WO2012168453A1 - Methods and uses relating to the diagnosis or prognosis of pain-related tissue states or pain-related diseases such as pain - Google Patents

Methods and uses relating to the diagnosis or prognosis of pain-related tissue states or pain-related diseases such as pain Download PDF

Info

Publication number
WO2012168453A1
WO2012168453A1 PCT/EP2012/060932 EP2012060932W WO2012168453A1 WO 2012168453 A1 WO2012168453 A1 WO 2012168453A1 EP 2012060932 W EP2012060932 W EP 2012060932W WO 2012168453 A1 WO2012168453 A1 WO 2012168453A1
Authority
WO
WIPO (PCT)
Prior art keywords
pain
slfn2
disease
protein
tissue status
Prior art date
Application number
PCT/EP2012/060932
Other languages
French (fr)
Inventor
Mathias Gebauer
Martin Michaelis
Danping Ding-Pfennigdorff
Anke M. Schulte
Daniel Ziemek
Christiane Metz-Weidmann
Original Assignee
Sanofi
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sanofi filed Critical Sanofi
Publication of WO2012168453A1 publication Critical patent/WO2012168453A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • Background of the Invention Present invention relates to Slfn 2 for use in the diagnosis or prognosis of a pain-related tissue status or a pain related-disease, methods of identifying or prognosing a pain- related tissue status or disease, a kit for use in the prognosis or diagnosis of a pain- related tissue status or a pain-related disease, a kit for detecting Slfn2, and a means for detecting Slfn2 for use in the prognosis or diagnosis of a pain-related tissue status or a pain-related disease.
  • IASP International Association for the Study of Pain
  • Pain of any type is the most common reason for physician consultation in the United States, prompting half of all Americans to seek medical care annually. It is a major symptom in many medical conditions, significantly interfering with a person's quality of life and general functioning. Diagnosis is based on characterizing pain in various ways, according to duration, intensity, type (dull, burning, throbbing or stabbing), source, or location in body. Usually pain stops without treatment or responds to simple measures such as resting or taking an analgesic, and it is then called 'acute' pain. But it may also become intractable and develop into a condition called chronic pain, in which pain is no longer considered a symptom but an illness by itself.
  • Pain is part of the body's defense system, triggering a reflex reaction to retract from a painful stimulus, and helps adjust behavior to increase avoidance of that particular harmful situation in the future.
  • Acute pain is 'normal' pain, it is felt when hurting a toe, breaking a bone, having a toothache, or walking after an extensive surgical operation.
  • Chronic pain is a 'pain illness', it is felt day after day, month after month, and seems impossible to heal.
  • Diagnosis of pain is mostly based on symptomatic description by the patient. However, this description is naturally very subjective and hardly quantifiable, so that the choice of the correct dosage and drug mostly relates to the patients age and body weight, together with some symptomatic analyses as to the nature of the pain-related tissue state (such as inflammation, necrosis etc.), pain-related disease (such as osteoarthritis, cancer, migraine etc) or pain (such as neuropathic pain or inflammatory pain). Especially in children, animals and mentally-disabled people, a proper diagnosis of pain is mostly difficult. Objective parameters allowing for a qualitative and quantitative assessment of pain e.g.
  • neuropathic pain i.e. pain arising as a consequence of neuronal damage
  • Several animal models of neuropathic pain are known in the art, which mimic many aspects of the clinical condition.
  • One object of the present invention is thus to provide a target for diagnosis and prognosis for pain-related tissue states and pain-related diseases, especially pain.
  • Slfn2 is involved in pain, which, in light of the then existing knowledge of possible functions of Slfn2 that rather hinted to completely different functions (e.g. in cell cycle control, see below) was not to be expected:
  • a screening assay for the identification of genes involved in pain three different inbred mouse strains differing in their pain sensitivity were examined. The expression of various genes was correlated with the pain sensitivity of the mouse strains.
  • Slfn2 is an interesting target for the identification and profiling of compounds concerning their analgesic effects. The above overview does not necessarily describe all problems solved by present invention.
  • the invention relates to Slfn2 for use as an indicator of a pain-related tissue status or a pain related disease or as an indicator of the risk for developing a pain-related tissue status or disease.
  • a second aspect of present invention relates to Slfn2 for use in the diagnosis or prognosis of a pain-related tissue status or of a pain-related disease.
  • present invention relates to a method of identifying
  • the method comprising detecting the level of Slfn2.
  • present invention relates to a kit for use in a method of identifying a pain-related tissue status or the presence of a pain-related disease and/or the risk of developing a pain-related tissue status or a pain-related disease and/or the progression or a stage of a pain-related tissue status or a pain-related disease in an individual, the method comprising detecting the level of Slfn2 and the kit comprising one or means for detecting Slfn2.
  • present invention relates to a kit for detecting Slfn2 comprising
  • a data carrier comprising instructions for a method a pain-related tissue status or the presence of a pain-related disease and/or the risk of developing a pain-related tissue status or a pain-related disease and/or the progression or a stage of a pain-related tissue status or a pain-related disease in an individual, the method comprising detecting the level of Slfn2 (iii) a container.
  • present invention relates to a means of detecting Slfn2 for use in diagnosis or prognosis of a tissue status or a disease.
  • this invention relates to a method for diagnosing or prognosing a pain- related tissue status or a pain-related disease, comprising:
  • an elevated level of Slfn2 in the individual as compared to the reference samples indicates the presence, increased risk of, or progression of the pain- related tissue status or pain-related disease.
  • the Slfn2 is selected from the group consisting of SEQ ID NO: 1 , 2, 6, 7, 8, and 9.
  • the detecting step comprises detecting the level of Slfn2 from a sample from an individual.
  • the sample is selected from the group consisting of: cerebrospinal fluid, neural tissue, extraneural tissue, a neural cell, and an extraneural cell.
  • the reference sample is selected from the group consisting of: a sample from a subject not having a pain-related tissue status or a pain- related disease, a sample from a subject having a pain-related tissue status or a pain- related disease, and a sample from the individual from an earlier point in time.
  • the level of Slfn2 is either the level of Slfn2 nucleic acid or the level of Slfn2 protein.
  • this invention also pertains to a kit for detecting Schlafen 2 (Slfn2) comprising:
  • the means for detecting Slfn2 is a means for determining the expression level of Slfn2.
  • the means for detecting is selected from the group consisting of: a means for detecting the expression level of the Slfn2 gene, a means for detecting the expression level of the Slfn2 RNA, and a means for detecting the expression level of the Slfn2 protein.
  • said kit further comprises:
  • the means comprises one or more nucleic acids or derivatives for detecting a Slfn2 gene or gene product or functionally active variant thereof, wherein the nucleic acid or derivative is selected from the group consisting of a nucleic acid probe, a polyamide or peptide nucleic acid (PNA), microRNA (miRNA), small interfering RNA (siRNA), primers for polymerase chain reaction (PCR), primers for reverse transcription (RT) reaction, and primers for DNA sequencing.
  • PNA polyamide or peptide nucleic acid
  • miRNA microRNA
  • siRNA small interfering RNA
  • PCR primers for reverse transcription
  • RT reverse transcription
  • the means comprises a peptide, polypeptide, or protein for detecting a Slfn2 gene or gene product or functionally active variant thereof, wherein the protein or polypeptide is a protein ligand, an antibody, a fragment or derivate thereof, a protein scaffold, a darpin or an anticalin, or wherein the polypeptide or peptide is a probe or a mass spectrometry probe.
  • the means comprises:
  • Slfn2 refers to murine Slfn2 comprising the nucleic acid sequence shown in SEQ ID NO: 1 , and the amino acid sequence shown in SEQ ID NO: 2, or to a functional variant thereof as defined below.
  • functional variant of Slfn2 encompasses naturally occurring variants, e.g. the below listed homologs, especially the human and/or rat protein and/or nucleic acid as identified below.
  • Slfn2 is a member of the (Schlafen) Slfn gene family comprising 10 family members in mouse and 7 family members in human (Schwarz et al., 1998 infra; Berger et al., Nature Immunology Vol.1 1 , Nr.4 (2010), p.335-343). Slfn2 is sometimes also referred to as schlafen family member 12 like (see e.g. accession number NP_035538 in the NCBI database that corresponds to the murine Slfn2 protein and coding sequence).
  • Slfn 2 has strongest homology to human SLFN12L, rat slfn 2, orang utan SLFN12L and chimpanzee SLFN12L (see below).
  • the gene of murine Slfn 2 is located on chromosome 4 (Ferguson et al., 2005).
  • the reference sequence of the murine Slfn2 coding sequence is known and publicly available under the accession number: AF099973 from the NCBI (National Centre for Biotechnology Information; National Library of Medicine 38A, Bethesda, MD20894, USA; www.ncbi.nih.gov). Version AF099973.1 of the coding sequence (SEQ ID NO: 1 , positions 1 to 1684) is given in the following: 1 gctttaatgc agcaaggaac aaagaactca catgggctta gaggtgaaca cacaaccagg
  • coding sequences comprised herein are indicated as cDNA sequences corresponding to the respective mRNA sequences.
  • the above coding sequence (SEQ ID NO: 1 ) encodes a 378 amino acid protein the reference sequence of which is publicly available at the NCBI database under accession number AAC83826.
  • the translated region starts at position 183 of SEQ ID NO: 1 and ends at position 1319.
  • Version AAC83826.1 (SEQ ID NO: 2) is given in the following:
  • the promoter region of murine Slfn2 has been cloned and analysed (see Wern-Joo et al., 2007). According to Wern-Joo et al., Molecular Immunology 44 (2007), 3273-3282 (the content of which is specifically incorporated herein by reference, see especially also page 3278, e.g. figure 3 of that article), the transcription start site is located 12 nucleotides upstream of the first nucleotide according to SEQ ID NO: 1 . In the sequence according to SEQ ID NO: 3 as given below, the nucleotides according to Wern-Joo infra (capital letters) are attached to the first part of SEQ ID NO: 1 (small letters):
  • the translation start (methionin/atg) is underlined and typed bold.
  • the promoter region of murine Slfn2 has been cloned and analysed (see Wern-Joo et al., 2007).
  • the promoter of murine Slfn2 plus some downstream sequences according to Wern-Joo et al., 2007 is given in figure 4 (the sequence according to figure 4 includes some downstream positions incl. the translation start - Met).
  • the upstream regulatory region, i.e. the promoter from position -1766 to +1 (+1 is underlined) is given in the following as SEQ ID NO: 4:
  • Slfn2 sequence containing the upstream regulatory sequence according to Wern- Joo et al., 2007 (capital letters) and the complete coding sequence according to SEQ ID NO: 1 (small letters) comprises positions -1766 to +1696 and is given in the following:
  • the upstream sequence comprises two AP-1 sites (-917 to -908 and -703 to -695) and one NF-kappa B binding site (-21 to -12). According to Wern-Joo et al., 2007 infra both AP-1 sites are important for maximal promoter activity upon LPS or CpG-DNA
  • Slfn-2 promoter fragments usable for cloning of reporter-gene constructs are nucleic acids comprising or having the following positions according to SEQ ID NO:5: from -1766/-1265 to +1/+50/
  • Slfn2 has been reported to be implicated in the regulation of quiescence in T-cells and monocytes (Berger et al., 2010 infra) and in interferon alpha-induced growth arrest in fibroblasts (Katsoulidis et al., Journal of Biological Chemistry Vol.284, Nr. 37 (2009)).
  • a point mutation on position 135 of the 278 amino acid protein caused by a thymidine-to- adenosine transversion in the Slfn2 gene and leading to an isoleucine to asparagine substitution leads to a loss-of-f unction mutation causing lymphoid and myeloid immunodeficiency due to loss of quiescence of T-cells and monocytes in mice, and causes the "electra" phenotype, leaving natural killer cells or B cells unaffected (Berger et al., 2010).
  • NFAT and NF-kB and Erk and Akt appear to be normally activated in electra mice, the mitogen-activated protein kinases p38 and Jnk are constitutively phosphorylated.
  • expression levels of the anti-apoptotic Bcl-2 is reported to be lower in T-cells derived from electra mice, wherein transgenic expression of Bcl-2 rescues CD4+ and CD8+ T-cells derived from electra mice from apoptotic cell death (all Berger et al., 2010 infra, see also Horton and Powell, Nature Immunology Vol. 1 1 , Nr.4 (2010), p.281 -282).
  • the reported mutation of Slfn2 appears to affect T- cell antigen receptor-induced signaling pathways, wherein other pathways are not affected.
  • the isoleucine at position 135 of murine Slfn2 according to SEQ ID NO:2 (and corresponding to position 135 in rat Slfn2 according to SEQ ID NO:7 and position 123 in hsSLFN12L according to SEQ ID NO:9) is conserved among murine Slfn2, rat Slfn2 and homo sapiens SLFN12L hinting to an equal importance of said amino acid or surrounding protein region within the respective rat and homo sapiens proteins.
  • Percent identity has been determined using the EMBL ClustalW program.
  • the rat homolog of Slfn2 is schlafen family member 12-like (Slfn12L).
  • the protein consists of 381 amino acids and the protein sequence is available under accession number NP_001 100501 .1 (SEQ ID NO: 7) and is encoded by the coding sequence NM_001 107031 .1 (from positions 193-1338).
  • rat Slfn2 The coding sequence of rat Slfn2 according to NM_001 107031 .1 is given following (SEQ ID NO: 6):
  • the divergent AAA domain spans positions 231 to 361 of the rat Slfn2 protein.
  • the Human homolog of Slfn2 is schlafen family member 12-like (SLFN12L).
  • the coding sequence can be retrieved from the NCBI database under accession number
  • NM_001 195790.1 (SEQ ID NO: 8), coding for a 588 amino acid protein with sequence according to NCBI accession number NP_001 182719.1 (SEQ ID NO:9) .
  • the divergent AAA domain spans positions 218 to 349 of human SLFN12L.
  • SLFN2 homologs are known in Orang Utan (Pongo abelii, see Gene ID: 100190853 of NCBI database) and Chimpanzee (Pan troglodytes, see Gene ID: 454585 of NCBI database).
  • Slfn2 protein refers e.g to a protein having or comprising
  • amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
  • amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
  • amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to SEQ ID NO:2, and/or
  • amino acid sequence with at least 50% identity with the amino acid sequence according to SEQ ID NO:2 and/or 6.
  • amino acid sequence with one of the following sequence motifs: YxxlxV, YxxlxVxxW, YxxlxVxxWS or YxxlxVxxWSxS with "x" standing for any amino acid and/or
  • 7. carries a variant AAA domain, preferably with 50, 60, 70, 80, 90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2.
  • the Slfn2 protein or the functionally active variant can comprise, consist of or have an amino acid sequence with the following features: An amino acid sequence having (1 ), (2), (3), (4), (5), (6) and/or (7).
  • the protein can also have (1 ) and (2) or (1 ) and (3) or (1 ) and (4) or (1 ) and (5) or (1 ) and (6) or (1 ) and (7) or (2) and (3) or (2) and (6) or (2) and (5) or (2) and (6) or (2) and (7) or (3) and (4) or (3) and (5) or (3) and (6) or (3) and (7) or (6) and (5) or (4) and (6) or (4) and (7) or (5) and (6) or (5) and (7) or (6) and (7);
  • the data carrier can also comprise (1 ) and (2) and (3) or (1 ) and (2) and (4) or (1 ) and (2) and (5) or (1 ) and
  • the data carrier can also comprise (1 ) and (2) and (3) and (4) or (1 ) and (2) and (3) and (5) or (1 ) and (2) and (3) and (6) or (1 ) and (2) and (3) and (7) or (2) and (3) and (4) and (5) or (2) and (3) and (4) and (6) or (2) and (3) and (4) and (7) or (4) and (5) and (6) or (4) and (5) and (7) or (5) and (6) and (7);
  • the data carrier can also comprise (1 ) and (2) and (3) and (4) or (1 ) and (2) and (3) and (5) or (1 ) and (2) and (3) and (6) or (1 ) and (2) and (3) and (7) or (2) and (3) and (4) and (5) or (2) and (3) and (4) and (6) or (2) and (3) and (4) and (7) or
  • the data carrier can also comprise (1 ) and (2) and (3) and (4) and (5) or (1 ) and (2) and (3) and (4) and (6) or (1 ) and (2) and (3) and (4) and (7) or (2) and (3) and (4) and (5) and (6) or (2) and (3) and (4) and (5) and (7) or (3) and (4) and (5) and (6) and (7).
  • pain refers to a complex subjective sensation reflecting real or potential tissue damage and the affective response to it.
  • the term “pain” encompasses acute pain, subacute pain and chronic pain. It further encompasses disease-associated pain, algesia and migraine.
  • Acute pain is a physiological signal indicating a possible or actual injury. Acute pain starts abruptly and is generally sharp in quality. It serves as a warning of disease or a threat to the body. Acute pain might be mild and last just a moment, or it might be severe and last for weeks. In most cases, acute pain disappears when the underlying cause of pain has been treated or healed. Acute pain might be somatogenetic (organic).
  • Organic pain might be any pain resulting from a disorder, abnormality or chemical imbalance in an organ system, for example, the human or animal body.
  • organic pain is a term covering pain causes that range in diversity from heartburn to multiples sclerosis.
  • any system or body organ can be affected by organic pain including, but not limited to, the circulatory system, musculoskeletal system, or neurological system.
  • Further examples of organic pain are constipation, diarrhoea, gastrointestinal tract infections, enteritis, colitis, or urinary tract infections.
  • Unrelieved acute pain might lead to chronic pain. Subacute pain can be classified as a status between acute pain and chronic pain.
  • Chronic pain can either be somatogenetic (organic) (see above) or psychogenic (psychosomatic). Chronic pain is frequently accompanied or followed by vegetative signs, which often result in depression.
  • acute pain is pain that lasts less than 30 days
  • chronic pain is pain of more than six months duration
  • subacute pain is pain that lasts from one to six months
  • acute pain may refer to pain that lasts less than 30 days (about 1 month)
  • chronic pain may refer to pain that lasts more than six months
  • subacute pain may refer to pain that lasts from one to six months.
  • acute pain may refer to pain that does not extend beyond the expected period of healing and chronic pain may refer to pain that extends beyond the expected period of healing.
  • pain means pain of nociceptive, inflammatory or neuropathic origin.
  • Nociceptive pain is part of a rapid warning relay instructing the motor neurons of the central nervous system to minimize detected physical harm.
  • Nociceptive pain is caused by activation of nociceptors. These nociceptors are free nerve endings that terminate just below the skin, in tendons, joints, and in body organs.
  • Nociceptive pain may be divided into “superficial somatic” pain and “deep” pain.
  • “Deep” pain in turn, may be divided into “deep somatic” pain and "visceral” pain.
  • Superficial somatic pain is initiated by activation of nociceptors in the skin or superficial tissues, while “deep somatic” pain is initiated by stimulation of nociceptors in ligaments, tendons, bones, blood vessels, fasciae and muscles, and is dull, aching, poorly-localized pain.
  • "Visceral” pain originates in the viscera (organs).
  • “Visceral” pain may be well-localized, but often it is extremely difficult to locate, and several visceral regions produce "referred” pain when injured, where the sensation is located in an area distant from the site of pathology or injury. Examples of nociceptive pain include, but are not limited to, sprains, bone fractures, burns or obstructions.
  • nociceptive pain can be associated with nerve damage caused by trauma, diseases such as diabetes, shingles, irritable bowel syndrome, late-stage cancer or the toxic effects of chemotherapy.
  • Nociceptive pain is generally time-limited. This means that when the tissue damage heals, the pain typically resolves. Thus, nociceptive pain is usually acute pain. Another characteristic of nociceptive pain is that it tends to respond well to treatment with opioids.
  • Neuropathic pain or neuralgia can be defined as non-nociceptive pain or as pain that is not related to activation of pain receptor cells in any part of the body.
  • Neuropathic pain or neuralgia is rather caused by damage to or malfunction of the nervous system.
  • neuropathic pain exists with no continuous nociceptive input.
  • Neuralgia or neuropathic pain can be defined as non-nociceptive pain, or in other words, pain that is not related to activation of pain receptor cells in any part of the body. It is believed that neuralgia is pain produced by a change in neurological structure or function. Unlike nociceptive pain, neuralgia exists with no continuous nociceptive input.
  • Neuralgia falls into two categories: central neuralgia and peripheral neuralgia. This unusual pain is thought to be linked to four possible mechanisms: ion gate malfunctions; the nerve becomes mechanically sensitive and creates an ectopic signal; cross signals between large and small fibers; and malfunction due to damage in the central processor.
  • Neuralgia falls into two categories: central neuropathic pain or neuralgia (originating in the brain or spinal cord) and peripheral neuropathic pain or neuralgia (originating in the peripheral nervous system).
  • Peripheral neuropathic pain can be described as burning, tingling, stabbing or as pins and needles. Bumping the "funny bone", for example, elicits peripheral neuropathic pain.
  • Neuropathic pain is often difficult to diagnose, and most treatments show little or no effectiveness. Diagnosis typically involves locating the damaged nerve by identifying missing sensory or motor function. This may involve tests such as an EMG test or a nerve conduction test. Neuralgia is more difficult to treat than other types of pain because it does not respond well to normal pain medications. Examples of neuropathic pain include, but are not limited to, reflex sympathetic dystrophy, nerve trauma, components of cancer pain, phantom limb pain, entrapment neuropathy (e.g.
  • peripheral neuropathy widespread nerve damage
  • bony hyperostosis casts crutches, prolonged cramped postures; haemorrhage into a nerve; exposure to cold or radiation; collagen-vascular disorders; infectious diseases such as Lyme disease and HIV; toxins such as emetine, hexobarbital, barbital, chlorobutanol, sulfonamides, phenytoin, nitrofurantoin, the vinca alkaloids, heavy metals, carbon monoxide, triorthocresylphosphate, orthodinitrophenol, and other solvents and industrial poisons; autoimmune reactions; nutritional deficiency, and vitamin B deficiency in particular; and metabolic disorders such as hypothyroidism, porphyria, sarcoidosis, amyloidosis, uremia and diabetes (see e.g.
  • Peripheral neuropathy may have several causes, for example, diabetes, chronic alcohol use, exposure to toxins (applied in chemotherapies), or vitamin deficiencies. Neuropathic pain is frequently chronic. It tends to have a less robust response to treatment with opioids. However, it may respond well to other drugs such as anti-seizure and anti-depressant drugs.
  • the pain may also be a mixed category pain.
  • the pain appears to be caused by a complex mixture of nociceptive pain and neuropathic pain.
  • an initial nervous system dysfunction or injury may trigger the neural release of inflammatory mediators and subsequent neurogenic inflammation.
  • Migraine headaches for example, probably represent a mixture of nociceptive pain and neuropathic pain.
  • Inflammatory pain is associated with tissue damage and the resulting inflammatory process.
  • Chronic pain may involve a mix of both inflammatory and neuropathic components, whereas inflammation may cause damage to the neurons and produce neuropathic pain or neuronal injury may cause an inflammatory reaction that contributes to the inflammatory pain.
  • Algesia from the Greek word algesis, is the sensitivity to pain.
  • Hyperalgesia is an increased sensitivity to pain, which may be caused by damage to nociceptors or peripheral nerves. Hyperalgesia can be experienced in focal, discrete areas, or as a more diffuse, body-wide form. Conditioning studies have established that it is possible to experience a learned hyperalgesia of the latter, diffuse form.
  • the focal form is typically associated with injury, and is divided into two subtypes: (i) primary hyperalgesia which describes pain sensitivity that occurs directly in the damaged tissues or (ii) secondary hyperalgesia which describes pain sensitivity that occurs in surrounding undamaged tissues.
  • disease-associated pain examples include, but are not limited to, pain associated with arthritis, back pain, cancer, epilepsy, lumbago, sciatica, lumbar spinal stenosis, cervical spinal stenosis, (clinical) depression, Fibromyalgia, Chronic Fatigue Syndrome, Complex Regional Pain Syndrome, Irritable Bowel Syndrome, Myofascial Pain Syndrome, Post-Vasectomy Pain Syndrome or Restless Leg Syndrome.
  • Disease- associated pain may also be pain associated with diabetes, e.g. diabetic neuropathy or the diabetic foot, pain associated with Parkinson's Disease, pain associated with viral infections, e.g. influenza or HIV, or bacterial infections, pain associated with fever, pain associated with levodopa, e.g.
  • Parkinson's disease pain associated with coronary heart disease, pain associated with shingles, pain associated with headache, pain associated with migraine, pain associated with muscular tension, especially chronic muscular tension, osteoarthritis as well as the pain-associated diseases listed under causes of neuropathic pain above.
  • Migraine (from the Greek words hemi, meaning half, and kranion, meaning skull) is a debilitating condition characterized by moderate to severe headaches. Migraine headache is typically associated with unilateral pain (affecting one half of the head) and pulsating in nature. It usually lasts between 2 to 70 hours. Symptoms of migraine include, but are not limited to, nausea, vomiting, photophobia (increased sensitivity to light), and phonophobia (increased sensitivity to sound). The cause of migraine headache is unknown. The most common theory is a disorder of the serotonergic control system. Migraine is usually treated with analgesics for the headache and antimetics for the nausea. In addition, triggering conditions such as noise or bright light should be avoided.
  • migraine is a disease that is associated with pain but is a disease that comprises more and different symptoms than symptoms of pain.
  • Said types of disease that are intimately connected with symptoms of pain but that have in addition symptoms outside of pain/algesia, are referred to, herein, as pain-associated diseases.
  • Some pain associated diseases are listed above in connection with disease-associated pain states and display pain as one characteristic disease symptom (e.g. migraine).
  • Other pain associated diseases may be increased, provoked or even caused by pain, such as depression or epilepsy.
  • pain also comprises equivalent states, stages, sensations and signalling cascades in lower animals or organisms, such as c. elegans.
  • pain also comprises pre-stages of pain.
  • tissue refers to an ensemble of cells of the same origin which fulfil a specific function concertedly.
  • tissue include but are not limited to bone, cartilage, connective tissue, muscle tissue, nervous tissue, and epithelial tissue. Multiple tissues together form an "organ” to carry out a specific function.
  • organ include but are not limited to brain, spinal cord, joint, skeleton, muscle, blood, brain, heart, liver, kidney, stomach, and skin.
  • a joint is formed of many different tissues such as but not limited to bone, cartilage, synovium, muscle, ligament, and tendon.
  • tissue furthermore relates to body liquids such as blood, liquor cerebrospinalis, saliva and lymph.
  • tissue status means the condition of a tissue.
  • the state of a tissue may be characterized by a specific morphology of such tissue or may be characterised by the expression of one or more specific molecules such as but not limited to peptides, proteins, and nucleic acids, or combinations thereof.
  • the status of a tissue may be regarded as "healthy” or “normal” in case it resembles the condition of such tissue when being free from illness or injury and efficiently fulfilling its specific function.
  • the status of a tissue may be regarded as "degenerative”, “diseased” or “abnormal” in case such tissue fails to fulfil its function due to an illness or injury.
  • the status of a tissue may be regarded as "degenerative”, “diseased” or “abnormal” in case the morphology of the tissue or its molecule expression pattern is “altered” or “changed” in comparison to normal tissue.
  • the morphology of a tissue or the expression pattern of specific molecules in a tissue may be an indicator for the state of a tissue.
  • a tissues status include but are not limited to tissue degradation such as cartilage degradation, bone degradation, and degradation of the synovium, tissue inflammation such as cartilage inflammation, or inflammation of the synovium, tissue remodelling such as bon remodelling or cartilage remodelling, sclerosis, liquid accumulation, or proliferative tissue such as proliferation in wound healing processes, cystic formations, or in cancer.
  • an altered (pathological) tissue status is preferably characterised by the tissue exhibiting a different (higher) Slfn2 level than normal (healthy) tissue, wherein the normal tissue can be tissue from another individual that is known not to have a pain-related tissue status or disease (e.g. not to experience pain) or the same tissue at an earlier time point (preferably before onset or worsening/progressing of the pain-related tissue status or disease, e.g. of pain).
  • a disease status is preferably characterised by an organ or individual exhibiting different slfn2 level in comparison to a normal organ or individual or in comparison to the same organ or individual at an earlier time point.
  • pain-related or pain-associated tissue status refers to a status of a tissue typically associated with pain or a pain-associated disease, typically a diseased or abnormal tissue state such as but not limited to an infection of tissue, infarction, necrosis or other states of tissue associated with pain.
  • tissue refers to an ensemble of cells of the same origin which concertedly fulfil a specific function.
  • tissue include but are not limited to bone, cartilage, connective tissue, muscle tissue, nervous tissue, and epithelial tissue. Multiple tissues together form an "organ” to carry out a specific function.
  • organ include but are not limited to joint, skeleton, muscle, blood, brain, heart, liver, kidney, stomach, and skin.
  • a joint is formed of many different tissues such as but not limited to bone, cartilage, synovium, muscle, ligament, and tendon.
  • disease and “disorder” are used interchangeably herein, referring to an abnormal condition, especially an abnormal medical condition such as an illness or injury, wherein a tissue, an organ or an individual is not able to efficiently fulfil its function anymore.
  • a disease is associated with specific symptoms or signs indicating the presence of such disease. The presence of such symptoms or signs may thus, be indicative for a tissue, an organ or an individual suffering from a disease. An alteration of these symptoms or signs may be indicative for the progression of such a disease.
  • a progression of a disease is typically characterized by an increase or decrease of such symptoms or signs which may indicate a "worsening" or “bettering” of the disease.
  • tissue, an organ or an individual being at "risk of developing" a disease is in a healthy state but shows potential of a disease emerging.
  • the risk of developing a disease is associated with early or weak signs or symptoms of such disease. In such case, the onset of the disease may still be prevented by treatment.
  • a disease include but are not limited to traumatic diseases, inflammatory diseases, infectious diseases, cutaneous conditions, endocrine diseases, intestinal diseases, neurological disorders, joint diseases, genetic disorders, autoimmune diseases, and various types of cancer.
  • Symptoms of a disease are implication of the disease noticeable by the tissue, organ or organism having such disease and include but are not limited to pain, weakness, tenderness, strain, stiffness, and spasm of the tissue, an organ or an individual.
  • “Signs” or “signals” of a disease include but are not limited to the change or alteration such as the presence, absence, increase or elevation, decrease or decline, of specific indicators such as biomarkers or molecular markers, or the development, presence, or worsening of symptoms.
  • Symptoms of pain include, but are not limited to an unpleasant sensation that may be felt as a persistent or varying burning, throbbing, itching or stinging ache.
  • biomarker or “indicator” are used interchangeably herein.
  • a biomarker can be defined as a substance within a biological system that is used as an indicator of a biological state of said system.
  • biomarker is sometimes also applied to means for the detection of said endogenous substances (e.g. antibodies, nucleic acid probes etc, imaging systems). In the context of present invention, however, the term “biomarker” shall be only applied for the substance, not for the detection means.
  • biomarkers can be any kind of molecule present in a living organism, such as a nucleic acid (DNA, mRNA, miRNA, rRNA etc.), a protein (cell surface receptor, cytosolic protein etc.), a metabolite or hormone (blood sugar, insulin, estrogen, etc.), a molecule characteristic of a certain modification of another molecule (e.g. sugar moieties or phosphoryl residues on proteins, methyl-residues on genomic DNA) or a substance that has been internalized by the organism or a metabolite of such a substance.
  • Slfn2 can thus be used as an indicator of pain, i.e. qualifies as biomarker for pain.
  • the indicator or biomarker refers to a sign or signal for a condition or is used to monitor a condition.
  • a condition refers to the biological status of a cell, tissue or organ or to the health and/or disease status of an individual.
  • An indicator may be the presence or absence of a molecule, including but not limited to peptide, protein, and nucleic acid, or may be a change in the expression level or pattern of such molecule in a cell, or tissue, organ or individual.
  • An indicator may be a sign for the onset, development or presence of a disease in an individual or for the further progression of such disease.
  • An indicator may also be a sign for the risk of developing a disease in an individual.
  • Known indicators of pain include but are not limited to: Cystatin C (Pain 102 (2003) p.251 -256), Cathepsin H and Cathepsin C and the circulatory pain-indicators beta endorphin, serotonin, 5-hydroxy indole acetic acid, anandamide, N-palmitoylethanolamide (for an overview see e.g. Journal of the American Osteopathic Association (2007) 107; p.387- 400).
  • level of Slfn2 refers to the expression level or the level of gene copies (also absence of Slfn2 or parts thereof or presence of fragments) present in a cell, tissue, organ or individual.
  • expression level refers to the amount of gene product present in the body or a sample at a certain point of time.
  • the expression level can e.g. be measured/quantified/detected by means of the protein or mRNA expressed from the gene.
  • the expression level can for example be quantified by normalizing the amount of gene product of interest (e.g. Slfn2 mRNA or protein) present in a sample with the total amount of gene product of the same category (total protein or mRNA) in the same sample or a reference sample (e.g.
  • the expression level can be measured or detected by means of any method as known in the art, e.g. methods for the direct detection and quantification of the gene product of interest (such as mass spectrometry) or methods for the indirect detection and measurement of the gene product of interest that usually work via binding of the gene product of interest with one or more different molecules or detection means (e.g. primer(s), probes, antibodies, protein scaffolds) specific for the gene product of interest, here Slfn2.
  • methods for the direct detection and quantification of the gene product of interest such as mass spectrometry
  • methods for the indirect detection and measurement of the gene product of interest that usually work via binding of the gene product of interest with one or more different molecules or detection means (e.g. primer(s), probes, antibodies, protein scaffolds) specific for the gene product of interest, here Slfn2.
  • the determination of the level of gene copies of Slfn2 comprising also the determination of the the absence or presence of one or more fragments (e.g. via nucleic acid probes or primers, e.g. quantitative PCR, Multiplex ligation-dependent probe amplification (MLPA) PCR (http://www.mlpa.com/) is also within the knowledge of the skilled artisan.
  • nucleic acid probes or primers e.g. quantitative PCR, Multiplex ligation-dependent probe amplification (MLPA) PCR (http://www.mlpa.com/)
  • the term "variant" is to be understood as a polynucleotide or protein which differs in comparison to the polynucleotide or protein from which it is derived by one or more changes in the sequence and comprises fragments or derivatives.
  • the polypeptide or polynucleotide from which a protein or nucleic acid variant is derived is also known as the parent polypeptide or nucleic acid.
  • a variant is constructed artificially, preferably by gene-technological means.
  • the parent polypeptide or polynucleotide is a wild-type protein or nucleic acid.
  • variants usable in the present invention may also be derived from homologs, orthologs, or paralogs of the parent molecule or from artificially constructed variant, provided that the variant exhibits at least one biological activity of the parent molecule, i.e. is functionally active, if the variant is meant to be a functional variant.
  • a "variant" as used herein can be characterized by a certain degree of sequence identity to the parent polypeptide or parent nucleic acid from which it is derived. More precisely, a protein variant in the context of the present invention exhibits 80% or more sequence identity to its parent polypeptide. A nucleic acid variant in the context of the present invention exhibits 80% or more sequence identity to its parent nucleic acid. The term "80% or more sequence identity” is used throughout the specification with regard to polypeptide and nucleic acid sequence comparisons.
  • This expression preferably refers to a sequence identity of 80% or more, 81 % or more, 82% or more, 83% or more, 84% or more, 85% or more, 86% or more, 87% or more, 88% or more, 89% or more, 90% or more, 91 % or more, 92% or more, 93% or more, 94% or more, 95% or more 96% or more, 97% or more, 98% or more, or 99% or more to the respective reference polypeptide or to the respective reference nucleic acid.
  • sequence alignments can be carried out with several art-known algorithms, e.g. with the mathematical algorithm of Karl in and Altschul (Karlin & Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5877), with hmmalign (HMMER package, http://hmmer.wustl.edu/) or with the CLUSTAL algorithm (Thompson, J. D., Higgins, D. G. & Gibson, T. J. (1994) Nucleic Acids Res. 22, 4673- 80) available e.g.
  • sequence identity may be calculated using e.g.
  • BLAST, BLAT or BlastZ (or BlastX).
  • BLASTN and BLASTP programs of Altschul et al. (1990) J. Mol. Biol. 215: 403-410.
  • Gapped BLAST is utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25: 3389-3402.
  • Sequence matching analysis may be supplemented by established homology mapping techniques like Shuffle- LAGAN (Brudno M., Bioinformatics 2003b, 19 Suppl 1 :154-162) or Markov random fields.
  • Shuffle- LAGAN Brudno M., Bioinformatics 2003b, 19 Suppl 1 :154-162
  • Markov random fields a homology mapping technique like Shuffle- LAGAN (Brudno M., Bioinformatics 2003b, 19 Suppl 1 :154-162) or Markov random fields.
  • Hybridization can also be used as a measure of sequence identity or homology between two nucleic acid sequences.
  • a nucleic acid sequence encoding miRNAs or a portion thereof can be used as a "hybridization probe” according to standard hybridization techniques.
  • the hybridization of a Slfn2 probe to DNA or RNA from a test source is an indication of the presence of the Slfn2 DNA or RNA in the test source.
  • Hybridization conditions are known to those skilled in the art and can be found, for example, in Current Protocols in Molecular Biology, John Wiley & Sons, N. Y., 6.3.1 - 6.3.6, 1991 .
  • Mode hybridization conditions are defined as equivalent to hybridization in 2X sodium chloride/sodium citrate (SSC) at 30°C, followed by a wash in 1 X SSC, 0.1 % SDS at 50°C.
  • Highly stringent conditions are defined as equivalent to hybridization in 6X sodium chloride/sodium citrate (SSC) at 45°C, followed by a wash in 0.2 X SSC, 0.1 % SDS at 65°C.
  • SSC sodium chloride/sodium citrate
  • Lowly stringent conditions are defined as equivalent to hybridization in 6X sodium chloride/sodium citrate (SSC) at 45°C, followed by a wash in 0.2 X SSC, 0.1 % SDS at 65°C.
  • protein and “polypeptide” are used interchangeably herein and refer to any peptide-linked chain of amino acids, regardless of length or post-translational modification.
  • Proteins usable in the present invention can be further modified by chemical or biological modification.
  • This means such a biologically or chemically modified polypeptide comprises other chemical groups than the 20 naturally occurring amino acids. Examples of such other chemical groups include without limitation glycosylated amino acids, phosphorylated amino acids or covalent attachment of amino-acid chains e.g. for stabilization of the protein/polypeptide (such as attachment of, e.g. rPEG, XTEN or PAS).
  • Modification of a polypeptide may provide advantageous properties as compared to the parent polypeptide, e.g. one or more of enhanced stability, increased biological half-life, or increased water solubility.
  • Chemical modifications applicable to the variants usable in the present invention include without limitation: PEGylation, glycosylation of non-glycosylated parent polypeptides, or the modification of the glycosylation pattern present in the parent polypeptide, rPEGylation, XTENylation or PASylation.
  • peptide refers to a short polymer of amino acids linked by peptide bonds. It has the same chemical (peptide) bonds as proteins, but is commonly shorter in length.
  • the shortest peptide is a dipeptide, consisting of two amino acids joined by a single peptide bond. There can also be a tripeptide, tetrapeptide, pentapeptide, etc.
  • the peptide has a length of up to 8, 10, 12, 15, 18 or 20 amino acids.
  • a peptide has an amino end and a carboxyl end, unless it is a cyclic peptide.
  • polypeptide refers to a single linear chain of amino acids bonded together by peptide bonds and preferably comprises at least about 21 amino acids.
  • a polypeptide can be one chain of a protein that is composed of more than one chain or it can be the protein itself if the protein is composed of one chain.
  • protein refers to a molecule comprising one or more polypeptides that resume a secondary and tertiary structure and additionally refers to a protein that is made up of several polypeptides, i.e. several subunits, forming quaternary structures.
  • the protein has sometimes non- peptide groups attached, which can be called prosthetic groups or cofactors.
  • the primary structure of a protein or polypeptide is the sequence of amino acids in the polypeptide chain.
  • the secondary structure in a protein is the general three-dimensional form of local segments of the protein. It does not, however, describe specific atomic positions in three-dimensional space, which are considered to be tertiary structure.
  • the secondary structure is defined by patterns of hydrogen bonds between backbone amide and carboxyl groups.
  • the tertiary structure of a protein is the three-dimensional structure of the protein determined by the atomic coordinates.
  • the quaternary structure is the arrangement of multiple folded or coiled protein or polypeptide molecules molecules in a multi-subunit complex.
  • amino acid chain and “polypeptide chain” are used synonymously in the context of present invention.
  • Slfn2 or Slfn2 protein also encompasses naturally occurring variants such as homologs and orthologs in same or different species, in particular in human.
  • the different aspects of present invention also relate to functional variants of Slfn2 protein, either naturally occurring or non-naturally occurring.
  • variant of a Slfn2 protein comprises fragments and derivatives of a protein.
  • a fragment is a protein or polypeptide that carries one or more end-terminal (n- and/or c-terminal) or internal deletions of one, two or more amino acids, when compared to the full-length protein.
  • a functional fragment of the protein is any fragment of this protein having at least one and preferably two or more of the functional characteristics of the full-length protein.
  • the term derivative of a protein comprises any type of modification of the protein in comparison to the naturally-occurring form, and in the context of present invention especially in comparison to the Slfn2 protein according to SEQ IDs no. 2 and 9, which is not a deletion.
  • a functional derivative of the protein is any derivative of this protein having at least one and preferably two or more of the functional characteristics of the unmodified protein.
  • the variants of Slfn2 of present invention also comprise functional derivatives of fragments of Slfn2.
  • Non-naturally occurring variants may be obtained by a limited number of amino acid deletions, insertions and/or substitutions, particularly deletions, insertions and/or substitutions of at most 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid(s).
  • a Slfn2 variant of the present invention can e.g. be a functionally active variant, i.e. the variant maintains at least one of the biological functions of Slfn2 as described herein, e.g. its function in the context of pain (e.g. as manifestation of the pain phenotype "mechanic hyperalgesia") or as known in the literature.
  • maintenance of biological function is defined as having 50 % or more, 60 % or more, 70 % or more, 80 % or more or 90 % or more or 95 % or more of the activity of the natural occurring Slfn2.
  • a non-functional variant of a Slfn2 protein may also be subject of one of the aspects of present invention, e.g. as negative control.
  • the biological activity may be determined as known to the skilled person.
  • the manifestation of the pain phenotype "mechanic hyperalgesia" can be determined as detailed in the Examples and in Persson et al., 2009, Molecular Pain 5:7.
  • the Slfn2 variant may be modified in order to comprise a further component. Accordingly, the variant may be a molecule having a domain composed of a naturally occurring Slfn2 protein or a variant thereof as detailed herein and at least one further component.
  • variant may be a fusion protein comprising (i) a Slfn2 protein or functionally active variant and (ii) a further protein component.
  • the protein may be coupled to a marker, such as a tag used for purification purposes (e.g. 6 His (or HexaHis) tag, Strep tag, HA tag, c-myc tag or glutathione S-transferase (GST) tag).
  • a marker such as a tag used for purification purposes (e.g. 6 His (or HexaHis) tag, Strep tag, HA tag, c-myc tag or glutathione S-transferase (GST) tag).
  • a marker such as a tag used for purification purposes (e.g. 6 His (or HexaHis) tag, Strep tag, HA tag, c-myc tag or glutathione S-transferase (GST) tag).
  • GST glutathione S-transferas
  • the proteins are purified in two or more separation chromatography steps, in each case utilizing the affinity of a first and then of a second tag.
  • double or tandem tags are the GST-His-tag (glutathione-S-transferase fused to a polyhistidine- tag), the 6xHis-Strep-tag (6 histidine residues fused to a Strep-tag), the 6XH is-tag 100- tag (6 histidine residues fused to a 12-amino-acid protein of mammalian MAP-kinase 2), 8xHis-HA-tag (8 histidine residues fused to a hemagglutinin-epitope-tag), His-MBP (His- tag fused to a maltose-binding protein, FLAG-HA-tag (FLAG-tag fused to a hemagglutinin-epitope-tag), and the FLAG-Strep-tag.
  • GST-His-tag glutas
  • the marker could be used in order to detect the tagged protein, wherein specific antibodies could be used. Suitable antibodies include anti-HA (such as 12CA5 or 3F10), anti-6 His, anti-c-myc and anti- GST. Furthermore, the Slfn2 protein could be linked to a marker of a different category, such as a fluorescence marker or a radioactive marker, which allows for the detection of Slfn2. In a further embodiment, Slfn2 could be part of a fusion protein, wherein the second part could be used for detection, such as a protein component having enzymatic activity.
  • a marker of a different category such as a fluorescence marker or a radioactive marker
  • the Slfn2 variant could be a Slfn2 fragment, wherein the fragment is still functionally active.
  • This may include Slfn2 proteins with short internal and/or C- and/or N-terminal deletions (e.g. deletions of at most 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9, 8, 7, 6 5, 4, 3, 2, or 1 amino acid).
  • the Slfn2 fragment may be further modified as detailed above for the Slfn2 protein.
  • the Slfn2 variant may comprise one or more amino acid substitution(s).
  • Semi-conservative and especially conservative amino acid substitutions wherein an amino acid is substituted with a chemically related amino acid are preferred.
  • Typical substitutions are among the aliphatic amino acids, among the amino acids having aliphatic hydroxyl side chain, among the amino acids having acidic residues, among the amide derivatives, among the amino acids with basic residues, or the amino acids having aromatic residues.
  • Typical semi-conservative and conservative substitutions are:
  • the Slfn2 protein or fragment or variant with substitution may be modified as detailed above for the Slfn2 protein or fragment or variant.
  • all details given with respect to Slfn2 protein also relate to functionally active variants thereof, unless stated otherwise.
  • the above modifications of the Slfn2 protein may be combined.
  • the variant of the present invention may be e.g. fragment of Slfn2 having a marker fused to it, or a Slfn2 protein fragment comprising one or more amino acid substitutions.
  • a derivative of Slfn2 protein or a Slfn2 protein fragment is a Slfn2 protein or fragment carrying any kind of modification of the amino acid sequence that is not a deletion and/or carrying any other kind of modification, such as a chemical or biological modification.
  • Derivatives comprise proteins carrying one or more modifications e.g. leading to the stabilization of the polypeptide, or enabling a specific targeting of the polypeptide to certain cells or facilitating its entry into or uptake by cells (such as cell- permeant phosphopeptides ortho coupling to cell-permeant peptide vectors, e.g.
  • the Slfn2 protein is a naturally occurring Slfn2 protein as detailed above, such as a naturally occurring mouse Slfn2 protein such as comprising or having SEQ ID NO: 2, or a naturally occurring human orthologous protein, such as comprising or having SEQ ID NO: 9.
  • the terms "nucleic acid” or “nucleic acid molecule” are used synonymously and are understood as single or double-stranded oligo- or polymers of deoxyribonucleotide or ribonucleotide bases or both.
  • nucleic acid is formed through phosphodiester bonds between the individual nucleotide monomers.
  • nucleic acid includes but is not limited to ribonucleic acid (RNA) and deoxyribonucleic acid (DNA) molecules.
  • RNA ribonucleic acid
  • DNA deoxyribonucleic acid
  • the depiction of a single strand of a nucleic acid also defines (at least partially) the sequence of the complementary strand.
  • the nucleic acid may be single or double stranded, or may contain portions of both double and single stranded sequences.
  • the nucleic acid may be obtained by biological, biochemical or chemical synthesis methods or any of the methods known in the art.
  • nucleic acid comprises the terms "polynucleotide” and "oligonucleotide”.
  • nucleic acid comprises cDNA, genomic DNA, recombinant DNA, cRNA and mRNA.
  • a nucleic acid may consist of an entire gene, or a portion thereof, the nucleic acid may also be a microRNA (miRNA) or small interfering RNA (siRNA).
  • miRNAs are short ribonucleic acid (RNA) molecules, on average only 22 nucleotides long, found in all eukaryotic cells.
  • MircoRNAs miRNAs
  • miRNAs are post-transcriptional regulators that bind to complementary sequences on target messenger RNA transcripts (mRNAs), usually resulting in translational repression and gene silencing.
  • RNA molecules short ribonucleic acid
  • RNAi RNA interference
  • the nucleic acid can also be an artificial nucleic acid.
  • Artificial nucleic acids include polyamide or peptide nucleic acid (PNA), morpholino and locked nucleic acid (LNA), as well as glycol nucleic acid (GNA) and threose nucleic acid (TNA). Each of these is distinguished from naturally-occurring DNA or RNA by changes to the backbone of the molecule.
  • nucleic acids can e.g. be synthesized chemically, e.g. in accordance with the phosphotriester method (see, for example, Uhlmann, E. & Peyman, A. (1460) Chemical Reviews, 90, 543-584).
  • Aptamers are nucleic acids which bind with high affinity to a polypeptide, here Slfn2.
  • Aptamers can be isolated by selection methods such as SELESIfn2 (see e.g. Jayasena (1469) Clin. Chem., 45, 1628-50; Klug and Famulok (1464) M. Mol. Biol. Rep., 20, 97-107; US 5,582,981 ) from a large pool of different single-stranded RNA molecules.
  • Aptamers can also be synthesized and selected in their mirror-image form, for example as the L-ribonucleotide (Nolte et al. (1466) Nat. Biotechnol., 14, 1 1 16-9; Klussmann et al. (1466) Nat. Biotechnol., 14, 1 1 12-5).
  • L-ribonucleotide Nolte et al. (1466) Nat. Biotechnol., 14, 1 1 16-9; Klussmann et al. (1466) Nat. Biotechnol., 14, 1 1 12-5).
  • Forms which have been isolated in this way enjoy the advantage that they are not degraded by naturally occurring ribonucleases and, therefore, possess greater stability.
  • Nucleic acids may be degraded by endonucleases or exonucleases, in particular by DNases and RNases which can be found in the cell.
  • nucleic acids it is, therefore, advantageous to modify the nucleic acids in order to stabilize them against degradation, thereby ensuring that a high concentration of the nucleic acid is maintained in the cell over a long period of time (Beigelman et al. (1465) Nucleic Acids Res. 23:3989-94; WO 95/1 1910; WO 98/37240; WO 97/291 16).
  • stabilization can be obtained by introducing one or more internucleotide phosphorus groups or by introducing one or more non-phosphorus internucleotides.
  • Suitably modified internucleotides are compiled in Uhlmann and Peyman (1460), supra (see also Beigelman et al. (1465) Nucleic Acids Res.
  • Modified internucleotide phosphate radicals and/or non-phosphorus bridges in a nucleic acid which can be employed in one of the uses according to the invention contain, for example, methyl phosphonate, phosphorothioate, phosphoramidate, phosphorodithioate and/or phosphate esters, whereas non-phosphorus internucleotide analogues contain, for example, siloxane bridges, carbonate bridges, carboxymethyl esters, acetamidate bridges and/or thioether bridges. It is also the intention that this modification should improve the durability of a pharmaceutical composition which can be employed in one of the uses according to the invention.
  • oligonucleotide when used in the context of one of the different aspects of present invention, refers to a nucleic acid of up to about 50 nucleotides, e.g. 2 to about 50 nucleotides in length.
  • polynucleotide when used in the context of one of the different aspects of present invention, refers to a nucleic acid of more than about 50 nucleotides in length, e.g. 51 or more nucleotides in length.
  • means for detection or means for detecting as used herein refers to any means suitable for specific detection of Slfn or nucleic acid, especially in a probe, in isolated organic matter (e.g. isolated protein or nucleic acid), a tissue, an organ or an animal body. The detection is usually mediated by a specific binding to the target molecule, here Slfn2 nucleic acid or polypeptide.
  • Probes and primers are short polynucleotides or oligonucleotides for the detection of nucleic acids in a sample or in vivo by hybridizing (probe) to the target nucleic acid or by hybridization to and amplification of the target nucleic acid.
  • Nucleic acids suitable for use as probe or primer comprise e.g. a polynucleotide probe, one or more primers (e.g.
  • a primer pair preferably one or more primers for polymerase chain reaction (PCR), reverse transcription (RT) reaction, or DNA sequencing, a peptide- or polyamid- nucleic acid (PNA), a locked nucleic acid (LNA), a glycol nucleic acid (GNA), a threose nucleic acid (TNA), a microRNA (miRNA), and a small interfering RNA (siRNA).
  • the one or more primers e.g. a primer pair
  • RT-PCR real time polymerase chain reaction
  • qRT-PCR quantitative real time polymerase chain reaction
  • probe refers to a nucleic acid which is typically used for the detection of target RNA and/or DNA sequences that is complementary to the sequence of the probe.
  • a probe hybridizes to single-stranded nucleic acid (DNA or RNA) whose nucleotide sequence allows for nucleotide pairing due to complementarity between the probe and the target sequence.
  • the length of a probe depends on the intended use as well as the required specificity of the probe. Typically, a probe is 20-500 (i.e.
  • Probes are used in various experimental set ups such as but not limited to Southern and Northern Blots, for real-time PCR and In Situ Hybridization (ISH) as well as for microarray experiments.
  • a probe may be unlabeled, directly labelled, or indirectly labelled, such as with biotin to which a streptavidin complex may later bind.
  • Said label may be a molecule detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means.
  • suitable labels include 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens and other entities which are or can be made detectable.
  • a label may be incorporated into nucleic acids at any position, e.g. at the 3' end, at the 5' end or internally.
  • probe also encompasses nucleic acids differing in the composition of their backbone such as but not limited to peptide nucleic acids (PNAs), locked nucleic acids (LNAs), glycol nucleic acids (GNAs) and threose nucleic acids (TNAs).
  • PNAs peptide nucleic acids
  • LNAs locked nucleic acids
  • GNAs glycol nucleic acids
  • TAAs threose nucleic acids
  • Said nucleic acid as a probe may be unlabeled, directly labeled, or indirectly labeled, such as with biotin to which a streptavidin complex may later bind.
  • Said label may be a molecule detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means.
  • suitable labels include 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens and other entities which are or can be made detectable.
  • a label may be incorporated into nucleic acids at any position, e.g. at the 3' end, at the 5' end or internally.
  • primer refers to a single-strand nucleic acid which typically serves as a starting point for DNA-replicating enzymes.
  • a primer binds to or hybridises with a DNA template and typically comprises a sequence being complementary to the DNA sequence to which it is supposed to bind.
  • a primer may also comprise additional sequences e.g. sequences serving as nuclease cleavage sites (e.g. Bam H1 , Hind III, etc.).
  • the length of a primer is chosen depending on the intended use. For instance, primers used for the amplification of DNA in Polymerase-Chain Reactions (PCR) typically have a length of at least 10 nucleotides, preferably between 10 to 50 (i.e.
  • primers are used for sequencing of DNA templates.
  • primer also encompassed in the term "primer” are "degenerate primers", which are a mixture of similar but not identical primers.
  • a primer may be tagged or labeled with a marker molecule detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means.
  • Probes and primers for the detection of Slfn2 are known in the art (see e.g. Wern-Joo et al, 2007) and can be ordered custom-made by commercial vendors against any known sequence including that of Slfn2. Examples include (all taken from Wern-Joo et al., 2007):
  • 5'-CTCACCTCAGAAAACAGGAGAATGC-3' (sense murine Slfn2; SEQ ID NO:10), 5'-CAGAAGTGAGTGACAGGCAGCTG-3' (anti-sense, murine Slfn2; SEQ ID NO:1 1 ), 5'-CGGCTAGCTTGGTGGCTCCTAGCGRR-3' (sense Slfn2 -1766; SEQ ID NO.12), 5'-CGGCTAGCGGAACTGAACTGGAGCT-3' (sense Slfn2 -1265; SEQ ID NO.13), 5'-CCCTCGAGCCATGTCAGCAGTTGGTAAAGC-3' (anti-sense SIfn 2 +198; SEQ ID NO:14).
  • the skilled artisan knows how to select suitable primers.
  • peptide nucleic acids PNAs
  • locked nucleic acids LNAs
  • glycol nucleic acids GAAs
  • threose nucleic acids TAAs
  • the above-mentioned nucleic acids are e.g. for detecting the SIfn nucleic acid sequences of present invention, e.g. one or more of the SIfn nucleic acid sequences of embodiment 20 and are e.g.
  • nucleic acids may allow the determination of the expression level of Slfn2 on the Slfn2 gene or Slfn2 mRNA level, e.g. in a sample, such as in a sample from a subject or in an assay, such as a biochemical or cellular assay (e.g. in a mixture in the context of a biochemical assay or in a cell in the context of a cellular assay).
  • variant of a nucleic acid comprises fragments and derivatives of a nucleic acid.
  • the variants of present invention also comprise functional derivatives of fragments of the nucleic acid.
  • Non-naturally occurring variants may be obtained by a limited number of nucleotide deletions, insertions and/or substitutions, particularly deletions, insertions and/or substitutions of at most 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 nucleotide(s).
  • a fragment of a nucleic acid is a nucleic acid that carries one or more terminal (5' (upstream) and/or 3' (downstream)) or internal deletions of one, two or more
  • a functional fragment of the nucleic acid is any fragment of the Slfn2 nucleic acid having at least one and preferably two or more of the functional characteristics of the full-length nucleic acid (e.g. the ability to confer Slfn2-characteristic transcriptional regulation of downstream elements or the characteristic to code for a Slfn2 protein).
  • derivative of a nucleic acid comprises any type of modification (e.g. a chemical or biological modification) of the nucleic acid in comparison to the naturally- occurring form, and in the context of present invention especially in comparison to the Slfn2 nucleic acid according to SEQ ID NOs. 1 , 3, 4, 6 or 8 that is not a deletion.
  • Derivatives comprise Slfn2 nucleic acids carrying one or more modifications leading to the stabilization of the nucleic acid (such as phosphoorothioate modification or modification of the nucleic acid backbone).
  • a functional derivative of the nucleic acid is any derivative of this nucleic acid having at least one and preferably two or more of the functional characteristics of the unmodified nucleic acid.
  • Slfn2 nucleic acid encompasses nucleic acids coding for the above Slfn2 protein and regulatory nucleic acids of the Slfn2 gene as well as naturally occurring and non-naturally occurring variants thereof (as defined herein).
  • the term relates to coding or non-coding regions of the Slfn2 gene, wherein these sections are of a relevant size in order to be specific for that gene. Examples of those regions are introns, exons or regulatory elements such as a Slfn2 promoter.
  • Slfn2 or Slfn2 nucleic acid also encompasses naturally occurring variants such as homologs and orthologs in same or different species, in particular in human (e.g.
  • Slfn2 nucleic acids according to SEQ IDs NO. 1 (mouse), 6 (rat) or 8 (human).
  • the different aspects of present invention also relate to functional variants of Slfn2 nucleic acid, either naturally occurring or non- naturally occurring.
  • Preferred aspects of a Slfn2 nucleic acid relate to the Slfn2 gene, promoter, DNA, cDNA or mRNA or oligonucleotides or polynucleotides able to specifically hybridize with one of these nucleic acids.
  • Slfn2 nucleic acids are e.g. a Slfn2 nucleic acid coding for the naturally occurring Slfn2 protein as detailed above (e.g.
  • a protein comprising or having an amino acid sequence according to SEQ ID NO: 2, 7 or 9) such as a nucleic acid comprising or having a nucleotide sequence according to SEQ ID NO: 1 , 3, 4, 6, or 8).
  • Slfn2 nucleic acid is a Slfn2 nucleic acid able to drive expression of a downstream element such as a nucleic acid derived from the Slfn2 gene and comprising regulatory elements thereof but lacking the Slfn2 coding sequence, e.g. a nucleic acid comprising or consisting of a nucleotide sequence according to SEQ ID NO:4.
  • a Slfn2 nucleic acid variant of the present invention can be a functionally active variant, i.e. the variant maintains at least one of the biological functions of Slfn2 nucleic acid as described herein, e.g. its function in the context of pain (e.g. as manifestation of the pain phenotype "mechanic hyperalgesia", the ability to confer Slfn2-characteristic transcriptional regulation of downstream elements or the characteristic to code for a Slfn2 protein) or as known in the literature.
  • maintenance of biological function is defined as having 50 % or more, 60 % or more, 70 % or more, 80 % or more or 90 % or more or 95 % or more of the activity of the natural occurring Slfn2.
  • a non- functional variant of a Slfn2 nucleic acid may also be subject of one of the aspects of present invention, e.g. as negative control.
  • the biological activity may be determined as known to the skilled person, for example, the manifestation of the transcriptional activity as detailed in Wern-Joo et al., 2007; examples of functional and non-functional variants (fragments and derivatives) of nucleic acids, especially different promoter variants are disclosed in Wern-Joo et al, 2007 the disclosure of which is explicitly included herein, as reference.
  • nucleic acids comprising a Slfn2 promoter comprise a Slfn2 nucleic acid from -1256 to +1 or from -1265 to +198 or from +1699 to +1 or from -1699 to +198 with respect to the sequence according to SEQ ID NO: 5.
  • antibody or fragment thereof refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e. molecules that contain an antigen binding site that specifically binds an antigen. Also comprised are immunoglobulin-like proteins that are selected through techniques including, for example, phage display to specifically bind to a target molecule or target protein.
  • the immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., lgG1 , lgG2, lgG3, lgG4, lgA1 and lgA2) or subclass of immunoglobulin molecule.
  • antibodies and fragments thereof include, but are not limited to, polyclonal, monoclonal, monovalent, bispecific, heteroconjugate, multispecific, human, humanized (in particular CDR-grafted), deimmunized, or chimeric antibodies, single chain antibodies (e.g. scFv), Fab fragments, F(ab')2 fragments, fragments produced by a Fab expression library, diabodies or tetrabodies (Holliger P. et al., 1993), nanobodies, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • the antibody fragments are mammalian, preferably human antigen-binding antibody fragments and include, but are not limited to, Fab, Fab' and F(ab')2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (dsFv) and fragments comprising either a VL or VH domain.
  • Antigen-binding antibody fragments, including single-chain antibodies may comprise the variable domain(s) alone or in combination with the entirety or a portion of the following: hinge region, CL, CH1 , CH2, and CH3 domains.
  • the antigen-binding fragments may also comprise any combination of variable domain(s) with a hinge region, CL, CH1 , CH2, and CH3 domains.
  • Antibodies usable in the invention may be from any animal origin including birds and mammals.
  • the antibodies are human, simian (e.g. chimpanzee, bonobo, macaque), rodent (e.g. mouse and rat), donkey, sheep rabbit, goat, guinea pig, camel, horse, or chicken. It is particularly preferred that the antibodies are of human or murine origin.
  • "human antibodies” include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins.
  • Antibodies or antibody fragments specific for the Slfn2 protein may be used in a variety of assays in order to quantitate the Slfn2 protein.
  • Well known methods include, for example, immunoprecipitation, antibody sandwich assays, ELISA and affinity chromatography methods (see above).
  • the antibody that recognizes the target antigen here Slfn2 or a functionally active variant thereof is generally called the "primary antibody".
  • Said antibody may be labeled with a detectable tag/label in order to allow direct detection of the target antigen.
  • Said detectable tag/label may be an enzymatic, fluorescent or radioisotope tag/label.
  • the primary antibody is not labeled for direct detection. Instead a "secondary antibody” that has been labeled with a detectable tag/label (e.g. enzymatic, fluorescent or radioisotope tag/label) is applied in a second step to probe for the primary antibody, which is bound to the target antigen.
  • the primary antibody or the secondary antibody may be labeled with an affinity tag such as biotin.
  • an affinity tag such as biotin.
  • Antibodies against Slfn2 are known in the art (see e.g. Katsoulidis et al., 2009) and can be purchased from commercial vendors such as antibodies-online.com (http://www.antibodies-online.com).
  • An example of an n-terminal anti Slfn2 antibody suitable for use in e.g. Western Blot or ELISA is a polyclonal rabbit anti-mouse antibody that has been generated using a synthetic peptide corresponding to the n-terminal residues of mouse Slfn2 and that can be purchased from antibodies-online under ABIN393095.
  • protein scaffold in the context of present invention refers to non-antibody recognition proteins derived from one or more of structurally different protein families and able to bind with high specificity to a selected target, thereby mimeting the binding principle of immunoglobulins to varying degrees (see e.g. Hey, T., et al., 2005: Artificial, non-antibody binding proteins for pharmaceutical and industrial applications, Trends in Biotechnology, Vol. 23, No. 10, the disclosure of which is incorporated herein, by reference).
  • Examples comprise lipocalin-derived repeat proteins such as darpins, ankyrin-repeat proteins such as anticalins, nanobodies, affibodies, maxibodies, trans- bodies, tetranectin, iMabs, Adnectin, domain antibodies, Kunitz-type domains, Evibodies, Affilins, Microbodies).
  • lipocalin-derived repeat proteins such as darpins, ankyrin-repeat proteins such as anticalins, nanobodies, affibodies, maxibodies, trans- bodies, tetranectin, iMabs, Adnectin, domain antibodies, Kunitz-type domains, Evibodies, Affilins, Microbodies).
  • biochip and “microarray” are interchangeably used herein
  • the terms “attached” or “immobilized”, as used herein, refer to the binding between the nucleic acid(s), protein, polypeptide, or peptide and the solid support and may mean that the binding between the nucleic acid, protein, polypeptide, or peptide and the solid support is sufficient to be stable under conditions of binding, washing, analysis and removal.
  • the binding may be covalent or non-covalent. Covalent bonds may be formed directly between the nucleic acid, protein, polypeptide, or peptide and the solid support or may be formed by a cross linker or by inclusion of specific reactive groups on either the solid support or the nucleic acid, protein, polypeptide, or peptide, or both.
  • Non- covalent binding may be electrostatic, hydrophilic and hydrophobic interactions or combinations thereof. Immobilization or attachment may also involve a combination of covalent and non-covalent interaction.
  • a marker is any kind of substance which is able to indicate the presence of another substance or complex of substances.
  • the marker can be a substance that is linked to or introduced in the substance to be detected.
  • Detectable markers are used in molecular biology and biotechnology to detect e.g. a protein, a product of an enzymatic reaction, a second messenger, DNA, interactions of molecules etc.
  • suitable marker or labels include a fluorophore, a chromophore, a radiolabel, a metal colloid, an enzyme, or a chemiluminescent or bioluminescent molecule.
  • fluorophores include fluorescein, rhodamine, and sulfoindocyanine dye Cy5.
  • radiolabels examples include 3 H, 14 C, 32 P, 33 P, 35 S, 99m Tc or 125 l.
  • enzymes include horseradish peroxidase, alkaline phosphatase, glucose oxidase, and urease. Further examples and preferred embodiments are detailed herein.
  • Radioisotopes were used extensively in the past, but they are expensive, have a short shelf-life, offer no improvement in signal:noise ratio and require special handling and disposal.
  • Enzymes and fluorophores have largely replaced radioactive isotopes as detectable tags for assays. A number of advancements in reagents and instrumentation make these newer technologies more versatile and powerful.
  • Enzymatic tags such as horseradish peroxidase (HRP) are most commonly used for blotting, immunoassays and immunohistochemistry methods. Fluorescent tags are used predominately for cellular imaging, nucleic acid amplification and sequencing and microarrays; however, fluorescence technology is developing rapidly for application in all types of assays.
  • Slfn2 protein or a variant thereof may include a specific Slfn2 antibody or protein scaffold.
  • antibodies can be raised using well established techniques for immunizing animals with prepared forms of the antigen.
  • a variety of reagents is available to assist in antibody production and purification, and various companies specialize in antibody production services.
  • different levels of purity and types of specificity are needed in a supplied primary antibody.
  • antibodies may be monoclonal or polyclonal, supplied as antiserum or affinity-purified solution, and validated for native protein or denatured protein detection.
  • An antibody that recognizes the target antigen here Slfn2 or fragment or variant thereof, is called the "primary antibody.” If this antibody is labeled with a tag, direct detection of the antigen is possible. Usually, however, the primary antibody is not labeled for direct detection. Instead a "secondary antibody” that has been labeled with a detectable tag is applied in a second step to probe for the primary antibody, which is bound to the target antigen. Thus, the antigen is detected indirectly. Another form of indirect detection involves using a primary or secondary antibody that is labeled with an affinity tag such as biotin.
  • a secondary (or tertiary) probe such as streptavidin that is labeled with the detectable enzyme or fluorophore tag
  • streptavidin that is labeled with the detectable enzyme or fluorophore tag
  • a primary antibody without a detectable label and some sort of secondary (indirect) detection method is required in assay methods. Nevertheless, nearly any antibody can be labeled with biotin, HRP enzyme or one of several fluorophores if needed.
  • Most primary antibodies are produced in mouse, rabbit or one of several other species. Nearly all of these are antibodies of the IgG class. Therefore, it is relatively easy and economical for manufacturers to produce and supply ready-to-use, labeled secondary antibodies for most applications and detection systems. Even so, several hundred options are available, differing in the level of purity, IgG- and species-specificity, and detection label.
  • the choice of secondary antibody depends upon the species of animal in which the primary antibody was raised (the host species). For example, if the primary antibody is a mouse monoclonal antibody then the secondary antibody must be an anti-mouse antibody obtained from a host other than the mouse.
  • biotin-binding proteins As probes, the highly specific affinity interaction between biotin and avidin or streptavidin protein is the basis for many kinds of detection and affinity-purification methods. Biotin is very small (244 Daltons), so its covalent attachment to antibodies or other probes rarely interferes with their functions. Yet its presence as a label on a probe allows efficient and specific secondary detection with either avidin or streptavidin. Both kinds of biotin-binding proteins are available in purified forms labeled with enzymatic or fluorescent tags that enable detection in many kinds of assays systems. Enzymatic labels are most commonly used as secondary antibody (or streptavidin) tags for detection in blotting and immunoassays.
  • Enzymes provide detectable signal via their activity; reaction with a specific substrate chemical yields a colored, light-emitting, or fluorescent product. While reporter enzymes like beta-galactosidase and luciferase have been successfully used to make probes, alkaline phosphatase (AP) and horseradish peroxidase (HRP) are the two enzymes used most extensively as labels for protein detection. An array of chromogenic, fluorogenic and chemiluminescent substrates is available for use with either enzyme.
  • Alkaline phosphatase is a large (140 kDa) protein that catalyzes the hydrolysis of phosphate groups from a substrate molecule resulting in a colored or fluorescent product or the release of light as a byproduct of the reaction.
  • AP has optimal enzymatic activity at a basic pH (pH 8-10) and can be inhibited by cyanides, arsenate, inorganic phosphate and divalent cation chelators, such as EDTA.
  • pH 8-10 basic pH
  • AP offers a distinct advantage over other enzymes. Because its reaction rate remains linear, detection sensitivity can be improved by simply allowing a reaction to proceed for a longer time period.
  • Horseradish peroxidase is a 40 kDa protein that catalyzes the oxidation of substrates by hydrogen peroxide, resulting in a colored or fluorescent product or the release of light as a byproduct of the reaction.
  • HRP functions optimally at a near-neutral pH and can be inhibited by cyanides, sulfides and azides.
  • Antibody-HRP conjugates are superior to antibody-AP conjugates with respect to the specific activities of both the enzyme and antibody. In addition, its high turnover rate, good stability, low cost and wide availability of substrates makes HRP the enzyme of choice for most applications.
  • Fluorescent labels for detection were historically used in a small number of cell biology applications such as flow cytometry (FC), fluorescence-activated cell sorting (FACS) and immunohistochemistry (IHC) using fluorescence microscopy.
  • FC flow cytometry
  • FACS fluorescence-activated cell sorting
  • IHC immunohistochemistry
  • fluorescent proteins such as the various forms of green fluorescent protein (GFP) and the phycobiliproteins (allophycocyanin, phycocyanin, phycoerythrin and phycoerythrocyanin). While having the ability to produce an intense fluorescent signal for detection, fluorescent proteins can be difficult to optimize for conjugation purposes and may create steric hindrance or background signal issues in binding assays.
  • GFP green fluorescent protein
  • phycobiliproteins allophycocyanin, phycocyanin, phycoerythrin and phycoerythrocyanin
  • fluorophore-conjugated probes require fewer steps compared to the use of enzymatic labels because there is no substrate development step to perform. While the protocol is shorter, fluorescent detection requires special equipment and the sensitivity is not a high as that which can be obtained with enzymatic chemiluminescent systems. Although not as sensitive as enzymatic detection, fluorescent detection methods reduce chemical waste and have the added advantage of multiplex compatibility (using more than one fluorophore in the same experiment).
  • two markers may be used in order to detect proximity of two substances, e.g. the test compound or the known Slfn2 ligand and the Slfn2 protein.
  • Papain and cathepsin L and H are known ligands for Slfn2.
  • the markers may be, e.g. one radioactive or fluorescent marker and one scintillator (e.g. for a scintillation proximity assay) or two fluorescent markers may be used (e.g. for FRET).
  • the Slfn2 protein and the test substance could be labeled with a first and a second marker.
  • test substance is bound to the protein, and the labels are therefore in close proximity, energy could be transferred from the first to the second label, thus detecting the interacting of Slfn2 protein and test substance.
  • This test could be designed as a competition binding test, wherein a known Slfn2 ligand carries one of the labels.
  • suitable marker combinations include:
  • radiolabels 3 H, 33 P, 35 S or 14 C, 125 l combined with scintillator such as Yttrium silicate or polyvinyl-toluene, e.g. compartmented in a microparticle or
  • a donor fluorescent markers such as fluorescein, Lucifer Yellow, B-phycoerythrin, 9- acridineisothiocyanate, Lucifer Yellow VS, 4-acetamido-4'-isothiocyanatostilbene- 2,2'-disulfonic acid, 7-diethylamino-3-(4'-isothiocyanatophenyl)-4-methylcoumarin, succinimdyl 1 -pyrenebutyrate, and 4-acetamido-4'-isothiocyanatostilbene-2,2'- disulfonic acid derivatives combined with a acceptor fluorescent marker such as LC- Red 610, LC -Red 640, LC-Red 670, LC -Red 705, Cy5, Cy5.5, Lissamine rhodamine B sulfonyl chloride, tetramethyl rhodamine isothiocyanate, rhodamine x isothiocyanate
  • Detection of Slfn2 can also refer to detection of Slfn2 mRNA:
  • RNA samples may be first separated by size via electrophoresis in an agarose gel under denaturing conditions. The RNA is then transferred to a membrane, crosslinked and hybridized with a labeled probe.
  • Nonisotopic or high specific activity radio labeled probes can be used including random- primed, nick-translated, or PCR-generated DNA probes, in vitro transcribed RNA probes, and oligonucleotides.
  • sequences with only partial homology e.g., cDNA from a different species or genomic DNA fragments that might contain an exon may be used as probes.
  • the Nuclease Protection Assay is an extremely sensitive method for the detection and quantitation of specific mRNAs.
  • the basis of the NPA is solution hybridization of an antisense probe (radio labeled or non-isotopic) to an RNA sample. After hybridization, single-stranded, unhybridized probe and RNA are degraded by nucleases. The remaining protected fragments are separated e.g. on an acrylamide gel. Solution hybridization is typically more efficient than membrane-based hybridization, and it can accommodate up to 100 g of sample RNA, compared with the 20-30 g maximum of blot hybridizations.
  • NPAs are also less sensitive to RNA sample degradation than Northern analysis since cleavage is only detected in the region of overlap with the probe (probes are usually about 100-400 bases in length).
  • RT-PCR an RNA template is copied into a complementary DNA (cDNA) using a retroviral reverse transcriptase. The cDNA is then amplified exponentially by PCR.
  • Relative quantitative RT-PCR involves amplifying an internal control simultaneously with the gene of interest. The internal control is used to normalize the samples. Once normalized, direct comparisons of relative abundance of a specific mRNA can be made across the samples.
  • Competitive RT-PCR is used for absolute quantitation.
  • This technique involves designing, synthesizing, and accurately quantitating a competitor RNA that can be distinguished from the endogenous target by a small difference in size or sequence.
  • Known amounts of the competitor RNA are added to experimental samples and RT-PCR is performed. Signals from the endogenous target are compared with signals from the competitor to determine the amount of target present in the sample.
  • the above methods may include nucleic acid labeling.
  • a series of techniques are known to the skilled person allowing for labeling of DNA, RNA or oligonuleotides. These include for example Nick translational labeling, random primed DNA labeling, PCR labeling of DNA probes and oligonucleotide 375' end labeling, transcriptional labeling of RNA probes, oligonucleotide 375' end labeling and oligonucleotide tailing.
  • the nick translation method is based on the ability of DNase I to introduce randomly distributed nicks into DNA.
  • DNA polymerase I synthesizes DNA complementary to the intact strand in a 5' ⁇ 3' direction using the 3'-OH termini of the nick as a primer.
  • the 5' ⁇ 3' exonucleolytic activity of DNA Polymerase I simultaneously removes nucleotides in the direction of synthesis.
  • the polymerase activity sequentially replaces the removed nucleotides with isotope-labeled or hapten-labeled deoxyribonucleoside triphosphates.
  • the unlabeled DNA in the reaction is thus replaced by newly synthesized labeled DNA.
  • Common labels include digoxigenin-, biotin-, or fluorochromes such as fluorescein or tetramethylrhodamin.
  • the method of "random primed" DNA labeling is based on the hybridization of a mixture of all possible hexanucleotides to the DNA to be labeled. All sequence combinations are represented in the hexanucleotide primer mixture, which leads to binding of primer to the template DNA in a statistic manner. Thus an equal degree of labeling along the entire length of the template DNA is guaranteed.
  • the complementary strand is synthesized from the 3' OH termini of the random hexanucleotide primer using Klenow enzyme, labeling grade. Modified deoxyribonucleoside triphosphates (e.g.
  • PCR polymerase chain reaction
  • digoxigenin a steroid hapten
  • the digoxigenin is usually coupled to dUTP via an alkali-labile ester bond.
  • the labeled dUTP can be easily incorporated by enzymatic nucleic-acid synthesis using DNA polymerases.
  • Oligonucleotides may enzymatically be labeled at their 3 ' -end with terminal transferase either by incorporation of a label such as single digoxigenin-labeled dideoxyuridine- triphosphate (DIG-ddUTP) or by the addition of a longer nucleotide tail.
  • Terminal Transferase catalyzes the template independent addition of deoxy- and dideoxynucleoside triphosphates to the 3 ⁇ ends of double and single-stranded DNA fragments and oligonucleotides.
  • Terminal transferase incorporates digoxigenin-, biotin-, and fluorochrome-labeled deoxy- and dideoxynucleotides as well as radioactive labeled deoxy-and dideoxynucleotides.
  • oligonucleotides may be labeled at the 5 ' -terminus, e.g. by reacting with a phosphoramidite in a final step according to the classical solid phase phosphoramidite synthesis method. By this process a 5 ' -terminal amino function is created. Treatment with ammonia releases the oligonucleotide from the support and cleaves the protecting groups. In the subsequent step the digoxigenin moiety is introduced at the 5 ' -position.
  • Different labels are known which may be used in the above labeling methods. Some of them including their detection are exemplarily described in the following:
  • Biotin-labeled compounds can be detected for example by anti-biotin antibodies or by streptavidin conjugates.
  • Anti-biotin antibodies e.g. monoclonal anti-biotin antibody or Fab-fragment, conjugated with alkaline phosphatase (AP)
  • AP alkaline phosphatase
  • Biotin-labeled compounds can be detected for example by anti-biotin antibodies or by streptavidin conjugates.
  • Anti-biotin antibodies e.g. monoclonal anti-biotin antibody or Fab-fragment, conjugated with alkaline phosphatase (AP)
  • AP alkaline phosphatase
  • This method of detection may be employed for detection of biotin labeled nucleic acids on membranes (e.g. Southern blots, dot blots), in cells and tissues (e.g. in situ hybridization), immunoblotting, immunohistochemistry or ELISA.
  • Streptavidin conjugates are used for the detection of biotin-labeled substances (e.g., biotinylated antibodies) which can be used for several immunological detection systems.
  • biotin-labeled substances e.g., biotinylated antibodies
  • streptavidin e.g. from Streptomyces avidinii could be coupled to alkaline phosphatase or to ⁇ -peroxidase. This method of detection may be employed with immunoblotting, immunohistochemistry or ELISA.
  • Probe-target hybrids may be detected with an enzyme-linked immunoassay. This immunochemical detection step is usually more sensitive than radioactive detection procedures. In this assay, the membrane may be blocked to prevent non-specific interaction of the antibody with the filter. Alkaline phosphatase-conjugated antibody, specific for digoxigenin, recognizes the digoxigenin molecule on the labeled hybrid. Addition of an alkaline phosphatase substrate allows the visualization of the hybrids.
  • suitable substrates for alkaline phosphatase such as disodium 3-(4-methoxyspiro ⁇ 1 ,2-dioxetane-3,2-(5-chloro)tricyclo [3.3.1 .1 3,7 ]decan ⁇ -4- yl)phenyl phosphate or disodium 4-chloro-3-(methoxyspiro ⁇ 1 ,2-dioxetane-3,2-(5- chloro)tricyclo [3.3.1 .1 3,7 ]decan ⁇ -4-yl)phenyl phosphate belong to the group of the dioxetane phenyl phosphates.
  • an intermediate Upon dephosphorylation by alkaline phosphatase, an intermediate is formed whose decomposition results in light emission which can be recorded e.g. on X-ray film.
  • Colorimetric detection of DIG-labeled probes is usually performed with colorless substrates which form a redox system.
  • Examples are like 5-bromo-4-chloro-3-indolyl- phosphate and 4-Nitro-blue-tetrazolium-chloride.
  • 5-bromo-4-chloro-3-indolyl-phosphate is oxidized by the alkaline phosphatase to indigo by release of a phosphate group.
  • 4-Nitro-blue-tetrazolium-chloride is reduced to diformazan. The reaction products form a water insoluble dark blue to brownish precipitate, depending on the type of membrane.
  • reporter molecules can be coupled to detecting antibodies to visualize the specific probe-target hybridization including, but not limited to, enzyme-coupled antibodies, fluorochrome-labeled antibodies (detection by fluorescent microscope and specific filters which allow visualization of the wavelength emitted by the fluorescent dye) and antibodies coupled to colloidal gold (detection by electron microscope on cryostatic sections).
  • Multiple simultaneous hybridizations can be performed by using combinations of digoxigenin-, biotin- and fluorochrome-labeled probes to localize different chromosomal regions or different RNA sequences in one preparation.
  • Such multiprobe experiments are made possible by the availability of different fluorescent dyes coupled to antibodies. These include fluorescein or FITC (fluorescein isothiocyanate; yellow), rhodamine or TRITC (tetramethylrhodamine isothiocyanate; red) and AMCA (amino-methylcoumarin acetic acid; blue).
  • Suitable diagnostic assays comprise the detection of Slfn2 protein:
  • Suitable methods for detecting a protein include e.g. detection of protein immunostaining, protein immunoprecipitation, Immunoelectrophoresis, immunoblotting, Western blotting, spectrophotometry, enzyme assays etc..
  • the method may require protein purification prior to the detection, which could involve protein isolation (e.g. by chromatography methods, protein extraction, protein solubilization, gel electrophoresis, and electrofocusing).
  • Protein immunostaining is an antibody-based method to detect a specific protein in a sample.
  • the term immunostaining was originally used to refer to the immunohistochemical staining of tissue sections. Now however, immunostaining encompasses a broad range of techniques used in histology, cell biology, and molecular biology that utilize antibody-based staining methods. Immunohisto- or -cytochemistry of tissue sections or cells which are preserved by fixation.
  • Western blotting allows the detection of specific proteins (native or denatured) from extracts made from cells or tissues, before or after any purification steps. Proteins are generally separated by size using gel electrophoresis before being transferred to a synthetic membrane (typically nitrocellulose or PVDF) via dry, semi-dry, or wet blotting methods. The membrane can then be probed using antibodies using methods similar to immunohistochemistry, but without a need for fixation. Detection is typically performed using peroxidase linked antibodies to catalyze a chemiluminescent reaction.
  • Western blotting is a routine molecular biology method that can be used to semi quantitatively or quantitatively compare protein levels between extracts.
  • Western blotting is an analytical technique used to detect specific proteins in a given sample of tissue homogenate or extract. It uses gel electrophoresis to separate proteins by the length of the polypeptide (denaturing conditions) or by the 3-D structure of the protein (native/ non-denaturing conditions).
  • the enzyme-linked immunosorbent assay or ELISA is a diagnostic method for quantitatively or semi-quantitatively determining protein concentrations from blood plasma, serum or cell/tissue extracts in a multi-well plate format (usually 96-wells per plate). Broadly, proteins in solution are adsorbed to ELISA plates. Antibodies specific for the protein of interest are used to probe the plate. Background is minimized by optimizing blocking and washing methods (as for IHC), and specificity is ensured via the presence of positive and negative controls. Detection methods are usually colorimetric or chemiluminescence based.
  • Electron microscopy or EM can be used to study the detailed micro architecture of tissues or cells.
  • Immuno-EM allows the detection of specific proteins in ultrathin tissue sections.
  • Antibodies labeled with heavy metal particles e.g. gold
  • transmission electron microscopy While powerful in detecting the sub- cellular localization of a protein, immuno-EM can be technically challenging, expensive, and require rigorous optimization of tissue fixation and processing methods.
  • an “individual” refers to a human or a non-human animal (e.g. a mammal, avian, reptile, fish, amphibian or invertebrate; preferably an individual that can either benefit from one of the different aspects of present invention (e.g. a method of treatment or a drug identified by present methods) or that can be used as laboratory animal for the identification or characterisation of a drug or a method of treatment.
  • the individual can e.g. be a human, a wild-animal, domestic animal or laboratory animal; examples comprise: mammal, e.g.
  • human, non-human primate chimpanzee, bonobo, gorilla
  • dog, cat rodent
  • rodent e.g. mouse, guinea pig, rat, hamster or rabbit, horse, donkey, cow, sheep, goat, pig, camel
  • avian such as duck, dove, turkey, goose or chick
  • reptile such as: turtle, tortoise, snake, lizard, amphibian such as frog (e.g. Xenopus laevis); fish such as koy or zebrafish
  • invertebrate such as a worm (e.g. c.elegans) or an insect (such as a fly, e.g. drosophila melanogaster).
  • the term individual also comprises the different morphological developmental stages of avian, fish, reptile or insects, such as egg, pupa, larva or imago.
  • the individual may be an individual which is suspected to experience pain.
  • the individual may be diagnosed and/or prognosed to experience pain.
  • the individual as mentioned in the method above may also be an individual which is experiencing pain.
  • the individual may be retested for experiencing pain and may be diagnosed and/or prognosed to still experiencing pain.
  • sample preferably refers to a biologically sample.
  • sample or “sample of interest” are used interchangeably herein, referring to a small part intended to represent the whole of a tissue, an organ or an individual.
  • samples include but are not limited to fluid samples such as cerebrospinal fluid, blood, serum, plasma, synovial fluid, urine, saliva, and lymphatic fluid, or solid samples such as biopsy samples, tissue, and tissue-extracts, e.g. taken from nervous tissue (e.g from the spinal cord), skin, muscle, cartilage, bone, synovium, perichondrium, capsule, and connective tissue.
  • cell cultures or tissue cultures such as but not limited to cultures of neural cells.
  • Analysis of a sample may be accomplished on a visual or chemical basis.
  • Visual analysis includes but is not limited to microscopic imaging or radiographic scanning of a tissue, organ or individual allowing for morphological evaluation of a sample.
  • Chemical analysis includes but is not limited to the detection of the presence or absence of specific indicators or alterations in their amount or level.
  • a tissue sample may be removed from a subject by conventional biopsy techniques or a blood sample may be taken from a subject by conventional blood collection techniques.
  • the sample e.g. tissue or blood sample, may be obtained from a subject prior to initiation of the therapeutic treatment, during the therapeutic treatment, and/or after the therapeutic treatment, e.g. with a pain reducing compound.
  • the sample is a body fluid sample, a tissue sample, a cell colony sample, a single cell sample or a cell culture sample. More preferably, the tissue sample is a section or an explant sample, e.g. an explant sample of dorsal root ganglia or spinal cord.
  • body fluid sample refers to a liquid sample derived from the body of a subject. Said body fluid sample may be a blood, urine, cerebrospinal fluid, cerumen (earwax), endolymph, perilymph, gastric juice, mucus, peritoneal fluid, pleural fluid, saliva, or sebum (skin oil) sample including components or fractions thereof. Said body fluid samples may be mixed or pooled.
  • a body fluid sample may be a mixture of a blood sample and anurine sample or a mixture of a blood sample and cerebrospinal fluid sample.
  • a "body fluid sample” may be provided by removing a body liquid from a subject, but may also be provided by using previously isolated body fluid sample material.
  • the blood sample of a subject is whole blood or a blood fraction such as serum or plasma. It is also particularly preferred to use blood cells also known as hemopoietic cells.
  • the tissue sample has a weight of between 0.1 and 500 mg, more preferably of between 0.5 and 250 mg, and most preferably of between 1 and 50 mg, i.e.
  • the cell sample e.g. cell colony sample or cell culture sample
  • the cell sample consists of between 100 and 1000 cells, more preferably of between 200 and 800 cells, and most preferably of between 400 and 600 cells.
  • the body fluid sample has a volume of between 0.1 and 20 ml, more preferably of between 0.5 and 10 ml, more preferably between 1 and 8 ml and most preferably between 2 and 5 ml, i.e. 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 ml. More preferably, the blood sample has a volume of between 0.1 and 20 ml, more preferably of between 0.5 and 10 ml, and most preferably of between 1 and 5 ml, i.e. 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 ml.
  • a reference value is a standard value for a certain parameter (e.g. expression level of a certain indicator/biomarker molecule) that is typical for a certain status (e.g. a disease status or health status)
  • a reference individual is an individual that has been selected for comparison and has a certain health state or disease
  • a reference sample can e.g. be a sample from a reference individual or an artificial sample with a characteristic level of a certain indicator or biomarker typical for a disease state or health state.
  • reference sample refers to a sample which is analysed in a substantially identical manner as the sample of interest and whose information is compared to that of the sample of interest. A reference sample thereby provides a standard allowing for the evaluation of the information obtained from the sample of interest.
  • a reference sample may be derived from a healthy or normal tissue, organ or individual, thereby providing a standard of a healthy status of a tissue, organ or individual. Differences between the status of the normal reference sample and the status of the sample of interest may be indicative of the risk of disease development or the presence or further progression of such disease or disorder.
  • a reference sample may be derived from an abnormal or diseased tissue, organ or individual thereby providing a standard of a diseased status of a tissue, organ or individual. Differences between the status of the abnormal reference sample and the status of the sample of interest may be indicative of a lowered risk of disease development or the absence or bettering of such disease or disorder.
  • a reference sample may also be derived from the same tissue, organ, or individual as the sample of interest but has been taken at an earlier time point. Differences between the status of the earlier taken reference sample and the status of the sample of interest may be indicative of the progression of the disease, i.e. a bettering or worsening of the disease over time.
  • a reference sample was taken at an earlier or later time point in case a period of time has lapsed between taking of the reference sample and taking of the sample of interest.
  • Such period of time may represent years (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 years), months (1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 months), weeks (e.g. 1 , 2, 3, 4, 5, 6, 7, 8 weeks), days (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500 days), hours (1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 hours), minutes (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60 minutes), or seconds (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60 seconds).
  • years e.g. 1
  • the reference sample representative for a status or stage of pain may be from a control subject known to experience pain.
  • the control sample is from a control subject known to experience algesia, particularly hyperalgesia.
  • the control subject may be a mammal such as a human, rodent (e.g. rat, hamster, or mouse) or monkey, or may be another animal than a mammal such as an avian.
  • both the sample or value and the reference sample or value are from subjects of the same species (e.g. human), more preferably of the same gender (e.g. female or male) and/or of a similar age or phase of life (e.g. infant, young child, juvenile, adult, or elderly).
  • the reference or reference sample in the different aspects and embodiments of present invention is preferably derived from a healthy individual, a diseased individual, or from the same individual as the sample of interest.
  • the reference (e.g. reference value) or reference sample was taken from the same individual as the sample of interest, the reference (e.g. reference value) or reference sample was preferably taken at an earlier or later time point then the sample of interest.
  • the time period which has lapsed between taking of the reference (e.g. reference value) or reference sample and taking of the reference (e.g. reference value) or sample or value of interest preferably represents years (e.g.
  • the reference sample is a reference sample with a level of Slfn2 representative for a healthy individual or representative for the presence or absence of an altered tissue status or disease or representative for an increased or decreased risk of developing an altered tissue status or disease.
  • an elevated level of Slfn2 in the reference or value or sample of interest in comparison to said reference (value) or reference sample indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
  • a similar level of Slfn2 indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
  • an elevated level of Slfn2 in the individual/value/sample of interest indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
  • a lowered level of Slfn2 in the sample of interest indicates (a) an alteration of the tissue status or an improvement or absence of a disease and/or (b) a decreased risk to develop a deteriorated tissue status or a disease and/or (c) a declined progression of the deteriorated tissue status or the disease.
  • a similar level of Slfn2 in the sample of interest indicates (a) a similar risk to develop a deteriorated tissue status or a disease and/or (b) a stagnation in the progression of a deteriorated tissue status or a disease, and/or (c) a persistence of the deteriorated tissue status or the disease in the individual.
  • the reference (value) or reference sample is derived from a healthy individual or from an individual with a decreased risk of developing an altered tissue status or disease or comprises a level of Slfn2 representative of a healthy individual or of a status of disease-absence or for a decreased risk of developing an altered tissue status or disease, wherein an elevated level of Slfn2 indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
  • the reference (value) or reference sample is derived from a diseased individual or from an individual with an increased risk of developing an altered tissue status or disease or comprises a level or amount of Slfn2 representative for a diseased individual or for a status of disease-presence or for an increased risk of developing an altered tissue status or disease, wherein a similar level of Slfn2 indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
  • an elevated level of Slfn2 in the sample of interest indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
  • a lowered level of Slfn2 in the sample of interest indicates (a) an alteration of the tissue status or an improvement or absence of a disease and/or (b) a decreased risk to develop a deteriorated tissue status or a disease and/or (c) a declined progression of the deteriorated tissue status or the disease.
  • a similar level of Slfn2 in the sample of interest indicates (a) a similar risk to develop a deteriorated tissue status or a disease and/or (b) a stagnation in the progression of a deteriorated tissue status or a disease, and/or (c) a persistence of the deteriorated tissue status or the disease in the individual.
  • lowered or “decreased”, especially in the context of the level of Slfn2 refer to the level of Slfn2 in the sample being reduced in comparison to the reference or reference sample.
  • elevated or “increased”, especially in the context of the level of Slfn2 refer to the level of Slfn2 in the sample being higher in comparison to the reference or reference sample.
  • a miRNA that is detectable in higher amounts in cerebrospinal fluid or spinal cord tissue of one individual suffering from pain than in cerebrospinal fluid or spinal cord tissue of individuals not suffering from pain has an elevated level.
  • an elevated level in a patient sample indicates the presence of a pain-associated disease or pain-associated tissue state or pain or an increased susceptibility or increased probability to develop a pain-associated disease or pain-associated tissue state or pain, especially the presence or an increased susceptibility to neuropathic algesia or neuropathic pain.
  • treat means accomplishing one or more of the following: (a) reducing the severity of the disorder; (b) limiting or preventing development of symptoms characteristic of the disorder(s) being treated; (c) inhibiting worsening of symptoms characteristic of the disorder(s) being treated; (d) limiting or preventing recurrence of the disorder(s) in an individual that have previously had the disorder(s); and (e) limiting or preventing recurrence of symptoms in individuals that were previously symptomatic for the disorder(s).
  • prevent means preventing that such disease or disorder occurs in patient.
  • a "patient” means any mammal, reptile or bird that may benefit from a treatment with the compounds described herein.
  • a “patient” is selected from the group consisting of laboratory animals (e.g. mouse or rat), domestic animals (including e.g. guinea pig, rabbit, chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog), or primates including chimpanzees and human beings. It is particularly preferred that the "patient” is a human being.
  • laboratory animals e.g. mouse or rat
  • domestic animals including e.g. guinea pig, rabbit, chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog
  • primates including chimpanzees and human beings. It is particularly preferred that the "patient” is a human being.
  • present invention relates to Slfn2 for use as an indicator of a pain- related tissue status or a pain related disease or as an indicator of the risk for developing a pain-related tissue status or disease.
  • Slfn2 correlates with pain, especially neuropathic pain in mice leads to the conclusion that Slfn2 can be used as indicator/biomarker for tissue statuses or diseases that correlate with pain and for pain itself.
  • Slfn2 is indicative of a pain-related tissue status or a pain-related disease.
  • the presence, absence or altered amounts of Slfn2 are thus a sign for the presence, absence or progression of a pain-related tissue status or disease.
  • a second aspect of present invention relates to Slfn2 for use in the diagnosis or prognosis of a pain-related tissue status or of a pain-related disease.
  • Slfn2 is used as an indicator of a tissue status or a disease in the context of present invention.
  • the level of Slfn2 is indicative of a pain-related tissue status or the presence of a pain-related disease and/or the risk of developing a pain-related tissue status or a pain-related disease and/or the progression or a stage of a pain-related tissue status or a pain-related disease in an individual.
  • the level of Slfn2 in the first and second aspect of present invention is the expression level of Slfn2 or the level or the amount (number) of Slfn2 gene (copies), wherein the level can be the level of Slfn2 in an individual (wherein Slfn2 needs to be determined by a method able to directly determine and quantify Slfn present in an individual without a sample needing to be taken) or in a sample of an individual.
  • the sample to be used in the first and second aspect of present invention can be any suitable sample, e.g. such as herein defined wherein cerebrospinal fluid is one of the preferred embodiments.
  • the reference(s) or reference sample(s) can be any suitable reference(s) or reference sample(s), e.g. such as herein defined.
  • the reference is selected from the group consisting of
  • an individual having a pain-related tissue status or disease especially one that is known to be associated with a certain level of Slfn2
  • a pain-related tissue status or disease especially one that is known to be associated with a certain level of Slfn2
  • the reference is (derived from) an individual with a decreased
  • the reference is a sample that is selected from the group consisting of a) a reference sample derived from an individual not having a pain-related tissue status or disease,
  • a reference sample from an individual having a pain-related tissue status or disease b) a reference sample from an individual having a pain-related tissue status or disease, c) a reference sample from the same individual as the sample to be tested taken at an earlier or later time point or
  • the reference sample is derived from an individual with a decreased risk of developing a pain-related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual not having a pain-related tissue status or disease or for a status of disease-absence or for a decreased risk of developing a pain-related tissue status or disease, wherein an elevated level of Slfn2 in the tested sample indicates
  • the reference is the same individual at an earlier time point or the sample is derived from the same individual as the tested sample and was taken at an earlier time point, wherein
  • the reference is the same individual as the individual of interest at an earlier time point or the sample is derived from the same individual as the individual of interest, wherein the sample is derived from the same individual as the tested sample and was taken at an earlier time point, and wherein
  • the sample e.g. the reference or tested sample of the first, second, third and fourth aspect of present invention can be any suitable sample, e.g. such as disclosed herein e.g. tissue such as extraneural or neural tissue or one or more extraneural or neural cells and/or body fluid such as cerebrospinal fluid. It can also be suitable to take samples of different origin such as a tissue sample and a body fluid sample.
  • the Slfn2 according to one of the embodiments of the first and second aspect of present invention can e.g. a Slfn2 gene or gene product, preferably a Slfn2 mRNA, or Slfn2 protein or a variant (such as a functionally active variant) thereof.
  • Slfn2 is a gene and the gene
  • a. is a naturally occurring or non-naturally occurring Slfn2 gene variant, or
  • amino acid sequence that has at least 80%, at least 90% at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
  • vii. carries a variant AAA domain, preferably with 50, 60, 70, 80, 90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2.
  • Slfn2 is a gene product, wherein the gene product
  • a) is a naturally occurring Slfn2 protein variant, preferably a homolog or
  • b) is a Slfn2 protein or a functionally active variant thereof having
  • amino acid sequence i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
  • amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
  • c) is a Slfn2 nucleic acid encoding a protein as defined in b), or
  • d) is a Slfn2 nucleic acid having
  • nucleic acid sequence that has at least 80%, at least 90%, at least
  • the first or second aspects of present invention are e.g. suitable for diagnosing and/or prognosing pain in an in vitro or in vivo test model (i.e. a test animal, such as a non- human test animal, e.g. in the context of clinical studies) or in a patient (such as a human or non-human animal).
  • an in vitro or in vivo test model i.e. a test animal, such as a non- human test animal, e.g. in the context of clinical studies
  • a patient such as a human or non-human animal
  • Another embodiment of the first and second aspect of present invention comprises determining the expression level of Slfn2 and preferably comprising determining the expression level of Slfn2 on the gene, mRNA or protein level.
  • the tissue status is a pain-associated tissue status, such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue, the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
  • a pain-associated tissue status such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue
  • the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by
  • the pain-related or pain-associated disease is a pain, e.g.
  • nociceptive pain nociceptive pain, neuropathic pain, inflammatory pain, or a mixture thereof, (iii) disease-associated pain, or
  • the acute pain is somatogenetic (organic) pain
  • the chronic pain is somatogenetic (organic) pain or psychogenic
  • the nociceptive pain is superficial somatic pain or deep pain, preferably deep somatic pain or visceral pain
  • the neuropathic pain is central neuropathic pain or peripheral neuropathic pain
  • the mixed pain is a mixture of nociceptive pain and neuropathic pain
  • the disease-associated pain is pain associated with depression, migraine, epilepsy, cancer, or diabetes, or
  • the algesia is hyperalgesia.
  • the pain-related or pain-associated disease is characterized by an altered level of pain- related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
  • the prognosis comprises predicting or estimating the occurrence or reoccurrence, preferably the severity of occurrence or reoccurrence, of pain.
  • present invention relates to a method of identifying the occurrence or reoccurrence, preferably the severity of occurrence or reoccurrence, of pain.
  • the method comprising detecting the level of Slfn2.
  • present invention relates to a method of identifying
  • the method comprising detecting the level of Slfn2.
  • the individual of the fourth aspect of present invention can be any suitable individual such as herein defined.
  • the fourth aspect of present invention further comprises comparing the level of Slfn2 (e.g. the level of Slfn2 in the individual of interest (i.e. the test animal or the patient)) to the level of Slfn2 in one or more references or reference samples.
  • the level of Slfn2 can e.g. be the level of Slfn2 in an individual (wherein Slfn2 needs to be determined by a method able to directly determine and quantify Slfn present in an individual without a sample needing to be taken) or in a sample of an individual.
  • the level of Slfn2 is the expression level of Slfn2 or the level or the amount (number) of Slfn2 gene (copies).
  • the reference is selected from the group consisting of an individual not having a pain-related tissue status or disease, an individual having a pain-related tissue status or disease or the same individual as the individual to be tested at an earlier or later time point or a value representative for the level of slfn2 in absence of the a pain-related tissue status or disease, in presence of a pain-related altered tissue status or disease or for an increased or decreased risk of developing a pain-related altered tissue status or disease.
  • the reference sample is selected from the group consisting of a reference sample derived from an not having a pain-related tissue status or disease individual, a reference sample from an individual having a pain-related tissue status or disease, and a reference sample from the same individual as the sample to be tested taken at an earlier or later time point or a reference sample with a level or amount of slfn2 representative for an individual having or not having a pain-related tissue status or disease or for the absence or presence of the pain-related disease or altered tissue status or representative for an increased or decreased risk of developing a pain-related tissue status or disease.
  • the reference is an individual not having a pain-related tissue status or disease or an individual with a decreased risk of developing a pain-related tissue status or disease or a value representative of the absence of a pain-related tissue status or disease, or the Slfn2 or method according to embodiment 8, wherein the reference sample is derived from an individual not having a pain-related tissue status or disease or from an individual with a decreased risk of developing a pain-related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual not having a pain-related tissue status or disease or for a status of disease-absence or for a decreased risk of developing a pain-related tissue status or disease, wherein an elevated level of Slfn2 in the tested sample indicates
  • the reference is an individual having a pain-related tissue status or disease or an individual with an increased risk of developing a pain-related tissue status or a disease or a value representative of the presence of a pain-related tissue status or disease, or Slfn2 or method according to embodiment 8, wherein the reference sample is derived from an individual having a pain-related tissue status or disease or from an individual with an increased risk of developing a pain-related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual having a pain-related tissue status or disease or for a status of disease-presence or for an increased risk of developing a pain-related tissue status or disease, wherein a similar or higher level of Slfn2 in the tested sample indicates
  • the reference is the same individual at an earlier time point or Slfn2 or method of embodiment 8, wherein the sample is derived from the same individual as the tested sample and was taken at an earlier time point, wherein
  • the tested sample is tissue and/or body fluid, wherein the tissue sample can e.g. be extraneural or neural tissue or an extraneural or a neural cell, and wherein the body fluid sample can e.g. be cerebrospinal fluid.
  • the pain- related disease is pain or a pain-associated disease, preferably a pain such as defined above and below.
  • Slfn2 is a Slfn2 gene or gene product, preferably a Slfn2 mRNA, or Slfn2 protein or a functionally active variant thereof.
  • Slfn2 is a gene, wherein the gene
  • a. is a naturally occurring or non-naturally occurring Slfn2 gene variant, or
  • amino acid sequence i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
  • amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
  • amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to
  • vii. carries a variant AAA domain, preferably with 50, 60, 70, 80, 90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2.
  • Slfn2 is a gene product, wherein the gene product
  • a) is a naturally occurring Slfn2 protein variant, preferably a homolog or
  • b) is a Slfn2 protein or a functionally active variant thereof having
  • amino acid sequence i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
  • amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
  • c) is a Slfn2 nucleic acid encoding a protein as defined in b), or
  • d) is a Slfn2 nucleic acid having
  • nucleic acid sequence that has at least 80%, at least 90%, at least
  • the fourth aspect and its different embodiments are especially suitable for diagnosing and/or prognosing pain in an in vitro or in vivo test model (i.e. a test animal, such as a non-human test animal, e.g. in the context of clinical studies) or in a patient (such as a human or non-human animal).
  • an in vitro or in vivo test model i.e. a test animal, such as a non-human test animal, e.g. in the context of clinical studies
  • a patient such as a human or non-human animal
  • the fourth aspect of present invention comprises determining the expression level of Slfn2 and preferably comprises determining the expression level of Slfn2 on the gene, mRNA or protein level.
  • the tissue status is a pain-associated tissue status, such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue, the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
  • a pain-associated tissue status such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue
  • the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by
  • the pain- related or pain-associated disease is a pain.
  • the pain is (i) acute pain, subacute pain, or chronic pain
  • the acute pain is somatogenetic (organic) pain
  • the chronic pain is somatogenetic (organic) pain or psychogenic
  • the nociceptive pain is superficial somatic pain or deep pain, preferably deep somatic pain or visceral pain
  • the neuropathic pain is central neuropathic pain or peripheral neuropathic pain
  • the mixed pain is a mixture of nociceptive pain and neuropathic pain
  • the disease-associated pain is pain associated with depression, migraine, epilepsy, cancer, or diabetes, or
  • the algesia is hyperalgesia.
  • the disease is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
  • the method relates to diagnosis, wherein the diagnosis comprises
  • the method relates to prognosis, wherein the prognosis comprises predicting or estimating the occurrence or reoccurrence, preferably the severity of occurrence or reoccurrence, of pain.
  • the kit for use in a method according to the fourth aspect and any of its embodiments, wherein the kit comprises one or more means of detecting Slfn2.
  • present invention relates to a kit for detecting Slfn2 comprising
  • a data carrier comprising instructions for a method according to the fourth aspect of present invention and any one its embodiments and/or
  • kit is an article of manufacture that comprises at least the components as defined above and optionally one or more further components, the term kit in the context of present invention is understood as herein defined.
  • the kit according to the fifth and sixth aspect of present invention comprises at least one or more means for detecting Slfn2, it can in addition, comprise one or more means for detecting another indicator of a pain-related tissue status or a pain-related disease such as pain, e.g. as listed above.
  • the kit comprises a means for detecting Slfn2 or a means for detecting Slfn2 and a data carrier comprising instructions for a method according to the fourth aspect of present invention and any one its embodiments, it can also comprise a means for detecting Slfn2 and a container, it can also comprise a means for detecting Slfn2 and a means for detecting another indicator of a pain-related tissue status or pain-related disease such as pain, it can also comprise a means for detecting Slfn2 and a data carrier comprising instructions for a method according to the fourth aspect of present invention and any one its embodiments and a container it can also comprise a means for detecting Slfn2 and a data carrier comprising instructions for a method according to the fourth aspect of present invention and any one its embodiments and another indicator of a pain-related tissue status or pain-related disease such as pain, it can also comprise a means for detecting Slfn2 and another indicator of a pain-related tissue status or pain-related disease such as pain
  • the means for detecting Slfn2 is a means for determining the expression level of Slfn2, preferably on the gene, protein or RNA level.
  • the term means of detecting Slfn2 or means of detection of Slfn2 is understood as herein defined.
  • the means for detecting Slfn2 is selected from the group consisting of nucleic acid, preferably DNA, RNA or PNA, peptide and protein, preferably monoclonal or polyclonal antibody or protein scaffold.
  • the kit according to the fifth and sixth aspect of present invention can also comprise more than one means, e.g. 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 means of detection.
  • the kit comprises
  • quality information such as information about the lot/batch number of the means for detecting Slfn2 and/or of the kit, the manufacturing or assembly site or the expiry or sell-by date, information concerning the correct storage or handling of the kit,
  • the data carrier can thus comprise information according to (1 ), (2), (3), (4), (5), (6) and/or (7).
  • the data carrier comprises at least information according to (1 ) or information according to (2) or information according to (3) or information according to (4) or information according to (5) or information according to (6) or information according to (7);
  • the data carrier can also comprise (1 ) and (2) or (1 ) and (3) or (1 ) and (4) or (1 ) and (5) or (1 ) and (6) or (1 ) and (7) or (2) and (3) or (2) and (6) or (2) and (5) or (2) and (6) or (2) and (7) or (3) and (4) or (3) and (5) or (3) and (6) or (3) and (7) or (6) and (5) or (4) and (6) or (4) and (7) or (5) and (6) or (5) and (7) or (6) and (7);
  • the data carrier can also comprise (1 ) and (2) and (3) or (1 ) and (2) and (4) or (1 ) and (2) and (5) or (1 ) and (2) and (6) or (1 ) and
  • the kit is for diagnosing and/or prognosing pain by determination of Slfn2 and preferably determining the level of Slfn2 in a sample.
  • the sample can be any suitable sample, e.g such as defined herein, e.g. in the definitions or above under the first and second aspect of present invention.
  • the kit comprises a means as defined below for the eighth aspect of present invention.
  • present invention refers to a means of detecting Slfn2 for use in the diagnosis or prognosis of a tissue status or a disease.
  • the means comprises one or more nucleic acids or derivatives for detecting a Slfn2 gene or gene product or a functionally active variant thereof, wherein the nucleic acid or derivative is preferably selected from the group consisting of a nucleic acid probe, a polyamide or peptide nucleic acid (PNA), microRNA (miRNA), small interfering RNA (siRNA), primers for polymerase chain reaction (PCR), primers for reverse transcription (RT) reaction, and primers for DNA sequencing.
  • PNA polyamide or peptide nucleic acid
  • miRNA microRNA
  • siRNA small interfering RNA
  • PCR primers for polymerase chain reaction
  • RT reverse transcription
  • the means comprises a peptide, polypeptide or protein for detecting a Slfn2 gene or gene product or functionally active variant thereof, wherein the protein or polypeptide is preferably a protein ligand (such as an antibody) a fragment or derivate thereof, a protein scaffold (such as a darpin or an anticalin) or wherein the polypeptide or peptide is a probe, preferably a mass spectrometry probe.
  • the tissue status is a pain-associated tissue status, such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue, the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
  • a pain-associated tissue status such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue
  • the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by
  • the disease is a pain wherein the pain is preferably
  • nociceptive pain nociceptive pain, neuropathic pain, inflammatory pain, or a mixture thereof, (iii) disease-associated pain, or
  • the acute pain is somatogenetic (organic) pain
  • the chronic pain is somatogenetic (organic) pain or psychogenic
  • the nociceptive pain is superficial somatic pain or deep pain, preferably deep somatic pain or visceral pain
  • the neuropathic pain is central neuropathic pain or peripheral neuropathic pain
  • the mixed pain is a mixture of nociceptive pain and neuropathic pain
  • the disease-associated pain is pain associated with depression, migraine, epilepsy, cancer, or diabetes, or
  • the algesia is hyperalgesia.
  • the pain- associated or pain-related disease is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
  • the means is for use in the diagnosis or prognosis or a pain-related tissue status or a pain- related disease, preferably in a method of the third aspect of present invention.
  • the diagnosis comprises
  • the prognosis comprises predicting or estimating the occurrence or reoccurrence, preferably the severity of occurrence or reoccurrence, of pain.
  • preferred embodiments of present invention are listed but are not to be understood as limiting present invention:
  • Slfn2 for use as an indicator of a pain-related tissue status or a pain related disease or as an indicator of the risk for developing a pain-related tissue status or disease.
  • Slfn2 for use in the diagnosis or prognosis of a pain-related tissue status or of a pain- related disease.
  • the method comprising detecting the level of Slfn2. 5.
  • the reference sample is selected from the group consisting of a reference sample derived from an not having a pain-related tissue status or disease individual, a reference sample from an individual having a pain-related tissue status or disease, and a reference sample from the same individual as the sample to be tested taken at an earlier or later time point or a reference sample with a level or amount of slfn2 representative for an individual having or not having a pain- related tissue status or disease or for the absence or presence of the pain-related disease or altered tissue status or representative for an increased or decreased risk of developing a pain-related tissue status or disease.
  • the reference sample is derived from an individual not having a pain-related tissue status or disease or from an individual with a decreased risk of developing a pain- related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual not having a pain-related tissue status or disease or for a status of disease-absence or for a decreased risk of developing a pain-related tissue status or disease, wherein an elevated level of Slfn2 in the tested sample indicates
  • tissue sample is extraneural or neural tissue or an extraneural or a neural cell
  • body fluid sample is cerebrospinal fluid
  • Slfn2 according to one of the embodiments 1 -3 or 5-14 or the method according to one of the embodiments 4-14, wherein Slfn2 is a Slfn2 gene or gene product, preferably a Slfn2 mRNA, or Slfn2 protein or a functionally active variant thereof. 16. The Slfn2 or method according to embodiment 15, wherein Slfn2 is a gene and wherein the gene
  • a. is a naturally occurring or non-naturally occurring Slfn2 gene variant, or b. codes for a Slfn2 protein or a functionally active variant thereof that has
  • amino acid sequence i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
  • amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
  • vii. carries a variant AAA domain, preferably with 50, 60, 70, 80, 90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2.
  • Slfn2 or method according to embodiment 16, wherein Slfn2 is a gene product and wherein the gene product
  • a) is a naturally occurring Slfn2 protein variant, preferably a homolog or
  • b) is a Slfn2 protein or a functionally active variant thereof having
  • amino acid sequence i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
  • iii an amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or iv. an amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to SEQ ID NO:2, and/or
  • vii. carries a variant AAA domain, preferably with 50, 60, 70, 80,
  • c) is a Slfn2 nucleic acid encoding a protein as defined in b), or
  • d) is a Slfn2 nucleic acid having
  • nucleic acid sequence that has at least 80%, at least 90%, at least
  • the Slfn2 according to one of the embodiments 1 -3 or 5-18 or the method according to one of the embodiments 4-18 comprising determining the expression level of Slfn2 and preferably comprising determining the expression level of Slfn2 on the gene, mRNA or protein level.
  • a kit for detecting Slfn2 comprising (i) a means for detecting Slfn2,
  • a data carrier comprising instructions for a method according to one of the embodiments 4-19 and optionally
  • kits according to embodiment 20 or 21 wherein the means for detecting Slfn2 is a means for determining the expression level of Slfn2, preferably on the gene, protein or RNA level.
  • the means for detecting Slfn2 is selected from the group consisting of nucleic acid, preferably DNA, RNA or PNA, peptide and protein, preferably monoclonal or polyclonal antibody or protein scaffold.
  • kit according to any of embodiments 20-23, wherein the kit comprises
  • quality information such as information about the lot/batch number of the means for detecting Slfn2 and/or of the kit, the manufacturing or assembly site or the expiry or sell-by date, information concerning the correct storage or handling of the kit,
  • kits according to any of embodiments 20-24, wherein the kit is for diagnosing and/or prognosing pain by determination of Slfn2 and preferably the level of Slfn2 in a sample.
  • kit according to any of embodiments 20-25, wherein the kit comprises a means according to any of the embodiments 27-39.
  • the means according to embodiment 28 comprising one or more nucleic acids or derivatives for detecting a Slfn2 gene or gene product or functionally active variant thereof.
  • nucleic acid or derivative is selected from the group consisting of a nucleic acid probe, a polyamide or peptide nucleic acid (PNA), microRNA (miRNA), small interfering RNA (siRNA), primers for polymerase chain reaction (PCR), primers for reverse transcription (RT) reaction, and primers for DNA sequencing.
  • PNA polyamide or peptide nucleic acid
  • miRNA microRNA
  • siRNA small interfering RNA
  • PCR primers for reverse transcription (RT) reaction
  • RT reverse transcription
  • the means according to embodiment 28 comprising a peptide, polypeptide or protein for detecting a Slfn2 gene or gene product or functionally active variant thereof.
  • the protein or polypeptide is a protein ligand, preferably an antibody, a fragment or derivate thereof, a protein scaffold, such as a darpin or an anticalin, or wherein the polypeptide or peptide is a probe, preferably a mass spectrometry probe.
  • tissue status is a pain-associated tissue status, such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue, the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue. 36. The Slfn2 according to any of embodiments 1 -3 or 5
  • nociceptive pain nociceptive pain, neuropathic pain, inflammatory pain, or a mixture thereof, (iii) disease-associated pain, or
  • the acute pain is somatogenetic (organic) pain
  • the chronic pain is somatogenetic (organic) pain or psychogenic
  • the nociceptive pain is superficial somatic pain or deep pain, preferably deep somatic pain or visceral pain
  • the neuropathic pain is central neuropathic pain or peripheral neuropath pain
  • the mixed pain is a mixture of nociceptive pain and neuropathic pain
  • the disease-associated pain is pain associated with depression, migraine, epilepsy, cancer, or diabetes, or
  • the algesia is hyperalgesia. 39.
  • FIG. 1 ClustalW 2.1 (available by EMBL-EBI (EMBL-European Bioinformatics
  • sequence 1 is murine (mus musculus) Slfn2 according to
  • sequence 2 is rat (rattus norwegicus) Slfn2 according to NP_001 100501/NM_001 107031 .1 (SEQ ID NO:7)
  • sequence 3 is homo sapiens SLFN12L according to NM_001 195790.1/NP_001 182719.1 (SEQ ID NO:9).
  • the identity score with ClustalW is 83% between mSlfn2 and ratSlfn2, 50% between mSlfn2 and hsSLFN12L and 50% between rat Slfn2 and hsSLFN12L.
  • the conserved variant AAA domain is shown underlined.
  • EMBOSS needle 6.3.1 program working on Needleman-Wunsch algorithm of the EMBL EBI database .
  • the divergent AAA domains (from position 231 to 355 of murine Slfn2 and from position 231 to 361 of rat Slfn2) as outlined in the NCBI database (see respective accession number) have been marked underlined.
  • FIG. 1 Sequence alignment of murine Slfn2 (SEQ ID NO:2, sequence 1 of
  • Figure 4 Promoter structure of murine Slfn2 according to Wern-Joo et al, 2009.
  • Figure 5 shows for every individual mouse its neuropathic pain phenotype (mechanical hypersensitivity, X-axis) and the corresponding gene expression (signal intensity, Y-axis) in the L5 DRG. Mouse data are colour-coded depending on the used strain. A Pearson correlation analysis has been performed and revealed a significant positive correlation of the two parameters pain phenotype and Slfn2 gene expression. This means for individual mice that the higher the L5 DRG expression of Slfn2 in Chung-operated neuropathic mice was, the more pronounced the mechanical hyperalgesia as exhibited in the behavioral test.
  • Figure 6 shows exemplary intensity data for Slfn2 of L5 DRG (3d p.o.).
  • RNA samples of dorsal root ganglia (DRGs) of inbred mouse strains AKR/J (AKR), C57BL/6J (C57/B6) and CBA J (CBA) were examined.
  • the left sciatic nerve was exposed at its trifurcation and the two major branches (tibial and common peroneal nerves) were ligated and transected distal to the ligation. Sham surgery was identical, the nerves were exposed but not ligated and transected (see Bourquin et al. 2006).
  • Paw withdrawal thresholds were assessed using a dynamic plantar aesthesiometer (see Szabo et al. 2005). After acclimation in a compartment with metal mesh floor, the stimulator was positioned under the animal's hindpaw, a straight metal filament driven by an electrodynamic actuator touched the plantar surface and exerted an increasing upward force until the animal removed the paw (paw withdrawal threshold, PWT). PWTs were assessed for hindpaws of the ipsilateral, operated side and of the contralateral side. RNA isolation from dorsal root ganglia:
  • RNA extraction for TaqMan and microarray analysis a method for isolating total RNA of murine DRGs was developed (Persson et al., supra, particularly section "RNA extraction for TaqMan and microarray analysis"), wherein the method provided for RNA in a sufficient amount (> 300 ng) and quality.
  • Total RNA from DRGs was isolated with the PicoPure RNA Isolation Kit (Arcturus) following the manufacturer's instructions. RNA quality was assessed using the 2100 Bioanalyzer and RNA 6000 Nano LabChip kit (Agilent). After having extracted RNA from L5 DRGs of the three mouse strains, either Chung-operated or sham-operated control animals, the RNA probes were hybridized on Affymetrix microarrays (MOE430 2.0).
  • Samples were profiled with Affymetrix microarrays (MOE430 2.0). At least five animals of each group were tested.
  • First-strand cDNA synthesis was performed using 500ng total RNA with a 100pM T7-(dT)24 oligomer (GGCCAGTGAATTGTAATACGACTCACTATAGGGAGGCGG-dT 24 ) (SEQ ID NO: 15) according to Baugh, L.R, Hill, A.A., Brown, E.L. and Hunter, CP. (2001 ) Nucleic Acids Res. 29, e29 and Superscript II Reverse Transcriptase following the manufacturer's instructions. Double-stranded cDNA was synthesized and then extracted using phenol- chloroform followed by an ethanol precipitation step.
  • RNA Transcription Labeling kit Enzo
  • Transcription reactions were incubated at 37°C for 16h.
  • cRNA was purified using the RNeasy Mini kit (Qiagen) protocol for RNA cleanup and quantified by a spectrophotometer.
  • the biotin-labeled cRNA was fragmented using a RNA fragmentation buffer (200mM Tris-acetate, 500mM KOAc, 150mM MgOAc, pH 8.1 ).
  • Hybridization and staining of mouse MOE430_2 GeneChipsTM was performed according to the manufacturer's instructions.
  • the microarrays were scanned using a GeneChip 3000 Scanner, and the scanned data were imported and analyzed using Resolver v5.1 expression data analysis software (Rosetta Biosoftware).
  • the Affymetrix gene expression data were statistically analyzed and filtered prior to a correlation analysis.
  • the following filter criteria were used: - Abs. fold-change in at least 60 % of all Chung-operated animals > 1 .5 or
  • S1 mean a ii sham anima is with in same strain ln(ipsilateral PWT / contralateral PWT).
  • corresponding gene expression had to be detectable ("present") in at least five animals as defined by a respective intensity p-value ⁇ 0.001 .

Abstract

Slfn2 (Schlafen 2) for use as an indicator of a pain-related tissue status or a pain-related disease or as an indicator of the risk for developing a pain-related tissue status or disease, Slfn2 for use in the diagnosis or prognosis of a pain-related tissue status or of a pain-related disease A method of identifying a pain-related tissue status or the presence of a pain-related disease and/or the risk of developing a pain-related tissue status or a pain-related disease and/or the progression or a stage of a pain-related tissue status or a pain-related disease in an individual, the method comprising detecting the level of Slfn2, a kit for detecting Slfn2 comprising a means for detecting Slfn2, a data carrier comprising instructions for a method of diagnosing a pain-related tissue state or disease and optionally a container.

Description

Methods and uses relating to the diagnosis or prognosis of pain-related tissue states or pain-related diseases such as pain.
Background of the Invention Present invention relates to Slfn 2 for use in the diagnosis or prognosis of a pain-related tissue status or a pain related-disease, methods of identifying or prognosing a pain- related tissue status or disease, a kit for use in the prognosis or diagnosis of a pain- related tissue status or a pain-related disease, a kit for detecting Slfn2, and a means for detecting Slfn2 for use in the prognosis or diagnosis of a pain-related tissue status or a pain-related disease.
Physical pain is a typical sensory experience that may be described as the unpleasant awareness of a noxious stimulus or bodily harm. Individuals experience pain by various daily hurts and aches, and sometimes through more serious injuries or illnesses. For scientific and clinical purposes, pain is defined by the International Association for the Study of Pain (IASP) as "an unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage".
Pain of any type is the most common reason for physician consultation in the United States, prompting half of all Americans to seek medical care annually. It is a major symptom in many medical conditions, significantly interfering with a person's quality of life and general functioning. Diagnosis is based on characterizing pain in various ways, according to duration, intensity, type (dull, burning, throbbing or stabbing), source, or location in body. Usually pain stops without treatment or responds to simple measures such as resting or taking an analgesic, and it is then called 'acute' pain. But it may also become intractable and develop into a condition called chronic pain, in which pain is no longer considered a symptom but an illness by itself. In recent years, the study of pain has attracted many different fields such as pharmacology, neurobiology, nursing, dentistry, physiotherapy, and psychology. Pain is part of the body's defense system, triggering a reflex reaction to retract from a painful stimulus, and helps adjust behavior to increase avoidance of that particular harmful situation in the future.
Medical management of pain has given rise to a distinction between acute pain and chronic pain. Acute pain is 'normal' pain, it is felt when hurting a toe, breaking a bone, having a toothache, or walking after an extensive surgical operation. Chronic pain is a 'pain illness', it is felt day after day, month after month, and seems impossible to heal.
In general, physicians are more comfortable treating acute pain, which usually is caused by soft tissue damage, infection and/or inflammation among other causes. It is usually treated simultaneously with pharmaceuticals, commonly analgesics, or appropriate techniques for removing the cause and for controlling the pain sensation. The failure to treat acute pain properly may lead to chronic pain in some cases.
Diagnosis of pain is mostly based on symptomatic description by the patient. However, this description is naturally very subjective and hardly quantifiable, so that the choice of the correct dosage and drug mostly relates to the patients age and body weight, together with some symptomatic analyses as to the nature of the pain-related tissue state (such as inflammation, necrosis etc.), pain-related disease (such as osteoarthritis, cancer, migraine etc) or pain (such as neuropathic pain or inflammatory pain). Especially in children, animals and mentally-disabled people, a proper diagnosis of pain is mostly difficult. Objective parameters allowing for a qualitative and quantitative assessment of pain e.g. allowing for a stageing of a pain-related tissue status or a pain related disease or allowing for the assessment which medicaments to use for the treatment and in which dosage, are presently scarce if present at all. There is thus great need for objective parameters for the diagnosis and prognosis of pain. A common feature of many clinical syndromes where chronic pain develops is the existence of previous nerve damage, affecting peripheral nerves, the spinal cord or (as in stroke) the brain. The concept of neuropathic pain (i.e. pain arising as a consequence of neuronal damage) has become accepted as the underlying cause of many different chronic pain conditions seen in the clinic. Several animal models of neuropathic pain are known in the art, which mimic many aspects of the clinical condition. These include lesions of the sciatic nerve (constriction or partial section), section of spinal nerves, ischemic lesions of the spinal cord, diabetic neuropathy, etc. Such models have been subjected to detailed study of the anatomical, biochemical and physiological changes that accompany the development of the pain state.
A number of experimental models for neuropathic pain have been developed (Bennett and Xie (1988), Pain 33: 87-107; Seltzer et al. (1990), Pain 43: 205-218; Kim and Chung (1992), Pain 50: 355-363; DeLeo et al. (1994) Pain 56: 9-16 ; Na et al. (1994), Neurosci. Lett. 177: 50- 52). The availability of these different models provides an opportunity to investigate mechanisms of neuropathic pain.
Further animal models for pain are considered in an article of Walker et al. (1999), Molecular Medicine Today 5: 319-321 , comparing models for different types of pain, which are acute pain, chronic/inflammatory pain and chronic/neuropathic pain, on the basis of behavioural signs.
Present invention is based on studies of the inventors using three different inbred mouse strains differing in their pain sensitivity in a murine model system of chronic neuropathic pain (Kim SH, Chung JM. Pain. 1992 Sep; 50(3): 355-63).
One object of the present invention is thus to provide a target for diagnosis and prognosis for pain-related tissue states and pain-related diseases, especially pain. Surprisingly, the inventors found that Slfn2 is involved in pain, which, in light of the then existing knowledge of possible functions of Slfn2 that rather hinted to completely different functions (e.g. in cell cycle control, see below) was not to be expected: In a screening assay for the identification of genes involved in pain, three different inbred mouse strains differing in their pain sensitivity were examined. The expression of various genes was correlated with the pain sensitivity of the mouse strains. Among the genes showing the best correlation between pain sensitivity and expression was Slfn2 (see examples). Therefore, Slfn2 is an interesting target for the identification and profiling of compounds concerning their analgesic effects. The above overview does not necessarily describe all problems solved by present invention.
Summary of the Invention
Accordingly, in a first aspect the invention relates to Slfn2 for use as an indicator of a pain-related tissue status or a pain related disease or as an indicator of the risk for developing a pain-related tissue status or disease. A second aspect of present invention relates to Slfn2 for use in the diagnosis or prognosis of a pain-related tissue status or of a pain-related disease.
In a third aspect, present invention relates to a method of identifying
(i) a pain-related tissue status or the presence of a pain-related disease and/or
(ii) the risk of developing a pain-related tissue status or a pain-related disease and/or
(iii) the progression or a stage of a pain-related tissue status or a pain-related disease
in an individual, the method comprising detecting the level of Slfn2.
In a fourth aspect, present invention relates to a kit for use in a method of identifying a pain-related tissue status or the presence of a pain-related disease and/or the risk of developing a pain-related tissue status or a pain-related disease and/or the progression or a stage of a pain-related tissue status or a pain-related disease in an individual, the method comprising detecting the level of Slfn2 and the kit comprising one or means for detecting Slfn2.
In a fifth aspect, present invention relates to a kit for detecting Slfn2 comprising
(i) a means for detecting Slfn2,
(ii) a data carrier comprising instructions for a method a pain-related tissue status or the presence of a pain-related disease and/or the risk of developing a pain-related tissue status or a pain-related disease and/or the progression or a stage of a pain-related tissue status or a pain-related disease in an individual, the method comprising detecting the level of Slfn2 (iii) a container. In a sixth aspect, present invention relates to a means of detecting Slfn2 for use in diagnosis or prognosis of a tissue status or a disease.
In another aspect this invention relates to a method for diagnosing or prognosing a pain- related tissue status or a pain-related disease, comprising:
(a) detecting the level of Schlafen 2 (Slfn2) in the individual,
(b) comparing the level of Slfn2 in the individual to one or more reference samples,
wherein an elevated level of Slfn2 in the individual as compared to the reference samples indicates the presence, increased risk of, or progression of the pain- related tissue status or pain-related disease.
In a further aspect in said method the Slfn2 is selected from the group consisting of SEQ ID NO: 1 , 2, 6, 7, 8, and 9. In yet another aspect in said method the detecting step comprises detecting the level of Slfn2 from a sample from an individual.
In yet a further aspect in said method the sample is selected from the group consisting of: cerebrospinal fluid, neural tissue, extraneural tissue, a neural cell, and an extraneural cell.
In yet a further aspect in said method the reference sample is selected from the group consisting of: a sample from a subject not having a pain-related tissue status or a pain- related disease, a sample from a subject having a pain-related tissue status or a pain- related disease, and a sample from the individual from an earlier point in time.
In yet another aspect in said method the level of Slfn2 is either the level of Slfn2 nucleic acid or the level of Slfn2 protein. In another aspect this invention also pertains to a kit for detecting Schlafen 2 (Slfn2) comprising:
- A mends for detecting Slfn2,
- instructions for detecting Slfn2.
In yet another aspect in said kit the means for detecting Slfn2 is a means for determining the expression level of Slfn2. In yet another aspect in said kit the means for detecting is selected from the group consisting of: a means for detecting the expression level of the Slfn2 gene, a means for detecting the expression level of the Slfn2 RNA, and a means for detecting the expression level of the Slfn2 protein. In yet another aspect said kit further comprises:
(a) a container, or
(b) a data carrier, wherein the data carrier comprises information selected from the group consisting of:
(i) instructions concerning methods for identifying the risk for developing and/or identifying the presence and/or monitoring progression of a pain-related tissue status or disease,
instructions for use of the means for detecting Slfn2,
(iii) quality information , the manufacturing or assembly site or expiry or sell-by date, information concerning the correct storage handling of the kit,
(iv) information concerning the composition of the buffer(s), diluent(s), reagent(s) for detecting Slfn2 and/or of the means for detecting Slfn2,
(v) information concerning the interpretation of information obtained when performing the above-mentioned methods identifying and/or monitoring progression of a pain-related tissue status or disease,
(vi) a warning concerning possible misinterpretations or wrong results when applying unsuitable methods and/or unsuitable means, and
(vii) a warning concerning possible misinterpretations or wrong results when using unsuitable reagent(s) and/or buffer(s). In yet another aspect in said kit the means comprises one or more nucleic acids or derivatives for detecting a Slfn2 gene or gene product or functionally active variant thereof, wherein the nucleic acid or derivative is selected from the group consisting of a nucleic acid probe, a polyamide or peptide nucleic acid (PNA), microRNA (miRNA), small interfering RNA (siRNA), primers for polymerase chain reaction (PCR), primers for reverse transcription (RT) reaction, and primers for DNA sequencing.
In yet another aspect in said kit the means comprises a peptide, polypeptide, or protein for detecting a Slfn2 gene or gene product or functionally active variant thereof, wherein the protein or polypeptide is a protein ligand, an antibody, a fragment or derivate thereof, a protein scaffold, a darpin or an anticalin, or wherein the polypeptide or peptide is a probe or a mass spectrometry probe.
In yet another aspect in said kit the means comprises:
(i) a biochip, or
(ii) a set of beads.
This summary of the invention does not necessarily describe all features of the present invention. Other embodiments will become apparent from a review of the ensuing detailed description.
DEFINITIONS AND METHODS:
In the following, definitions, methods and technologies applicable in the context of present invention and its different aspects and embodiments are listed.
Before the present invention is described in detail below, it is to be understood that this invention is not limited to the particular methodology, protocols and reagents described herein as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Preferably, the terms used herein are defined as described in "A multilingual glossary of biotechnological terms: (lUPAC Recommendations)", Leuenberger, H.G.W, Nagel, B. and Kolbl, H. eds. (1995), Helvetica Chimica Acta, CH-4010 Basel, Switzerland).
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to be inclusive terms that do not exclude additional, unrecited elements or steps, such as the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integer or step.
Several documents (for example: patents, patent applications, scientific publications, manufacturer's specifications, instructions, GenBank Accession Number sequence submissions etc.) are cited throughout the text of this specification. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention. All of the documents cited herein are herein incorporated by reference. In the event of a conflict between the definitions or teachings of such incorporated references and definitions or teachings recited in the present specification, the text of the present specification takes precedence.
Sequences: All sequences referred to herein are disclosed in the attached sequence listing that, with its whole content and disclosure, is a part of this specification.
The term "about" when used in connection with a numerical value is meant to encompass numerical values within a range having a lower limit that is 5% smaller than the indicated numerical value and having an upper limit that is 5% larger than the indicated numerical value.
The term "Schlafen 2" or "Slfn2", as used herein, refers to murine Slfn2 comprising the nucleic acid sequence shown in SEQ ID NO: 1 , and the amino acid sequence shown in SEQ ID NO: 2, or to a functional variant thereof as defined below. As will be detailed below, the term "functional variant of Slfn2" encompasses naturally occurring variants, e.g. the below listed homologs, especially the human and/or rat protein and/or nucleic acid as identified below. Cloning and characterization of Slfn: Identification and cloning of the Slfn family of proteins is described by Schwarz, Katayama and Hedrick, Immunity Vol.9 (1998), p.657-668. Schlafen 2 (in the following referred to as Slfn2) is a member of the (Schlafen) Slfn gene family comprising 10 family members in mouse and 7 family members in human (Schwarz et al., 1998 infra; Berger et al., Nature Immunology Vol.1 1 , Nr.4 (2010), p.335-343). Slfn2 is sometimes also referred to as schlafen family member 12 like (see e.g. accession number NP_035538 in the NCBI database that corresponds to the murine Slfn2 protein and coding sequence). Slfn 2 has strongest homology to human SLFN12L, rat slfn 2, orang utan SLFN12L and chimpanzee SLFN12L (see below). The gene of murine Slfn 2 is located on chromosome 4 (Ferguson et al., 2005).
The reference sequence of the murine Slfn2 coding sequence is known and publicly available under the accession number: AF099973 from the NCBI (National Centre for Biotechnology Information; National Library of Medicine 38A, Bethesda, MD20894, USA; www.ncbi.nih.gov). Version AF099973.1 of the coding sequence (SEQ ID NO: 1 , positions 1 to 1684) is given in the following: 1 gctttaatgc agcaaggaac aaagaactca catgggctta gaggtgaaca cacaaccagg
61 gctgcagtta gaactcttcc aagtgccaaa gcccaccggc gaagacatct gctggccagg 121 acttgttgca gacagagggt ctttctcagg tgacactagc ctgctttacc aagactgctg 181 acatgggtac tagacttgag gcaactgagc aaagcaacca cagaactcag aggaatgaca 241 ttcagctgga aaatgcaaag gctgctggga aaatgggcat cagtgttgat ctggaagcca 301 aatatgctaa gctgggtcta aatctcggag caatcacttt tggagagaag gataggaaga
361 aaatgaagaa ttctcacctc agaaaacagg agaatgcaaa catctctcta gctgtatgtg 421 ctctcctgaa ttcgggaggt ggagcaatca aggttaaaat tgaaaatgaa aattatagtc 481 tcactaggga tggcctggga ctagatttgg aagcctctct ttgtaaatgt ctgccctttg 541 tccagtggca cctggacttc acggagagcg aaggctacat ttatatctac gtgaaatcgt 601 ggagccaaga aatctttggg ctgcctattg gcaccctaag aaccaatttg tatgtaagga
661 gcatgtcatc ttctgtacaa gtgagcgccg ctgctgccct ggaatttctc caggacctgg 721 aggaaactgg agggagaccc tgtgtcagac cagagttgcc tgcaagcata gccttccctg 781 aagtggaagg agaatggcac ctggaggatt tggctgctgc attgtttaac aggacagaat 841 ttcagtacga ggaaactttc ccctttacca gatccagata tgttgaagtt acattgcttt 901 cagcgaaacg cctgcgaaaa cgcatcaaag agctcctccc tcaaactgtt tctgcttttg
961 caaacacgga tgggggattt ttgttcattg gtttggatgg caaaacccag caaattattg 1021 gttttgaagc agagaagagc gatctcgtgc ttctagagag tgaaatagaa aagcacatcc 1081 ggcagctgcc tgtcactcac ttctgtgagg agaaggagaa gatcaagtac acgtgcaaat 1141 tcatcgaagt gcacaaatcc ggagctgtgt gtgcatatgt gtgtgcgctc agagtggaga
1201 gattctgctg tgcagtattc gctgcagagc ccgagtcctg gcatgtggaa ggcggctgtg
1261 tgaagaggtt taccacagag gaatgggtga aactccagat gaatgcccca tcaggttgaa
1321 gggggattaa taatctgtat gagggcggca gagatggctc agtggctaag agcactgact
1381 gctcttccag agatctgagc tgtaaccccc cccccccagc ctgaaacctg gttgctcagg
1441 tttcactgtt agcaagaaga gagcatggga gaggggggag gagcctgccg ccctatcgtt
1501 catcctaccc tttgtccacc cgagactaga ggggtgtgtc cgttccacgc agacaaaggg
1561 gcccagaggg tctgtggcag tggtcacctg aaactggact ccaggatgat ttggcgggaa
1621 tgggcccctt ccccctcctt cataaaattg agctttgctc agaaaaaaaa aaaaaaaaaa
1681 aaaa (SEQ ID NO:1 )
It should be noted that the coding sequences comprised herein are indicated as cDNA sequences corresponding to the respective mRNA sequences. The above coding sequence (SEQ ID NO: 1 ) encodes a 378 amino acid protein the reference sequence of which is publicly available at the NCBI database under accession number AAC83826. The translated region starts at position 183 of SEQ ID NO: 1 and ends at position 1319. Version AAC83826.1 (SEQ ID NO: 2) is given in the following:
1 mgtrleateq snhrtqrndi qlenakaagk mgisvdleak yaklglnlga
51 Itfgekdrkk mknshlrkqe nanislavca llnsgggaik vkienenysl
101 trdglgldle aslckclpf qwhldftese gyiyiyvksw sqeifglpig
151 tlrtnlyvrs msssvqvsaa aaleflqdle etggrpcvrp elpasiafpe
201 vegewhledl aaalfnrtef qyeetfpftr sryvevtlls akrlrkrike
251 llpqtvsafa ntdggfIfig ldgktqqiig feaeksdlvl leseiekhir
301 qlpvthfcee kekikytckf ievhksgavc ayvcalrver fccavfaaep
351 eswhveggcv krftteewvk lqmnapsg (SEQ ID NO:2)
The divergent AAA domain (from position 231 to position 355), that is common to Slfn- family proteins is shown underlined. Position 135, the mutation of which gives rise to the "electra" phenotype in mice (see below) is marked bold and underlined.
The promoter region of murine Slfn2 has been cloned and analysed (see Wern-Joo et al., 2007). According to Wern-Joo et al., Molecular Immunology 44 (2007), 3273-3282 (the content of which is specifically incorporated herein by reference, see especially also page 3278, e.g. figure 3 of that article), the transcription start site is located 12 nucleotides upstream of the first nucleotide according to SEQ ID NO: 1 . In the sequence according to SEQ ID NO: 3 as given below, the nucleotides according to Wern-Joo infra (capital letters) are attached to the first part of SEQ ID NO: 1 (small letters):
+1
AGTCTGTGCA ATgctttaat gcagcaagga acaaagaact cacatgggct tagaggtgaa cacacaacca gggctgcagt tagaactctt ccaagtgcca aagcccaccg gcgaagacat ctgctggcca ggacttgttg cagacagagg gtctttctca ggtgacacta gcctgcttta ccaagactgc tgacatgggt ac... [see SEQ ID NO:l]...aaaa (pos. 1684)
(SEQ ID NO:3)
The translation start (methionin/atg) is underlined and typed bold.
The promoter region of murine Slfn2 has been cloned and analysed (see Wern-Joo et al., 2007). The promoter of murine Slfn2 plus some downstream sequences according to Wern-Joo et al., 2007 is given in figure 4 (the sequence according to figure 4 includes some downstream positions incl. the translation start - Met). The upstream regulatory region, i.e. the promoter from position -1766 to +1 (+1 is underlined) is given in the following as SEQ ID NO: 4:
TTGGTGGCTCCTAGCGTTGTACAATCACATTTTGACACAAGGGGTTGGGCAATGGTGTCT CCATTGCTCTTTTGCTCTGACTTAGGCAGGGCGGGGTGGGGGTGGGGGTTGGTAGGTGGG GCAGTTGTCAGTCATAGGACAGGAGAAAGTATTGGTGATAAATTGGTGATTAGGCCTTAT CTGGGCCACCTGGCTTGAAGGTCTTTGACTCCAAGCCCTGGTGGTCAGGCCTTACCCTGG ATATTTGACCTGTAACTCAAGATGAGAAGGCAACAAAGGAGCTAGTTCCTGGGAACATAG ACTCAGCTGGACCCTGCCAGCCGATGGAGAACTTGTCCCCGAGGTGGCTGGACTCGGAGA GCTGTCTCCTTATAAATGCTCTCCTGGCTGCCTCCTTCCTTAAGCCTGGGCACTGCACTT TGTCCTTGATGGAGAAGTCCCCAGAGTTTGCTGCAGTTCTAATCCCAGACCTTTGGAGTT GAATTCCTTCTCACTGACTTTGGAACTGAACTGGAGCTCGGAGGAGGAGCTCGGAGGAGG AGCTCGGAGGAGGAGGAGGAGGAGGAGGAGGAGGGAGGAGGGAGGAGAGTGAAGAGGGGA GAGAAAGAAGAGAAGACTTCAGGCACACAGAGCATGGGCACTGGACTTTCTCCACCCAGC AGCTGCCTCTGTCCAGATGCTGCTCCTCCCTGGCTCCCCCACTTTCCCTACCCCTGTTCT TTTCTTCCTCATGTCCCTCAAAGCCCTGGTTGTTGGCTTGCCCTGGTCTCTGGCTAGTCT TGTGACATTCCTTAGGAACGGGAGGGTACAGTTCTGAGCAAGAGGTTGTGCTTGGGATAC TGGAGGAGGGTGACTCATTGGACTCCCCCTAGCAACTTCTTCTTTCACCTCCTGACAAGT ACAGAAGGAAAATTTTGTCTTTCTTCATTTTTGGTCTTCTTCATTACTGATGCAAATATT CTCCTTTACGTTTTTGCATGTTTCCATCAACCCACGAGAACTCCAGAAACCTCCTGTACT TTATTTCTTAAATTAGAGATTAAGAAAGAGATTCTAAAGGTCTCTGAGTCAGTTTCCTCA GAGCTGGTCTGGTGGTGACAGGCAATGAAACAGCCCACAGGGAAGAAAGAGCCCTGTTCT GGACCCTTAGGCTGGAGGGACAGTGGAGTTCTGGACCTCTGGAAGGGCAGTCAGTGCTCT TATCCACTGAACCATCTCTCCAGCCCTATTTCTTTTCTTTTCTTATCTTTTCTTTTTTAA TATTTATTTTATTGGATATTTTCTTTATTTACATTTCAAATGTTATCCCCTTTCCTGGTC CTGGCCCCGGTCCTTCCTTCCTCCCCCAACTCCCTCTCCCATCCACCCCCAACTCCCTTT CCCTTCCCTCAAGCCCCCTCTCCCACCCCACCCCCCCTCCCCAATCAGTTTCTTAAGGCT GGTTAGGGAGGTGTTACTGTAACAAAGTAGCAAAAGAGGTGTACTGCTTTAGGGTAGCCC TGGGAATCCCATAGAGAGAAGGAATCTGCTGGCAGAGAGGATCCTGCTGTGCCCATCTTA AACCCGGGACTGGAGGGACTCTAAAGGCGTTAACAAGGGCACCTGCTCTTTTGAGCAAAT GGACTTTGCTGCTTGGGAATGCCTGAGGCAGATAGAAATCTGACTAGTCAACTGTGAGCT TGTGTTACAGTCTGCCCAATAGAAAAGTGGAGGCAGGGCTTTCACTTCCTGGAGGGTGAC AGTTCTGGGATTTCCAGTCTTCCACCA
(SEQ ID NO: 4)
The Slfn2 sequence containing the upstream regulatory sequence according to Wern- Joo et al., 2007 (capital letters) and the complete coding sequence according to SEQ ID NO: 1 (small letters) comprises positions -1766 to +1696 and is given in the following:
TTGGTGGCTCCTAGCGTTGTACAATCACATTTTGACACAAGGGGTTGGGCAATGGTGTCT CCATTGCTCTTTTGCTCTGACTTAGGCAGGGCGGGGTGGGGGTGGGGGTTGGTAGGTGGG GCAGTTGTCAGTCATAGGACAGGAGAAAGTATTGGTGATAAATTGGTGATTAGGCCTTAT CTGGGCCACCTGGCTTGAAGGTCTTTGACTCCAAGCCCTGGTGGTCAGGCCTTACCCTGG ATATTTGACCTGTAACTCAAGATGAGAAGGCAACAAAGGAGCTAGTTCCTGGGAACATAG ACTCAGCTGGACCCTGCCAGCCGATGGAGAACTTGTCCCCGAGGTGGCTGGACTCGGAGA GCTGTCTCCTTATAAATGCTCTCCTGGCTGCCTCCTTCCTTAAGCCTGGGCACTGCACTT TGTCCTTGATGGAGAAGTCCCCAGAGTTTGCTGCAGTTCTAATCCCAGACCTTTGGAGTT GAATTCCTTCTCACTGACTTTGGAACTGAACTGGAGCTCGGAGGAGGAGCTCGGAGGAGG AGCTCGGAGGAGGAGGAGGAGGAGGAGGAGGAGGGAGGAGGGAGGAGAGTGAAGAGGGGA GAGAAAGAAGAGAAGACTTCAGGCACACAGAGCATGGGCACTGGACTTTCTCCACCCAGC AGCTGCCTCTGTCCAGATGCTGCTCCTCCCTGGCTCCCCCACTTTCCCTACCCCTGTTCT TTTCTTCCTCATGTCCCTCAAAGCCCTGGTTGTTGGCTTGCCCTGGTCTCTGGCTAGTCT TGTGACATTCCTTAGGAACGGGAGGGTACAGTTCTGAGCAAGAGGTTGTGCTTGGGATAC TGGAGGAGGGTGACTCATTGGACTCCCCCTAGCAACTTCTTCTTTCACCTCCTGACAAGT ACAGAAGGAAAATTTTGTCTTTCTTCATTTTTGGTCTTCTTCATTACTGATGCAAATATT CTCCTTTACGTTTTTGCATGTTTCCATCAACCCACGAGAACTCCAGAAACCTCCTGTACT TTATTTCTTAAATTAGAGATTAAGAAAGAGATTCTAAAGGTCTCTGAGTCAGTTTCCTCA GAGCTGGTCTGGTGGTGACAGGCAATGAAACAGCCCACAGGGAAGAAAGAGCCCTGTTCT GGACCCTTAGGCTGGAGGGACAGTGGAGTTCTGGACCTCTGGAAGGGCAGTCAGTGCTCT TATCCACTGAACCATCTCTCCAGCCCTATTTCTTTTCTTTTCTTATCTTTTCTTTTTTAA TATTTATTTTATTGGATATTTTCTTTATTTACATTTCAAATGTTATCCCCTTTCCTGGTC CTGGCCCCGGTCCTTCCTTCCTCCCCCAACTCCCTCTCCCATCCACCCCCAACTCCCTTT CCCTTCCCTCAAGCCCCCTCTCCCACCCCACCCCCCCTCCCCAATCAGTTTCTTAAGGCT GGTTAGGGAGGTGTTACTGTAACAAAGTAGCAAAAGAGGTGTACTGCTTTAGGGTAGCCC TGGGAATCCCATAGAGAGAAGGAATCTGCTGGCAGAGAGGATCCTGCTGTGCCCATCTTA AACCCGGGACTGGAGGGACTCTAAAGGCGTTAACAAGGGCACCTGCTCTTTTGAGCAAAT GGACTTTGCTGCTTGGGAATGCCTGAGGCAGATAGAAATCTGACTAGTCAACTGTGAGCT TGTGTTACAGTCTGCCCAATAGAAAAGTGGAGGCAGGGCTTTCACTTCCTGGAGGGTGAC AGTTCTGGGATTTCCAGTCTTCCACCAGTCTGTGCAATgctttaatgcagcaaggaacaa agaactcacatgggcttagaggtgaacacacaaccagggctgcagttagaactcttccaa gtgccaaagcccaccggcgaagacatctgctggccaggacttgttgcagacagagggtct ttctcagtgacactagcctgctttaccaagactgctgacatgggtactagacttgaggca actgagcaaagcaaccacagaactcagaggaatgacattcagctggaaaatgcaaaggct gctgggaaaatgggcatcagtgttgatctggaagccaaatatgctaagctgggtctaaat ctcggagcaatcacttttggagagaaggataggaagaaaatgaagaattctcacctcaga aaacaggagaatgcaaacatctctctagctgtatgtgctctcctgaattcgggaggtgga gcaatcaaggttaaaattgaaaatgaaaattatagtctcactagggatggcctgggacta gatttggaagcctctctttgtaaatgtctgccctttgtccagtggcacctggacttcacg gagagcgaaggctacatttatatctacgtgaaatcgtggagccaagaaatctttgggctg cctattggcaccctaagaaccaatttgtatgtaaggagcatgtcatcttctgtacaagtg agcgccgctgctgccctggaatttctccaggacctggaggaaactggagggagaccctgt gtcagaccagagttgcctgcaagcatagccttccctgaagtggaaggagaatggcacctg gaggatttggctgctgcattgtttaacaggacagaatttcagtacgaggaaactttcccc tttaccagatccagatatgttgaagttacattgctttcagcgaaacgcctgcgaaaacgc atcaaagagctcctccctcaaactgtttctgcttttgcaaacacggatgggggatttttg ttcattggtttggatggcaaaacccagcaaattattggttttgaagcagagaagagcgat ctcgtgcttctagagagtgaaatagaaaagcacatccggcagctgcctgtcactcacttc tgtgaggagaaggagaagatcaagtacacgtgcaaattcatcgaagtgcacaaatccgga gctgtgtgtgcatatgtgtgtgcgctcagagtggagagattctgctgtgcagtattcgct gcagagcccgagtcctggcatgtggaaggcggctgtgtgaagaggtttaccacagaggaa tgggtgaaactccagatgaatgccccatcaggttgaagggggattaataatctgtatgag ggcggcagagatggctcagtggctaagagcactgactgctcttccagagatctgagctgt aaccccccccccccagcctgaaacctggttgctcaggtttcactgttagcaagaagagag catgggagaggggggaggagcctgccgccctatcgttcatcctaccctttgtccacccga gactagaggggtgtgtccgttccacgcagacaaaggggcccagagggtctgtggcagtgg tcacctgaaactggactccaggatgatttggcgggaatgggccccttccccctccttcat aaaattgagctttgctcagaaaaaaaaaaaaaaaaaaaaaa
(SEQ ID NO: 5)
The upstream sequence comprises two AP-1 sites (-917 to -908 and -703 to -695) and one NF-kappa B binding site (-21 to -12). According to Wern-Joo et al., 2007 infra both AP-1 sites are important for maximal promoter activity upon LPS or CpG-DNA
stimulation (8-9 fold induction), but activity is also exhibited by a fragment comprising only the second AP-1 site (-703 to -695) and the NF kappa B site (2 fold induction). Possible promoter fragments able to drive expression of downstream coding regions and the construction of fragments, point mutants and various luciferase constructs are disclosed in Wern-Joo et al., 2007 infra. Examples of Slfn-2 promoter fragments usable for cloning of reporter-gene constructs are nucleic acids comprising or having the following positions according to SEQ ID NO:5: from -1766/-1265 to +1/+50/
+100/+150/+198.
Slfn2 has been reported to be implicated in the regulation of quiescence in T-cells and monocytes (Berger et al., 2010 infra) and in interferon alpha-induced growth arrest in fibroblasts (Katsoulidis et al., Journal of Biological Chemistry Vol.284, Nr. 37 (2009)). A point mutation on position 135 of the 278 amino acid protein caused by a thymidine-to- adenosine transversion in the Slfn2 gene and leading to an isoleucine to asparagine substitution leads to a loss-of-f unction mutation causing lymphoid and myeloid immunodeficiency due to loss of quiescence of T-cells and monocytes in mice, and causes the "electra" phenotype, leaving natural killer cells or B cells unaffected (Berger et al., 2010). Markedly NFAT and NF-kB and Erk and Akt appear to be normally activated in electra mice, the mitogen-activated protein kinases p38 and Jnk are constitutively phosphorylated. Moreover, expression levels of the anti-apoptotic Bcl-2 is reported to be lower in T-cells derived from electra mice, wherein transgenic expression of Bcl-2 rescues CD4+ and CD8+ T-cells derived from electra mice from apoptotic cell death (all Berger et al., 2010 infra, see also Horton and Powell, Nature Immunology Vol. 1 1 , Nr.4 (2010), p.281 -282). Thus, the reported mutation of Slfn2 appears to affect T- cell antigen receptor-induced signaling pathways, wherein other pathways are not affected. As can be gained from sequence comparison of murine, rat and human proteins (see Figure 1 ) using ClustalW, the isoleucine at position 135 of murine Slfn2 according to SEQ ID NO:2 (and corresponding to position 135 in rat Slfn2 according to SEQ ID NO:7 and position 123 in hsSLFN12L according to SEQ ID NO:9) is conserved among murine Slfn2, rat Slfn2 and homo sapiens SLFN12L hinting to an equal importance of said amino acid or surrounding protein region within the respective rat and homo sapiens proteins.
The following species variants are exemplified:
Murine protein NCBI accession Symbol Chromosome
Organism
Identity* (%) numbers Location Mus 100 % AAC83826.1 SLFN2 1 1 (50.3 cM) musculus AF099973.1 82878614- 82884180(+)
NCBI Mouse Build 37
Rattus 83% NM_001 107031 .1 SLFN2 10(126) norvegicus NP_001 100501 .1
XP_220779.4
Human 50% NP_001 182719.1 SLFN12L 17 (q12)
NM_001 195790.1
Percent identity has been determined using the EMBL ClustalW program. The rat homolog of Slfn2 is schlafen family member 12-like (Slfn12L). The protein consists of 381 amino acids and the protein sequence is available under accession number NP_001 100501 .1 (SEQ ID NO: 7) and is encoded by the coding sequence NM_001 107031 .1 (from positions 193-1338).
The coding sequence of rat Slfn2 according to NM_001 107031 .1 is given following (SEQ ID NO: 6):
1 cttcaatgca gcaaagaaca gagaactcaa gtgagcttag aggtgagctc acagccaggg 61 ctgcagatag aactctccca agtgccaaca accagcctcg aagacgtccc cggccaggac 121 ttgttgcaga caacactagt tgtctttctc aagtgacact agcctgcctc accaaaggag
181 ctgactgctg gcatgggcgc tggacttcat gcagctgagc aagacaacca cagagctcac 241 aggaagggca ctgggcagca aaatgcaaaa gctgctggga aaatgggcat cagcgttgac 301 ctggaagctg agtatgctaa gctgggtcta aatctaggag caatcactct tggagagaag 361 gataggagga aaatgaagaa ctctcacctc aggaaaaagg agaatgagaa catctctctg 421 gctgtgtgtg ctctcctgaa ttcaggaggt ggagcaatca aggttaaaat tgaaaatgaa 481 aattatagtc tcactaggga tggcctggga ctagatttgg aaacctctct ttgtaaatgt
541 ctcccgtttg tccagtggta cctggacttc actgagagca aaggctacat ttatatctac
601 gtgagagcat ggagtcagga aatctttggg ctgccgattg gcaccctgag caccaatttg
661 tacgtaaggg tcgtttcatc ctccgtacaa gtgagcgctg atgctgcgct acaatttctc
721 caggacctgg aggaagctgg agggagaccc cgtgtcagac cagagttgcc tgcaaggaaa
781 gcctgccctg gagtggaagg agaatgtcac caggaggatt tggctgctgc attttttaac
841 aagaccgaat ttcagtatga ggaaactttc ttctttacca ggtccaggta tgttgaagtt
901 acatcacttt cagtgaaacg cctgcgaaaa cgcatcagag agatcctccc tcgaactgtt
961 tctgcattcg caaacacgga tgggggatat ttgttcattg gcttggatgg gaaaaaacag
1021 caaataattg gctttgaagc agagaagagc gaccttgtgc atctagagag tgaaatagaa
1081 aagtgcatcc gacagctgcc tgtcactcac ttctgtgagg agagggagaa gatcaagtac
1141 acgtgcaaat tcatggaagt gcacaaaccc ggagccgcgt gttcatacgt gtgtgcgctc
1201 cgagtggaga gattctgctg tgcagtgttc gctgcagagc ccgaatcctg gcacgtggaa
1261 gacagctgtg tgaagaggtt tgccgcagag gactgggtta ggctccagat gggttccagg
1321 gaagcttgga aaaggtgact aataatcagt tagtgaaaaa taaagatttt taagttcccc
1381 acttgaccct gctgtgagga caagtgcacc tcccaaaaga atagcatcac atgttttggt
1441 gaggtagcct ggctatagcc accacaaacc atctggcctc gataaggcct gacacaacag
1501 ttctgaggaa aacaagaact agggtcagct tgccccagaa gcagggtggc ctagagttga
1561 actgaggcca tgtagttttc cctataaatc ccttccccta attgggctcg gggttgactc
1621 ctctgtctcc tgtgtgagat acgtgtcatc cccagagctc tggcttctca aatatactta
1681 ttatatcaag accggtttct cgtgagttct tgggggttgc gtcatcttga gatttgagtg
1741 ggggtcttcc ccaccttggt ggtctttcat tacaaccatc tgtaatgata tctgatgccc
1801 tcttctaact attttccagc atccacatgg tgattcacaa ccacctagaa ccccagttta
1861 tttatttttt ttggctgttt tttttttttt tttggtttta catggagtgg tttaatgaga
1921 gaagaagggt agaagaggag aggaacaaag gaggctggcc atgggcacgt ggggggaagg
1981 ggggatgggg agagaaggga cagggacaaa gaggacacca gaagaggaga gcaagagaga
2041 ggagtaagag caaactgatg agcttccagg gactaagc
(SEQ ID NO:6)
The protein sequence of rat Slfn2 according to NP_001 100501 .1 (SEQ ID NO:7) given in the following:
1 mgaglhaaeq dnhrahrkgt gqqnakaagk mgisvdleae yaklglnlga itlgekdrrk
61 mknshlrkke nenislavca llnsgggaik vkienenysl trdglgldle tslckclpfv
121 qwyldftesk gyiyiyvraw sqeifglpig tlstnlyvrv vsssvqvsad aalqflqdle
181 eaggrprvrp elparkacpg vegechqedl aaaffnktef qyeetffftr sryvevtsls
241 vkrlrkrire ilprtvsafa ntdggylfig ldgkkqqiig feaeksdlvh leseiekcir
301 qlpvthfcee rekikytckf mevhkpgaac syvcalrver fccavfaaep eswhvedscv 361 krfaaedwvr lqmgsreawk r
(SEQ ID NO:7) The divergent AAA domain spans positions 231 to 361 of the rat Slfn2 protein.
The Human homolog of Slfn2 is schlafen family member 12-like (SLFN12L). The coding sequence can be retrieved from the NCBI database under accession number
NM_001 195790.1 (SEQ ID NO: 8), coding for a 588 amino acid protein with sequence according to NCBI accession number NP_001 182719.1 (SEQ ID NO:9) .
Human SLFN12L coding sequence according to NM_001 195790.1 (SEQ ID NO:8):
1 atggacctcg ccagaaaaga atttctgcgt ggaaatggct tagctgctgg gaaaatgaac
61 atcagtattg atttagacac aaactatgct gagctggttc taaatgtggg aagagtcact
121 cttggagaga acaatagaaa aaaaatgaag gattgtcaac tgagaaaaca gcagaatgaa
181 aatgtctcac gagctgtgtg tgctctgctg aattctggag ggggagtgat caaggctgaa
241 gttgagaata aaggctatag ttataaaaaa gatggaatag ggctagattt ggaaaattct
301 tttagtaaca tgctgccatt tgttcctaat ttcctggact tcatgcagaa tggtaactac
361 tttcacattt ttgtgaaatc atggagcttg gaaacctctg gtccgcagat tgccacgttg
421 agctccagtt tgtacaagag agatgtaacg tctgcaaaag tcatgaatgc ttctgctgca
481 ctggagttcc tcaaagacat ggaaaaaact ggagggagag catatttaag accagaattc
541 cctgcaaaaa gggcctgtgt tgatgtacaa gaagaaagta acatggaagc cttggctgct
601 gattttttta acagaacaga acttggttat aaagaaaaat tgacctttac tgaatccaca
661 cacgttgaaa taaaaaactt ctcgactgaa aagttgttac aacgaattac agagattctc
721 cctcaatatg tttctgcatt tgcaaatact gatggaggat atttattcgt tggtctaaat
781 gaagataaag aagtaattgg ctttaaagca gagaagagtt atcttactaa gttagaagaa
841 gtaacaaaaa attccattgg gaaactgcct gtgcatcact tctgtgtgga gaaggggacg
901 ataaattact tatgcaaatt ccttggagta tatgataaag gaaggctttg tggatatgtg
961 tatgcactca gagtggaacg cttctgctgt gcagtgtttg ctaaaaagcc tgattcctgg
1021 cacgtgaaag ataacagagt taagcagttg accgagaagg aatggatcca gttcatggtg
1081 gattcagaac cagtatgtga ggaactgccc tctccagcaa gtacatcatc acctgtctcc
1141 cagagttatc ctcttcgtga atatattaac ttcaaaattc agccactgag atatcacctt
1201 ccagggctat cagaaaagat aacttgtgct ccaaaaacct tctgcagaaa tctgttctca
1261 caacatgaag gacttaagca attaatatgt gaagaaatgg gctctgtcaa taagggctca
1321 ctgatcttct ctaggagctg gtctttggat ctgggcttgc aagagaacca caaagtcctc
1381 tgtgatgctc ttctgatttc ccaggacaag cctccagtcc tatacacctt ccacatggta
1441 caggatgagg agtttaaaga ctattctaca caaactgccc aaactttaaa acagaagctg
1501 gcaaaaattg gtggttacac taaaaaagtg tgtgtcatga caaagatctt ctacttgagc
1561 cctgaaggca agacaagctg ccagtatgat ttaaactcgc aagtaattta ccctgaatcc
1621 tactattgga caacagctca aacaatgaaa gacttggaaa aggccctttc aaatatctta
1681 cctaaggaga atcaaatctt tttgtttgtt tgtttgtttc gtttttgttt gtttgtttgt
1741 tggtttgttt gttttttctt gagatga
(SEQ ID NO:8) Human SLFN12L protein sequence according to NP_001 182719.1 (SEQ ID NO:9):
1 mdlarkeflr gnglaagkmn isidldtnya elvlnvgrvt lgennrkkmk dcqlrkqqne 61 nvsravcall nsgggvikae venkgysykk dgigldlens fsnmlpfvpn fldfmqngny 121 fhifvkswsl etsgpqiatl ssslykrdvt sakvmnasaa leflkdmekt ggraylrpef
181 pakracvdvq eesnmealaa dffnrtelgy kekltftest hveiknfste kllqriteil 241 pqyvsafant dggylfvgln edkevigfka eksyltklee vtknsigklp vhhfcvekgt 301 inylckflgv ydkgrlcgyv yalrverfcc avfakkpdsw hvkdnrvkql tekewiqfmv 361 dsepvceelp spastsspvs qsyplreyin fkiqplryhl pglsekitca pktfcrnlfs 421 qheglkqlic eemgsvnkgs lifsrswsld lglqenhkvl cdallisqdk ppvlytfhmv
481 qdeefkdyst qtaqtlkqkl akiggytkkv cvmtkifyls pegktscqyd lnsqviypes 541 yywttaqtmk dlekalsnil pkenqiflfv clfrfclfvc wfvcfflr
(SEQ ID NO:9) The divergent AAA domain spans positions 218 to 349 of human SLFN12L.
Further SLFN2 homologs (SLFN12L) are known in Orang Utan (Pongo abelii, see Gene ID: 100190853 of NCBI database) and Chimpanzee (Pan troglodytes, see Gene ID: 454585 of NCBI database).
The term Slfn2 protein as used herein refers e.g to a protein having or comprising
1 . an amino acid sequence according to SEQ ID NO: 2, 7 or 9,
2. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
3. an amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
4. an amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to SEQ ID NO:2, and/or
5. an amino acid sequence with at least 50% identity with the amino acid sequence according to SEQ ID NO:2 and/or 6. an amino acid sequence with one of the following sequence motifs: YxxlxV, YxxlxVxxW, YxxlxVxxWS or YxxlxVxxWSxS with "x" standing for any amino acid and/or
7. carries a variant AAA domain, preferably with 50, 60, 70, 80, 90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2.
This means that the Slfn2 protein or the functionally active variant can comprise, consist of or have an amino acid sequence with the following features: An amino acid sequence having (1 ), (2), (3), (4), (5), (6) and/or (7). This means that the Slfn2 protein at least has (1 ) or (2) or (3) or (4) or (5) or (6) or (7); the protein can also have (1 ) and (2) or (1 ) and (3) or (1 ) and (4) or (1 ) and (5) or (1 ) and (6) or (1 ) and (7) or (2) and (3) or (2) and (6) or (2) and (5) or (2) and (6) or (2) and (7) or (3) and (4) or (3) and (5) or (3) and (6) or (3) and (7) or (6) and (5) or (4) and (6) or (4) and (7) or (5) and (6) or (5) and (7) or (6) and (7); the data carrier can also comprise (1 ) and (2) and (3) or (1 ) and (2) and (4) or (1 ) and (2) and (5) or (1 ) and (2) and (6) or (1 ) and (2) and (7) or (1 ) and (3) and (4) or (1 ) and (3) and (5) or (1 ) and (3) and (6) or (1 ) and (3) and (7) or (1 ) and (4) and (5) or (1 ) and (4) and (6) or (1 ) and (4) and (7) or (1 ) and (5) and (6) or (1 ) and (5) and (7) or
(1 ) and (6) and (7) or (2) and (3) and (4) or (2) and (3) and (5) or (2) and (3) and (6) or (2) and (3) and (7) or (2) and (4) and (5) or (2) and (4) and (6) or (2) and (4) and (7) or
(2) and (5) and (6) or (2) and (5) and (7) or or (2) and (6) and (7) or (3) and (4) and (5) or (3) and (4) and (6) or (3) and (4) and (7) or (4) and (5) and (6) or (4) and (5) and (7) or (5) and (6) and (7); the data carrier can also comprise (1 ) and (2) and (3) and (4) or (1 ) and (2) and (3) and (5) or (1 ) and (2) and (3) and (6) or (1 ) and (2) and (3) and (7) or (2) and (3) and (4) and (5) or (2) and (3) and (4) and (6) or (2) and (3) and (4) and (7) or
(3) and (4) and (5) and (6) or (3) and (4) and (5) and (7) or (4) and (5) and (6) and (7); the data carrier can also comprise (1 ) and (2) and (3) and (4) and (5) or (1 ) and (2) and (3) and (4) and (6) or (1 ) and (2) and (3) and (4) and (7) or (2) and (3) and (4) and (5) and (6) or (2) and (3) and (4) and (5) and (7) or (3) and (4) and (5) and (6) and (7).
The term "pain", as used in the context of the present invention, refers to a complex subjective sensation reflecting real or potential tissue damage and the affective response to it. The term "pain" encompasses acute pain, subacute pain and chronic pain. It further encompasses disease-associated pain, algesia and migraine. Acute pain is a physiological signal indicating a possible or actual injury. Acute pain starts abruptly and is generally sharp in quality. It serves as a warning of disease or a threat to the body. Acute pain might be mild and last just a moment, or it might be severe and last for weeks. In most cases, acute pain disappears when the underlying cause of pain has been treated or healed. Acute pain might be somatogenetic (organic). Organic pain might be any pain resulting from a disorder, abnormality or chemical imbalance in an organ system, for example, the human or animal body. Thus, organic pain is a term covering pain causes that range in diversity from heartburn to multiples sclerosis. Almost any system or body organ can be affected by organic pain including, but not limited to, the circulatory system, musculoskeletal system, or neurological system. Further examples of organic pain are constipation, diarrhoea, gastrointestinal tract infections, enteritis, colitis, or urinary tract infections. Unrelieved acute pain, however, might lead to chronic pain. Subacute pain can be classified as a status between acute pain and chronic pain.
Chronic pain can either be somatogenetic (organic) (see above) or psychogenic (psychosomatic). Chronic pain is frequently accompanied or followed by vegetative signs, which often result in depression.
Traditionally, the discrimination between acute and chronic pain has relied upon an arbitrary interval of time from onset, the two most commonly used markers being 3 months and 6 months from the initiation of pain (Turk, D.C., Okifuji, A. (2001 ), "Pain terms and taxonomies". In Loeser, D., Butler, S. H., Chapman, J.J. et al. Bonica's management of pain (3 ed.), Lippincott Williams & Wilkins, pages 18 to 25). Some researchers have placed the transition from acute to chronic pain at 12 months (Main, C.J., Spanswick, C.C. (2001 ), Pain management: an interdisciplinary approach, Elsevier, page 93). For other researchers acute pain is pain that lasts less than 30 days, chronic pain is pain of more than six months duration, and subacute pain is pain that lasts from one to six months (Thienhaus, O., Cole, B.E. (2002), "Classification of pain", In Weiner, R.S. Pain management: A practical guide for clinicians (6 ed.), American Academy of Pain Management). In the context of the present invention, acute pain may refer to pain that lasts less than 30 days (about 1 month), chronic pain may refer to pain that lasts more than six months, and subacute pain may refer to pain that lasts from one to six months. Alternatively, in the context of the present invention, acute pain may refer to pain that does not extend beyond the expected period of healing and chronic pain may refer to pain that extends beyond the expected period of healing.
Further, the term "pain", as used herein, means pain of nociceptive, inflammatory or neuropathic origin.
Nociceptive pain is part of a rapid warning relay instructing the motor neurons of the central nervous system to minimize detected physical harm. Nociceptive pain is caused by activation of nociceptors. These nociceptors are free nerve endings that terminate just below the skin, in tendons, joints, and in body organs. Nociceptive pain may be divided into "superficial somatic" pain and "deep" pain. "Deep" pain, in turn, may be divided into "deep somatic" pain and "visceral" pain. "Superficial somatic pain" is initiated by activation of nociceptors in the skin or superficial tissues, while "deep somatic" pain is initiated by stimulation of nociceptors in ligaments, tendons, bones, blood vessels, fasciae and muscles, and is dull, aching, poorly-localized pain. "Visceral" pain originates in the viscera (organs). "Visceral" pain may be well-localized, but often it is extremely difficult to locate, and several visceral regions produce "referred" pain when injured, where the sensation is located in an area distant from the site of pathology or injury. Examples of nociceptive pain include, but are not limited to, sprains, bone fractures, burns or obstructions. Further, nociceptive pain can be associated with nerve damage caused by trauma, diseases such as diabetes, shingles, irritable bowel syndrome, late-stage cancer or the toxic effects of chemotherapy. Nociceptive pain is generally time-limited. This means that when the tissue damage heals, the pain typically resolves. Thus, nociceptive pain is usually acute pain. Another characteristic of nociceptive pain is that it tends to respond well to treatment with opioids.
Neuropathic pain or neuralgia (both terms are used synonymously herein) can be defined as non-nociceptive pain or as pain that is not related to activation of pain receptor cells in any part of the body. Neuropathic pain or neuralgia is rather caused by damage to or malfunction of the nervous system. Unlike nociceptive pain, neuropathic pain exists with no continuous nociceptive input. Neuralgia or neuropathic pain can be defined as non-nociceptive pain, or in other words, pain that is not related to activation of pain receptor cells in any part of the body. It is believed that neuralgia is pain produced by a change in neurological structure or function. Unlike nociceptive pain, neuralgia exists with no continuous nociceptive input. Neuralgia falls into two categories: central neuralgia and peripheral neuralgia. This unusual pain is thought to be linked to four possible mechanisms: ion gate malfunctions; the nerve becomes mechanically sensitive and creates an ectopic signal; cross signals between large and small fibers; and malfunction due to damage in the central processor. Neuralgia falls into two categories: central neuropathic pain or neuralgia (originating in the brain or spinal cord) and peripheral neuropathic pain or neuralgia (originating in the peripheral nervous system). Peripheral neuropathic pain can be described as burning, tingling, stabbing or as pins and needles. Bumping the "funny bone", for example, elicits peripheral neuropathic pain. Neuropathic pain is often difficult to diagnose, and most treatments show little or no effectiveness. Diagnosis typically involves locating the damaged nerve by identifying missing sensory or motor function. This may involve tests such as an EMG test or a nerve conduction test. Neuralgia is more difficult to treat than other types of pain because it does not respond well to normal pain medications. Examples of neuropathic pain include, but are not limited to, reflex sympathetic dystrophy, nerve trauma, components of cancer pain, phantom limb pain, entrapment neuropathy (e.g. carpal tunnel syndrome) and peripheral neuropathy (widespread nerve damage), bony hyperostosis casts: crutches, prolonged cramped postures; haemorrhage into a nerve; exposure to cold or radiation; collagen-vascular disorders; infectious diseases such as Lyme disease and HIV; toxins such as emetine, hexobarbital, barbital, chlorobutanol, sulfonamides, phenytoin, nitrofurantoin, the vinca alkaloids, heavy metals, carbon monoxide, triorthocresylphosphate, orthodinitrophenol, and other solvents and industrial poisons; autoimmune reactions; nutritional deficiency, and vitamin B deficiency in particular; and metabolic disorders such as hypothyroidism, porphyria, sarcoidosis, amyloidosis, uremia and diabetes (see e.g. The Merck Manual, 16th ed. 1518 (1992). Peripheral neuropathy may have several causes, for example, diabetes, chronic alcohol use, exposure to toxins (applied in chemotherapies), or vitamin deficiencies. Neuropathic pain is frequently chronic. It tends to have a less robust response to treatment with opioids. However, it may respond well to other drugs such as anti-seizure and anti-depressant drugs.
The pain may also be a mixed category pain. In some conditions the pain appears to be caused by a complex mixture of nociceptive pain and neuropathic pain. For example, an initial nervous system dysfunction or injury may trigger the neural release of inflammatory mediators and subsequent neurogenic inflammation. Migraine headaches, for example, probably represent a mixture of nociceptive pain and neuropathic pain. Inflammatory pain is associated with tissue damage and the resulting inflammatory process. Chronic pain may involve a mix of both inflammatory and neuropathic components, whereas inflammation may cause damage to the neurons and produce neuropathic pain or neuronal injury may cause an inflammatory reaction that contributes to the inflammatory pain.
Algesia, from the Greek word algesis, is the sensitivity to pain. Hyperalgesia is an increased sensitivity to pain, which may be caused by damage to nociceptors or peripheral nerves. Hyperalgesia can be experienced in focal, discrete areas, or as a more diffuse, body-wide form. Conditioning studies have established that it is possible to experience a learned hyperalgesia of the latter, diffuse form. The focal form is typically associated with injury, and is divided into two subtypes: (i) primary hyperalgesia which describes pain sensitivity that occurs directly in the damaged tissues or (ii) secondary hyperalgesia which describes pain sensitivity that occurs in surrounding undamaged tissues.
For an overview of pain mechanisms, it is referred to Scholz and Woolf, 2002; Julius and Basbaum, 2001 , Woolf and Mannion, 1999; Wood, J.D., 2000; Woolf and Salter, 2000. Many diseases and diseased states are associated with pain (disease-associated pain or diseased state-associated pain). These pain types may also fall under the above pain classifications. Examples of disease-associated pain include, but are not limited to, pain associated with arthritis, back pain, cancer, epilepsy, lumbago, sciatica, lumbar spinal stenosis, cervical spinal stenosis, (clinical) depression, Fibromyalgia, Chronic Fatigue Syndrome, Complex Regional Pain Syndrome, Irritable Bowel Syndrome, Myofascial Pain Syndrome, Post-Vasectomy Pain Syndrome or Restless Leg Syndrome. Disease- associated pain may also be pain associated with diabetes, e.g. diabetic neuropathy or the diabetic foot, pain associated with Parkinson's Disease, pain associated with viral infections, e.g. influenza or HIV, or bacterial infections, pain associated with fever, pain associated with levodopa, e.g. in Parkinson's disease, pain associated with coronary heart disease, pain associated with shingles, pain associated with headache, pain associated with migraine, pain associated with muscular tension, especially chronic muscular tension, osteoarthritis as well as the pain-associated diseases listed under causes of neuropathic pain above.
Migraine (from the Greek words hemi, meaning half, and kranion, meaning skull) is a debilitating condition characterized by moderate to severe headaches. Migraine headache is typically associated with unilateral pain (affecting one half of the head) and pulsating in nature. It usually lasts between 2 to 70 hours. Symptoms of migraine include, but are not limited to, nausea, vomiting, photophobia (increased sensitivity to light), and phonophobia (increased sensitivity to sound). The cause of migraine headache is unknown. The most common theory is a disorder of the serotonergic control system. Migraine is usually treated with analgesics for the headache and antimetics for the nausea. In addition, triggering conditions such as noise or bright light should be avoided. Pericranial (jaw and neck) muscle tenderness is a most common finding in migraine. Thus, tender muscle trigger points can be at least part of the cause, and perpetuate most kinds of headaches. As described above, migraine is a disease that is associated with pain but is a disease that comprises more and different symptoms than symptoms of pain. Said types of disease that are intimately connected with symptoms of pain but that have in addition symptoms outside of pain/algesia, are referred to, herein, as pain-associated diseases. Some pain associated diseases are listed above in connection with disease-associated pain states and display pain as one characteristic disease symptom (e.g. migraine). Other pain associated diseases (either psychogenic or somatogenic) may be increased, provoked or even caused by pain, such as depression or epilepsy. The herein described definitions of pain, pain-stages, pain-states, pain-sensations and pain-sensitivities etc. also comprise equivalent states, stages, sensations and signalling cascades in lower animals or organisms, such as c. elegans. Moreover, the term pain also comprises pre-stages of pain.
The term "tissue" as used herein, refers to an ensemble of cells of the same origin which fulfil a specific function concertedly. Examples of a tissue include but are not limited to bone, cartilage, connective tissue, muscle tissue, nervous tissue, and epithelial tissue. Multiple tissues together form an "organ" to carry out a specific function. Examples of an organ include but are not limited to brain, spinal cord, joint, skeleton, muscle, blood, brain, heart, liver, kidney, stomach, and skin. For example, a joint is formed of many different tissues such as but not limited to bone, cartilage, synovium, muscle, ligament, and tendon. The term "tissue" furthermore relates to body liquids such as blood, liquor cerebrospinalis, saliva and lymph.
The terms "tissue status", "status of a tissue", "tissue state" and "state of a tissue" are used interchangeably herein referring to the condition of a tissue. The state of a tissue may be characterized by a specific morphology of such tissue or may be characterised by the expression of one or more specific molecules such as but not limited to peptides, proteins, and nucleic acids, or combinations thereof. The status of a tissue may be regarded as "healthy" or "normal" in case it resembles the condition of such tissue when being free from illness or injury and efficiently fulfilling its specific function. The status of a tissue may be regarded as "degenerative", "diseased" or "abnormal" in case such tissue fails to fulfil its function due to an illness or injury. Additionally or alternatively, the status of a tissue may be regarded as "degenerative", "diseased" or "abnormal" in case the morphology of the tissue or its molecule expression pattern is "altered" or "changed" in comparison to normal tissue. Accordingly, the morphology of a tissue or the expression pattern of specific molecules in a tissue may be an indicator for the state of a tissue. Examples of a tissues status include but are not limited to tissue degradation such as cartilage degradation, bone degradation, and degradation of the synovium, tissue inflammation such as cartilage inflammation, or inflammation of the synovium, tissue remodelling such as bon remodelling or cartilage remodelling, sclerosis, liquid accumulation, or proliferative tissue such as proliferation in wound healing processes, cystic formations, or in cancer. In the context of the present invention, an altered (pathological) tissue status is preferably characterised by the tissue exhibiting a different (higher) Slfn2 level than normal (healthy) tissue, wherein the normal tissue can be tissue from another individual that is known not to have a pain-related tissue status or disease (e.g. not to experience pain) or the same tissue at an earlier time point (preferably before onset or worsening/progressing of the pain-related tissue status or disease, e.g. of pain). A disease status is preferably characterised by an organ or individual exhibiting different slfn2 level in comparison to a normal organ or individual or in comparison to the same organ or individual at an earlier time point.
The term "pain-related or pain-associated tissue status" as used herein, refers to a status of a tissue typically associated with pain or a pain-associated disease, typically a diseased or abnormal tissue state such as but not limited to an infection of tissue, infarction, necrosis or other states of tissue associated with pain.
The term "tissue" as used herein, refers to an ensemble of cells of the same origin which concertedly fulfil a specific function. Examples of a tissue include but are not limited to bone, cartilage, connective tissue, muscle tissue, nervous tissue, and epithelial tissue. Multiple tissues together form an "organ" to carry out a specific function. Examples of an organ include but are not limited to joint, skeleton, muscle, blood, brain, heart, liver, kidney, stomach, and skin. For example, a joint is formed of many different tissues such as but not limited to bone, cartilage, synovium, muscle, ligament, and tendon.
The terms "disease" and "disorder" are used interchangeably herein, referring to an abnormal condition, especially an abnormal medical condition such as an illness or injury, wherein a tissue, an organ or an individual is not able to efficiently fulfil its function anymore. Typically, but not necessarily, a disease is associated with specific symptoms or signs indicating the presence of such disease. The presence of such symptoms or signs may thus, be indicative for a tissue, an organ or an individual suffering from a disease. An alteration of these symptoms or signs may be indicative for the progression of such a disease. A progression of a disease is typically characterized by an increase or decrease of such symptoms or signs which may indicate a "worsening" or "bettering" of the disease. The "worsening" of a disease is characterized by a decreasing ability of the tissue, organ or organism to fulfil its function efficiently, whereas the "bettering" of a disease is typically characterized by an increase in the ability of the tissue, an organ or an individual to fulfil its function efficiently. A tissue, an organ or an individual being at "risk of developing" a disease is in a healthy state but shows potential of a disease emerging. Typically, the risk of developing a disease is associated with early or weak signs or symptoms of such disease. In such case, the onset of the disease may still be prevented by treatment. Examples of a disease include but are not limited to traumatic diseases, inflammatory diseases, infectious diseases, cutaneous conditions, endocrine diseases, intestinal diseases, neurological disorders, joint diseases, genetic disorders, autoimmune diseases, and various types of cancer.
"Symptoms" of a disease are implication of the disease noticeable by the tissue, organ or organism having such disease and include but are not limited to pain, weakness, tenderness, strain, stiffness, and spasm of the tissue, an organ or an individual. "Signs" or "signals" of a disease include but are not limited to the change or alteration such as the presence, absence, increase or elevation, decrease or decline, of specific indicators such as biomarkers or molecular markers, or the development, presence, or worsening of symptoms. Symptoms of pain include, but are not limited to an unpleasant sensation that may be felt as a persistent or varying burning, throbbing, itching or stinging ache.
The term "biomarker" or "indicator" are used interchangeably herein. In the context of present invention, a biomarker can be defined as a substance within a biological system that is used as an indicator of a biological state of said system. In the art, the term "biomarker" is sometimes also applied to means for the detection of said endogenous substances (e.g. antibodies, nucleic acid probes etc, imaging systems). In the context of present invention, however, the term "biomarker" shall be only applied for the substance, not for the detection means. Thus biomarkers can be any kind of molecule present in a living organism, such as a nucleic acid (DNA, mRNA, miRNA, rRNA etc.), a protein (cell surface receptor, cytosolic protein etc.), a metabolite or hormone (blood sugar, insulin, estrogen, etc.), a molecule characteristic of a certain modification of another molecule (e.g. sugar moieties or phosphoryl residues on proteins, methyl-residues on genomic DNA) or a substance that has been internalized by the organism or a metabolite of such a substance. As shown by the inventors, there is surprisingly a close correlation between pain and the expression/steady state level of Slfn2. Slfn2 can thus be used as an indicator of pain, i.e. qualifies as biomarker for pain.
The indicator or biomarker refers to a sign or signal for a condition or is used to monitor a condition. Such a "condition" refers to the biological status of a cell, tissue or organ or to the health and/or disease status of an individual. An indicator may be the presence or absence of a molecule, including but not limited to peptide, protein, and nucleic acid, or may be a change in the expression level or pattern of such molecule in a cell, or tissue, organ or individual. An indicator may be a sign for the onset, development or presence of a disease in an individual or for the further progression of such disease. An indicator may also be a sign for the risk of developing a disease in an individual. Known indicators of pain include but are not limited to: Cystatin C (Pain 102 (2003) p.251 -256), Cathepsin H and Cathepsin C and the circulatory pain-indicators beta endorphin, serotonin, 5-hydroxy indole acetic acid, anandamide, N-palmitoylethanolamide (for an overview see e.g. Journal of the American Osteopathic Association (2007) 107; p.387- 400). The term "level of Slfn2" as used herein, refers to the expression level or the level of gene copies (also absence of Slfn2 or parts thereof or presence of fragments) present in a cell, tissue, organ or individual.
The term "expression level" (of a gene, here e.g. Slfn2) refers to the amount of gene product present in the body or a sample at a certain point of time. The expression level can e.g. be measured/quantified/detected by means of the protein or mRNA expressed from the gene. The expression level can for example be quantified by normalizing the amount of gene product of interest (e.g. Slfn2 mRNA or protein) present in a sample with the total amount of gene product of the same category (total protein or mRNA) in the same sample or a reference sample (e.g. a sample taken at the same time from the same individual or a part of identical size (weight, volume) of the same sample) or by identifying the amount of gene product of interest per defined sample size (weight, volume, etc.). The expression level can be measured or detected by means of any method as known in the art, e.g. methods for the direct detection and quantification of the gene product of interest (such as mass spectrometry) or methods for the indirect detection and measurement of the gene product of interest that usually work via binding of the gene product of interest with one or more different molecules or detection means (e.g. primer(s), probes, antibodies, protein scaffolds) specific for the gene product of interest, here Slfn2. The determination of the level of gene copies of Slfn2 comprising also the determination of the the absence or presence of one or more fragments (e.g. via nucleic acid probes or primers, e.g. quantitative PCR, Multiplex ligation-dependent probe amplification (MLPA) PCR (http://www.mlpa.com/) is also within the knowledge of the skilled artisan.
As used herein, the term "variant" is to be understood as a polynucleotide or protein which differs in comparison to the polynucleotide or protein from which it is derived by one or more changes in the sequence and comprises fragments or derivatives. The polypeptide or polynucleotide from which a protein or nucleic acid variant is derived is also known as the parent polypeptide or nucleic acid. Typically, a variant is constructed artificially, preferably by gene-technological means. Typically, the parent polypeptide or polynucleotide is a wild-type protein or nucleic acid. Further, the variants usable in the present invention may also be derived from homologs, orthologs, or paralogs of the parent molecule or from artificially constructed variant, provided that the variant exhibits at least one biological activity of the parent molecule, i.e. is functionally active, if the variant is meant to be a functional variant.
Alternatively or additionally, a "variant" as used herein, can be characterized by a certain degree of sequence identity to the parent polypeptide or parent nucleic acid from which it is derived. More precisely, a protein variant in the context of the present invention exhibits 80% or more sequence identity to its parent polypeptide. A nucleic acid variant in the context of the present invention exhibits 80% or more sequence identity to its parent nucleic acid. The term "80% or more sequence identity" is used throughout the specification with regard to polypeptide and nucleic acid sequence comparisons. This expression preferably refers to a sequence identity of 80% or more, 81 % or more, 82% or more, 83% or more, 84% or more, 85% or more, 86% or more, 87% or more, 88% or more, 89% or more, 90% or more, 91 % or more, 92% or more, 93% or more, 94% or more, 95% or more 96% or more, 97% or more, 98% or more, or 99% or more to the respective reference polypeptide or to the respective reference nucleic acid.
The similarity of nucleotide and amino acid sequences, i.e. the percentage of sequence identity, can be determined via sequence alignments. Such alignments can be carried out with several art-known algorithms, e.g. with the mathematical algorithm of Karl in and Altschul (Karlin & Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5877), with hmmalign (HMMER package, http://hmmer.wustl.edu/) or with the CLUSTAL algorithm (Thompson, J. D., Higgins, D. G. & Gibson, T. J. (1994) Nucleic Acids Res. 22, 4673- 80) available e.g. on http://www.ebi.ac.uk/Tools/clustalw/ or on http://www.ebi.ac.uk/Tools/clustalw2/index.html or on http://npsa-pbil.ibcp.fr/cgi- bin/npsa_automat.pl?page=/NPSA/npsa_clustalw.html. Preferred parameters used are the default parameters as they are set on http://www.ebi.ac.uk/Tools/clustalw/ or http://www.ebi.ac.uk/Tools/clustalw2/index.html. The grade of sequence identity (sequence matching) may be calculated using e.g. BLAST, BLAT or BlastZ (or BlastX). A similar algorithm is incorporated into the BLASTN and BLASTP programs of Altschul et al. (1990) J. Mol. Biol. 215: 403-410. BLAST nucleic acid searches are performed with the BLASTN program, score = 100, word length = 12, to obtain nucleic acid sequences that are homologous to those nucleic acids which encode Slfn2. BLAST protein searches are performed with the BLASTP program, score = 50, word length = 3, to obtain amino acid sequences homologous to Slfn2. To obtain gapped alignments for comparative purposes, Gapped BLAST is utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25: 3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs are used. Sequence matching analysis may be supplemented by established homology mapping techniques like Shuffle- LAGAN (Brudno M., Bioinformatics 2003b, 19 Suppl 1 :154-162) or Markov random fields. When percentages of sequence identity are referred to in the present application, these percentages are calculated in relation to the full length of the longer sequence, if not specifically indicated otherwise.
"Hybridization" can also be used as a measure of sequence identity or homology between two nucleic acid sequences. A nucleic acid sequence encoding miRNAs or a portion thereof can be used as a "hybridization probe" according to standard hybridization techniques. The hybridization of a Slfn2 probe to DNA or RNA from a test source is an indication of the presence of the Slfn2 DNA or RNA in the test source. Hybridization conditions are known to those skilled in the art and can be found, for example, in Current Protocols in Molecular Biology, John Wiley & Sons, N. Y., 6.3.1 - 6.3.6, 1991 . "Moderate hybridization conditions" are defined as equivalent to hybridization in 2X sodium chloride/sodium citrate (SSC) at 30°C, followed by a wash in 1 X SSC, 0.1 % SDS at 50°C. "Highly stringent conditions" are defined as equivalent to hybridization in 6X sodium chloride/sodium citrate (SSC) at 45°C, followed by a wash in 0.2 X SSC, 0.1 % SDS at 65°C. The terms "protein" and "polypeptide" are used interchangeably herein and refer to any peptide-linked chain of amino acids, regardless of length or post-translational modification. Proteins usable in the present invention (including protein derivatives, protein variants, protein fragments, protein segments, protein epitopes and protein domains) can be further modified by chemical or biological modification. This means such a biologically or chemically modified polypeptide comprises other chemical groups than the 20 naturally occurring amino acids. Examples of such other chemical groups include without limitation glycosylated amino acids, phosphorylated amino acids or covalent attachment of amino-acid chains e.g. for stabilization of the protein/polypeptide (such as attachment of, e.g. rPEG, XTEN or PAS). Modification of a polypeptide may provide advantageous properties as compared to the parent polypeptide, e.g. one or more of enhanced stability, increased biological half-life, or increased water solubility. Chemical modifications applicable to the variants usable in the present invention include without limitation: PEGylation, glycosylation of non-glycosylated parent polypeptides, or the modification of the glycosylation pattern present in the parent polypeptide, rPEGylation, XTENylation or PASylation.
In the context of the different aspects of present invention, the term "peptide" refers to a short polymer of amino acids linked by peptide bonds. It has the same chemical (peptide) bonds as proteins, but is commonly shorter in length. The shortest peptide is a dipeptide, consisting of two amino acids joined by a single peptide bond. There can also be a tripeptide, tetrapeptide, pentapeptide, etc. Preferably, the peptide has a length of up to 8, 10, 12, 15, 18 or 20 amino acids. A peptide has an amino end and a carboxyl end, unless it is a cyclic peptide. In the context of the different aspects of present invention, the term "polypeptide" refers to a single linear chain of amino acids bonded together by peptide bonds and preferably comprises at least about 21 amino acids. A polypeptide can be one chain of a protein that is composed of more than one chain or it can be the protein itself if the protein is composed of one chain.
In the context of the different aspects of present invention, the term "protein" refers to a molecule comprising one or more polypeptides that resume a secondary and tertiary structure and additionally refers to a protein that is made up of several polypeptides, i.e. several subunits, forming quaternary structures. The protein has sometimes non- peptide groups attached, which can be called prosthetic groups or cofactors.
In the context of present invention, the primary structure of a protein or polypeptide is the sequence of amino acids in the polypeptide chain. The secondary structure in a protein is the general three-dimensional form of local segments of the protein. It does not, however, describe specific atomic positions in three-dimensional space, which are considered to be tertiary structure. In proteins, the secondary structure is defined by patterns of hydrogen bonds between backbone amide and carboxyl groups. The tertiary structure of a protein is the three-dimensional structure of the protein determined by the atomic coordinates. The quaternary structure is the arrangement of multiple folded or coiled protein or polypeptide molecules molecules in a multi-subunit complex. The terms "amino acid chain" and "polypeptide chain" are used synonymously in the context of present invention. The term Slfn2 or Slfn2 protein also encompasses naturally occurring variants such as homologs and orthologs in same or different species, in particular in human. The different aspects of present invention also relate to functional variants of Slfn2 protein, either naturally occurring or non-naturally occurring. The term "variant" of a Slfn2 protein comprises fragments and derivatives of a protein. A fragment is a protein or polypeptide that carries one or more end-terminal (n- and/or c-terminal) or internal deletions of one, two or more amino acids, when compared to the full-length protein. A functional fragment of the protein is any fragment of this protein having at least one and preferably two or more of the functional characteristics of the full-length protein. The term derivative of a protein comprises any type of modification of the protein in comparison to the naturally-occurring form, and in the context of present invention especially in comparison to the Slfn2 protein according to SEQ IDs no. 2 and 9, which is not a deletion. A functional derivative of the protein is any derivative of this protein having at least one and preferably two or more of the functional characteristics of the unmodified protein. The variants of Slfn2 of present invention also comprise functional derivatives of fragments of Slfn2.
Non-naturally occurring variants may be obtained by a limited number of amino acid deletions, insertions and/or substitutions, particularly deletions, insertions and/or substitutions of at most 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid(s).
A Slfn2 variant of the present invention can e.g. be a functionally active variant, i.e. the variant maintains at least one of the biological functions of Slfn2 as described herein, e.g. its function in the context of pain (e.g. as manifestation of the pain phenotype "mechanic hyperalgesia") or as known in the literature. Preferably, maintenance of biological function is defined as having 50 % or more, 60 % or more, 70 % or more, 80 % or more or 90 % or more or 95 % or more of the activity of the natural occurring Slfn2. A non-functional variant of a Slfn2 protein (with less than the above-identified biological activity or even no detectable biological activity) may also be subject of one of the aspects of present invention, e.g. as negative control. The biological activity may be determined as known to the skilled person. For example, the manifestation of the pain phenotype "mechanic hyperalgesia" can be determined as detailed in the Examples and in Persson et al., 2009, Molecular Pain 5:7. The Slfn2 variant may be modified in order to comprise a further component. Accordingly, the variant may be a molecule having a domain composed of a naturally occurring Slfn2 protein or a variant thereof as detailed herein and at least one further component. In one embodiment variant may be a fusion protein comprising (i) a Slfn2 protein or functionally active variant and (ii) a further protein component. For example, the protein may be coupled to a marker, such as a tag used for purification purposes (e.g. 6 His (or HexaHis) tag, Strep tag, HA tag, c-myc tag or glutathione S-transferase (GST) tag). If e.g. a highly purified Slfn2 protein or variant should be required, double or multiple markers (e.g. combinations of the above markers or tags) may be used. In this case the proteins are purified in two or more separation chromatography steps, in each case utilizing the affinity of a first and then of a second tag. Examples of such double or tandem tags are the GST-His-tag (glutathione-S-transferase fused to a polyhistidine- tag), the 6xHis-Strep-tag (6 histidine residues fused to a Strep-tag), the 6XH is-tag 100- tag (6 histidine residues fused to a 12-amino-acid protein of mammalian MAP-kinase 2), 8xHis-HA-tag (8 histidine residues fused to a hemagglutinin-epitope-tag), His-MBP (His- tag fused to a maltose-binding protein, FLAG-HA-tag (FLAG-tag fused to a hemagglutinin-epitope-tag), and the FLAG-Strep-tag. The marker could be used in order to detect the tagged protein, wherein specific antibodies could be used. Suitable antibodies include anti-HA (such as 12CA5 or 3F10), anti-6 His, anti-c-myc and anti- GST. Furthermore, the Slfn2 protein could be linked to a marker of a different category, such as a fluorescence marker or a radioactive marker, which allows for the detection of Slfn2. In a further embodiment, Slfn2 could be part of a fusion protein, wherein the second part could be used for detection, such as a protein component having enzymatic activity.
In another embodiment of the present invention, the Slfn2 variant could be a Slfn2 fragment, wherein the fragment is still functionally active. This may include Slfn2 proteins with short internal and/or C- and/or N-terminal deletions (e.g. deletions of at most 20, 19, 18, 17, 16, 15, 14, 13, 12, 1 1 , 10, 9, 8, 7, 6 5, 4, 3, 2, or 1 amino acid). Additionally, the Slfn2 fragment may be further modified as detailed above for the Slfn2 protein.
Alternatively or additionally, the Slfn2 variant may comprise one or more amino acid substitution(s). Semi-conservative and especially conservative amino acid substitutions, wherein an amino acid is substituted with a chemically related amino acid are preferred. Typical substitutions are among the aliphatic amino acids, among the amino acids having aliphatic hydroxyl side chain, among the amino acids having acidic residues, among the amide derivatives, among the amino acids with basic residues, or the amino acids having aromatic residues. Typical semi-conservative and conservative substitutions are:
Amino acid Conservative substitution Semi-conservative substitution
A G; S; T N; V; C
C A; V; L M; I; F; G D E; N; Q A; S; T; K; R; H
E D; Q; N A; S; T; K; R; H
F W; Y; L; M; H 1; V; A
G A S; N; T; D; E; N; Q
H Y; F; K; R L; M; A
1 V; L; M; A F; Y; W; G
K R; H D; E; N; Q; S; T; A
L M; 1; V; A F; Y; W; H; C
M L; 1; V; A F; Y; W; C;
N Q D; E; S; T; A; G; K; R
P V; 1 L; A; M; W; Y; S; T; C; F
Q N D; E; A; S; T; L; M; K; R
R K; H N; Q; S; T; D; E; A
S A; T; G; N D; E; R; K
T A; S; G; N; V D; E; R; K; 1
V A; L; l M; T; C; N
w F; Y; H L; M; 1; V; C
Y F; W; H L; M; 1; V; C
Changing from A, F, H, I, L, M, P, V, W or Y to C is semi-conservative if the new cysteine remains as a free thiol. Furthermore, the skilled person will appreciate that glycines at sterically demanding positions should not be substituted and that P should not be introduced into parts of the protein which have an alpha-helical or a beta-sheet structure.
The Slfn2 protein or fragment or variant with substitution may be modified as detailed above for the Slfn2 protein or fragment or variant. In the following description of the invention all details given with respect to Slfn2 protein also relate to functionally active variants thereof, unless stated otherwise. The above modifications of the Slfn2 protein may be combined. The variant of the present invention may be e.g. fragment of Slfn2 having a marker fused to it, or a Slfn2 protein fragment comprising one or more amino acid substitutions. A derivative of Slfn2 protein or a Slfn2 protein fragment is a Slfn2 protein or fragment carrying any kind of modification of the amino acid sequence that is not a deletion and/or carrying any other kind of modification, such as a chemical or biological modification. Derivatives comprise proteins carrying one or more modifications e.g. leading to the stabilization of the polypeptide, or enabling a specific targeting of the polypeptide to certain cells or facilitating its entry into or uptake by cells (such as cell- permeant phosphopeptides ortho coupling to cell-permeant peptide vectors, e.g. based on the antennapedia/penetratin, TAT, and signal-peptide based sequences; or coupling to parts of ligands for specific transporters or importers). According to one embodiment, the Slfn2 protein is a naturally occurring Slfn2 protein as detailed above, such as a naturally occurring mouse Slfn2 protein such as comprising or having SEQ ID NO: 2, or a naturally occurring human orthologous protein, such as comprising or having SEQ ID NO: 9. The terms "nucleic acid" or "nucleic acid molecule" are used synonymously and are understood as single or double-stranded oligo- or polymers of deoxyribonucleotide or ribonucleotide bases or both. Typically, a nucleic acid is formed through phosphodiester bonds between the individual nucleotide monomers. In the context of the present invention, the term nucleic acid includes but is not limited to ribonucleic acid (RNA) and deoxyribonucleic acid (DNA) molecules. The depiction of a single strand of a nucleic acid also defines (at least partially) the sequence of the complementary strand. The nucleic acid may be single or double stranded, or may contain portions of both double and single stranded sequences. The nucleic acid may be obtained by biological, biochemical or chemical synthesis methods or any of the methods known in the art. As used herein, the term "nucleic acid" comprises the terms "polynucleotide" and "oligonucleotide".
In the context of the different aspects of present invention, the term nucleic acid comprises cDNA, genomic DNA, recombinant DNA, cRNA and mRNA. A nucleic acid may consist of an entire gene, or a portion thereof, the nucleic acid may also be a microRNA (miRNA) or small interfering RNA (siRNA). MiRNAs are short ribonucleic acid (RNA) molecules, on average only 22 nucleotides long, found in all eukaryotic cells. MircoRNAs (miRNAs) are post-transcriptional regulators that bind to complementary sequences on target messenger RNA transcripts (mRNAs), usually resulting in translational repression and gene silencing. Small interfering RNAs (siRNAs), sometimes known as short interfering RNA or silencing RNA, are short ribonucleic acid (RNA molecules), between 20-25 nucleotides in length. They are involved in the RNA interference (RNAi) pathway, where they interfere with the expression of specific genes. The nucleic acid can also be an artificial nucleic acid. Artificial nucleic acids include polyamide or peptide nucleic acid (PNA), morpholino and locked nucleic acid (LNA), as well as glycol nucleic acid (GNA) and threose nucleic acid (TNA). Each of these is distinguished from naturally-occurring DNA or RNA by changes to the backbone of the molecule.
The nucleic acids, can e.g. be synthesized chemically, e.g. in accordance with the phosphotriester method (see, for example, Uhlmann, E. & Peyman, A. (1460) Chemical Reviews, 90, 543-584). Aptamers are nucleic acids which bind with high affinity to a polypeptide, here Slfn2. Aptamers can be isolated by selection methods such as SELESIfn2 (see e.g. Jayasena (1469) Clin. Chem., 45, 1628-50; Klug and Famulok (1464) M. Mol. Biol. Rep., 20, 97-107; US 5,582,981 ) from a large pool of different single-stranded RNA molecules. Aptamers can also be synthesized and selected in their mirror-image form, for example as the L-ribonucleotide (Nolte et al. (1466) Nat. Biotechnol., 14, 1 1 16-9; Klussmann et al. (1466) Nat. Biotechnol., 14, 1 1 12-5). Forms which have been isolated in this way enjoy the advantage that they are not degraded by naturally occurring ribonucleases and, therefore, possess greater stability. Nucleic acids may be degraded by endonucleases or exonucleases, in particular by DNases and RNases which can be found in the cell. It is, therefore, advantageous to modify the nucleic acids in order to stabilize them against degradation, thereby ensuring that a high concentration of the nucleic acid is maintained in the cell over a long period of time (Beigelman et al. (1465) Nucleic Acids Res. 23:3989-94; WO 95/1 1910; WO 98/37240; WO 97/291 16). Typically, such stabilization can be obtained by introducing one or more internucleotide phosphorus groups or by introducing one or more non-phosphorus internucleotides. Suitably modified internucleotides are compiled in Uhlmann and Peyman (1460), supra (see also Beigelman et al. (1465) Nucleic Acids Res. 23:3989- 94; WO 95/1 1910; WO 98/37240; WO 97/291 16). Modified internucleotide phosphate radicals and/or non-phosphorus bridges in a nucleic acid which can be employed in one of the uses according to the invention contain, for example, methyl phosphonate, phosphorothioate, phosphoramidate, phosphorodithioate and/or phosphate esters, whereas non-phosphorus internucleotide analogues contain, for example, siloxane bridges, carbonate bridges, carboxymethyl esters, acetamidate bridges and/or thioether bridges. It is also the intention that this modification should improve the durability of a pharmaceutical composition which can be employed in one of the uses according to the invention.
The term "oligonucleotide" when used in the context of one of the different aspects of present invention, refers to a nucleic acid of up to about 50 nucleotides, e.g. 2 to about 50 nucleotides in length.
The term "polynucleotide" when used in the context of one of the different aspects of present invention, refers to a nucleic acid of more than about 50 nucleotides in length, e.g. 51 or more nucleotides in length. The term means for detection or means for detecting (especially in the context of detecting Slfn2) as used herein refers to any means suitable for specific detection of Slfn or nucleic acid, especially in a probe, in isolated organic matter (e.g. isolated protein or nucleic acid), a tissue, an organ or an animal body. The detection is usually mediated by a specific binding to the target molecule, here Slfn2 nucleic acid or polypeptide.
Probes and primers are short polynucleotides or oligonucleotides for the detection of nucleic acids in a sample or in vivo by hybridizing (probe) to the target nucleic acid or by hybridization to and amplification of the target nucleic acid. Nucleic acids suitable for use as probe or primer comprise e.g. a polynucleotide probe, one or more primers (e.g. a primer pair), preferably one or more primers for polymerase chain reaction (PCR), reverse transcription (RT) reaction, or DNA sequencing, a peptide- or polyamid- nucleic acid (PNA), a locked nucleic acid (LNA), a glycol nucleic acid (GNA), a threose nucleic acid (TNA), a microRNA (miRNA), and a small interfering RNA (siRNA). The one or more primers (e.g. a primer pair) can e.g. be for real time polymerase chain reaction (RT-PCR) or for quantitative real time polymerase chain reaction (qRT-PCR).
The term "probe" as used herein refers to a nucleic acid which is typically used for the detection of target RNA and/or DNA sequences that is complementary to the sequence of the probe. A probe hybridizes to single-stranded nucleic acid (DNA or RNA) whose nucleotide sequence allows for nucleotide pairing due to complementarity between the probe and the target sequence. The length of a probe depends on the intended use as well as the required specificity of the probe. Typically, a probe is 20-500 (i.e. 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 1 10, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500) nucleotides long, preferably 20-100 nucleotides, more preferably 20-50. Probes are used in various experimental set ups such as but not limited to Southern and Northern Blots, for real-time PCR and In Situ Hybridization (ISH) as well as for microarray experiments. A probe may be unlabeled, directly labelled, or indirectly labelled, such as with biotin to which a streptavidin complex may later bind. Said label may be a molecule detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means. For example, suitable labels include 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens and other entities which are or can be made detectable. A label may be incorporated into nucleic acids at any position, e.g. at the 3' end, at the 5' end or internally. The term "probe" also encompasses nucleic acids differing in the composition of their backbone such as but not limited to peptide nucleic acids (PNAs), locked nucleic acids (LNAs), glycol nucleic acids (GNAs) and threose nucleic acids (TNAs).
Said nucleic acid as a probe may be unlabeled, directly labeled, or indirectly labeled, such as with biotin to which a streptavidin complex may later bind. Said label may be a molecule detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means. For example, suitable labels include 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, digoxigenin, or haptens and other entities which are or can be made detectable. A label may be incorporated into nucleic acids at any position, e.g. at the 3' end, at the 5' end or internally. The term "primer" as used herein refers to a single-strand nucleic acid which typically serves as a starting point for DNA-replicating enzymes. A primer binds to or hybridises with a DNA template and typically comprises a sequence being complementary to the DNA sequence to which it is supposed to bind. A primer may also comprise additional sequences e.g. sequences serving as nuclease cleavage sites (e.g. Bam H1 , Hind III, etc.). The length of a primer is chosen depending on the intended use. For instance, primers used for the amplification of DNA in Polymerase-Chain Reactions (PCR) typically have a length of at least 10 nucleotides, preferably between 10 to 50 (i.e. 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 45, 50) nucleotides, more preferably between 15 and 30 nucleotides. Shorter primers of at least 5 nucleotides are used for sequencing of DNA templates. Also encompassed in the term "primer" are "degenerate primers", which are a mixture of similar but not identical primers. A primer may be tagged or labeled with a marker molecule detectable by spectroscopic, photochemical, biochemical, immunochemical, chemical, or other physical means.
Probes and primers for the detection of Slfn2 are known in the art (see e.g. Wern-Joo et al, 2007) and can be ordered custom-made by commercial vendors against any known sequence including that of Slfn2. Examples include (all taken from Wern-Joo et al., 2007):
5'-CTCACCTCAGAAAACAGGAGAATGC-3' (sense murine Slfn2; SEQ ID NO:10), 5'-CAGAAGTGAGTGACAGGCAGCTG-3' (anti-sense, murine Slfn2; SEQ ID NO:1 1 ), 5'-CGGCTAGCTTGGTGGCTCCTAGCGRR-3' (sense Slfn2 -1766; SEQ ID NO.12), 5'-CGGCTAGCGGAACTGAACTGGAGCT-3' (sense Slfn2 -1265; SEQ ID NO.13), 5'-CCCTCGAGCCATGTCAGCAGTTGGTAAAGC-3' (anti-sense SIfn 2 +198; SEQ ID NO:14). In addition, using standard software, the skilled artisan knows how to select suitable primers.
The above-mentioned peptide nucleic acids (PNAs), locked nucleic acids (LNAs), glycol nucleic acids (GNAs) and threose nucleic acids (TNAs) are artificial nucleic acids which can also be used as probes or primers. Each of these is distinguished from naturally- occurring DNA or RNA by changes to the backbone of the molecule. MiRNAs and siRNAs can also be used as probes. The above-mentioned nucleic acids are e.g. for detecting the SIfn nucleic acid sequences of present invention, e.g. one or more of the SIfn nucleic acid sequences of embodiment 20 and are e.g. for use in one of the methods according to one of the aspects of present invention. The above-mentioned nucleic acids may allow the determination of the expression level of Slfn2 on the Slfn2 gene or Slfn2 mRNA level, e.g. in a sample, such as in a sample from a subject or in an assay, such as a biochemical or cellular assay (e.g. in a mixture in the context of a biochemical assay or in a cell in the context of a cellular assay).
The term "variant" of a nucleic acid comprises fragments and derivatives of a nucleic acid. The variants of present invention also comprise functional derivatives of fragments of the nucleic acid. Non-naturally occurring variants may be obtained by a limited number of nucleotide deletions, insertions and/or substitutions, particularly deletions, insertions and/or substitutions of at most 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 nucleotide(s).
A fragment of a nucleic acid is a nucleic acid that carries one or more terminal (5' (upstream) and/or 3' (downstream)) or internal deletions of one, two or more
nucleotides, when compared to the full-length nucleic acid. A functional fragment of the nucleic acid is any fragment of the Slfn2 nucleic acid having at least one and preferably two or more of the functional characteristics of the full-length nucleic acid (e.g. the ability to confer Slfn2-characteristic transcriptional regulation of downstream elements or the characteristic to code for a Slfn2 protein).
The term "derivative of a nucleic acid" comprises any type of modification (e.g. a chemical or biological modification) of the nucleic acid in comparison to the naturally- occurring form, and in the context of present invention especially in comparison to the Slfn2 nucleic acid according to SEQ ID NOs. 1 , 3, 4, 6 or 8 that is not a deletion.
Derivatives comprise Slfn2 nucleic acids carrying one or more modifications leading to the stabilization of the nucleic acid (such as phosphoorothioate modification or modification of the nucleic acid backbone). A functional derivative of the nucleic acid is any derivative of this nucleic acid having at least one and preferably two or more of the functional characteristics of the unmodified nucleic acid.
The term "Slfn2 nucleic acid" encompasses nucleic acids coding for the above Slfn2 protein and regulatory nucleic acids of the Slfn2 gene as well as naturally occurring and non-naturally occurring variants thereof (as defined herein). Preferably, the term relates to coding or non-coding regions of the Slfn2 gene, wherein these sections are of a relevant size in order to be specific for that gene. Examples of those regions are introns, exons or regulatory elements such as a Slfn2 promoter. The term Slfn2 or Slfn2 nucleic acid also encompasses naturally occurring variants such as homologs and orthologs in same or different species, in particular in human (e.g. the nucleic acids according to SEQ IDs NO. 1 (mouse), 6 (rat) or 8 (human). The different aspects of present invention also relate to functional variants of Slfn2 nucleic acid, either naturally occurring or non- naturally occurring. Preferred aspects of a Slfn2 nucleic acid relate to the Slfn2 gene, promoter, DNA, cDNA or mRNA or oligonucleotides or polynucleotides able to specifically hybridize with one of these nucleic acids. Examples of Slfn2 nucleic acids are e.g. a Slfn2 nucleic acid coding for the naturally occurring Slfn2 protein as detailed above (e.g. a protein comprising or having an amino acid sequence according to SEQ ID NO: 2, 7 or 9) such as a nucleic acid comprising or having a nucleotide sequence according to SEQ ID NO: 1 , 3, 4, 6, or 8). Another example of a Slfn2 nucleic acid is a Slfn2 nucleic acid able to drive expression of a downstream element such as a nucleic acid derived from the Slfn2 gene and comprising regulatory elements thereof but lacking the Slfn2 coding sequence, e.g. a nucleic acid comprising or consisting of a nucleotide sequence according to SEQ ID NO:4.
A Slfn2 nucleic acid variant of the present invention can be a functionally active variant, i.e. the variant maintains at least one of the biological functions of Slfn2 nucleic acid as described herein, e.g. its function in the context of pain (e.g. as manifestation of the pain phenotype "mechanic hyperalgesia", the ability to confer Slfn2-characteristic transcriptional regulation of downstream elements or the characteristic to code for a Slfn2 protein) or as known in the literature. Preferably, maintenance of biological function is defined as having 50 % or more, 60 % or more, 70 % or more, 80 % or more or 90 % or more or 95 % or more of the activity of the natural occurring Slfn2. A non- functional variant of a Slfn2 nucleic acid (with less than the above-identified biological activity or even no detectable biological activity) may also be subject of one of the aspects of present invention, e.g. as negative control. The biological activity may be determined as known to the skilled person, for example, the manifestation of the transcriptional activity as detailed in Wern-Joo et al., 2007; examples of functional and non-functional variants (fragments and derivatives) of nucleic acids, especially different promoter variants are disclosed in Wern-Joo et al, 2007 the disclosure of which is explicitly included herein, as reference. Preferred examples nucleic acids comprising a Slfn2 promoter comprise a Slfn2 nucleic acid from -1256 to +1 or from -1265 to +198 or from +1699 to +1 or from -1699 to +198 with respect to the sequence according to SEQ ID NO: 5.
The term "antibody or fragment thereof, as used herein, refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e. molecules that contain an antigen binding site that specifically binds an antigen. Also comprised are immunoglobulin-like proteins that are selected through techniques including, for example, phage display to specifically bind to a target molecule or target protein. The immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., lgG1 , lgG2, lgG3, lgG4, lgA1 and lgA2) or subclass of immunoglobulin molecule. The "antibodies and fragments thereof include, but are not limited to, polyclonal, monoclonal, monovalent, bispecific, heteroconjugate, multispecific, human, humanized (in particular CDR-grafted), deimmunized, or chimeric antibodies, single chain antibodies (e.g. scFv), Fab fragments, F(ab')2 fragments, fragments produced by a Fab expression library, diabodies or tetrabodies (Holliger P. et al., 1993), nanobodies, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
In some embodiments, the antibody fragments are mammalian, preferably human antigen-binding antibody fragments and include, but are not limited to, Fab, Fab' and F(ab')2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (dsFv) and fragments comprising either a VL or VH domain. Antigen-binding antibody fragments, including single-chain antibodies, may comprise the variable domain(s) alone or in combination with the entirety or a portion of the following: hinge region, CL, CH1 , CH2, and CH3 domains. The antigen-binding fragments may also comprise any combination of variable domain(s) with a hinge region, CL, CH1 , CH2, and CH3 domains.
Antibodies usable in the invention may be from any animal origin including birds and mammals. Preferably, the antibodies are human, simian (e.g. chimpanzee, bonobo, macaque), rodent (e.g. mouse and rat), donkey, sheep rabbit, goat, guinea pig, camel, horse, or chicken. It is particularly preferred that the antibodies are of human or murine origin. As used herein, "human antibodies" include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins.
The techniques for preparing antibodies and antibody fragments are well known in the art. Means of preparing and characterizing antibodies or antibody fragments are also well known in the art (see, for example, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988; incorporated herein by reference). Antibodies or antibody fragments specific for the Slfn2 protein may be used in a variety of assays in order to quantitate the Slfn2 protein. Well known methods include, for example, immunoprecipitation, antibody sandwich assays, ELISA and affinity chromatography methods (see above).
The antibody that recognizes the target antigen, here Slfn2 or a functionally active variant thereof is generally called the "primary antibody". Said antibody may be labeled with a detectable tag/label in order to allow direct detection of the target antigen. Said detectable tag/label may be an enzymatic, fluorescent or radioisotope tag/label. Usually, however, the primary antibody is not labeled for direct detection. Instead a "secondary antibody" that has been labeled with a detectable tag/label (e.g. enzymatic, fluorescent or radioisotope tag/label) is applied in a second step to probe for the primary antibody, which is bound to the target antigen. For example, the primary antibody or the secondary antibody may be labeled with an affinity tag such as biotin. As to detectable tags/labels which may be used for the detection of Slfn2 and/or as to the specific detection methods it is also referred to the information provided with respect to the methods of certain aspects of the present invention.
Antibodies against Slfn2 are known in the art (see e.g. Katsoulidis et al., 2009) and can be purchased from commercial vendors such as antibodies-online.com (http://www.antibodies-online.com). An example of an n-terminal anti Slfn2 antibody suitable for use in e.g. Western Blot or ELISA is a polyclonal rabbit anti-mouse antibody that has been generated using a synthetic peptide corresponding to the n-terminal residues of mouse Slfn2 and that can be purchased from antibodies-online under ABIN393095. The term "protein scaffold" in the context of present invention refers to non-antibody recognition proteins derived from one or more of structurally different protein families and able to bind with high specificity to a selected target, thereby mimeting the binding principle of immunoglobulins to varying degrees (see e.g. Hey, T., et al., 2005: Artificial, non-antibody binding proteins for pharmaceutical and industrial applications, Trends in Biotechnology, Vol. 23, No. 10, the disclosure of which is incorporated herein, by reference). Examples comprise lipocalin-derived repeat proteins such as darpins, ankyrin-repeat proteins such as anticalins, nanobodies, affibodies, maxibodies, trans- bodies, tetranectin, iMabs, Adnectin, domain antibodies, Kunitz-type domains, Evibodies, Affilins, Microbodies).
The terms "biochip" and "microarray" are interchangeably used herein
The terms "attached" or "immobilized", as used herein, refer to the binding between the nucleic acid(s), protein, polypeptide, or peptide and the solid support and may mean that the binding between the nucleic acid, protein, polypeptide, or peptide and the solid support is sufficient to be stable under conditions of binding, washing, analysis and removal. The binding may be covalent or non-covalent. Covalent bonds may be formed directly between the nucleic acid, protein, polypeptide, or peptide and the solid support or may be formed by a cross linker or by inclusion of specific reactive groups on either the solid support or the nucleic acid, protein, polypeptide, or peptide, or both. Non- covalent binding may be electrostatic, hydrophilic and hydrophobic interactions or combinations thereof. Immobilization or attachment may also involve a combination of covalent and non-covalent interaction.
A marker (or tag or label) is any kind of substance which is able to indicate the presence of another substance or complex of substances. The marker can be a substance that is linked to or introduced in the substance to be detected. Detectable markers are used in molecular biology and biotechnology to detect e.g. a protein, a product of an enzymatic reaction, a second messenger, DNA, interactions of molecules etc. Examples of suitable marker or labels include a fluorophore, a chromophore, a radiolabel, a metal colloid, an enzyme, or a chemiluminescent or bioluminescent molecule. Examples of fluorophores include fluorescein, rhodamine, and sulfoindocyanine dye Cy5. Examples of radiolabels include 3H, 14C, 32P, 33P, 35S, 99mTc or 125l. Examples of enzymes include horseradish peroxidase, alkaline phosphatase, glucose oxidase, and urease. Further examples and preferred embodiments are detailed herein.
Different types of chemical labels or tags can be conjugated to secondary or primary antibodies and other molecules to facilitate their visualization (i.e., detection and measurement) by various methods. Radioisotopes were used extensively in the past, but they are expensive, have a short shelf-life, offer no improvement in signal:noise ratio and require special handling and disposal. Enzymes and fluorophores have largely replaced radioactive isotopes as detectable tags for assays. A number of advancements in reagents and instrumentation make these newer technologies more versatile and powerful. Enzymatic tags such as horseradish peroxidase (HRP) are most commonly used for blotting, immunoassays and immunohistochemistry methods. Fluorescent tags are used predominately for cellular imaging, nucleic acid amplification and sequencing and microarrays; however, fluorescence technology is developing rapidly for application in all types of assays.
The detection of protein often involves the use of specific antibodies. Accordingly, the detection of Slfn2 protein or a variant thereof may include a specific Slfn2 antibody or protein scaffold. Alternatively, antibodies can be raised using well established techniques for immunizing animals with prepared forms of the antigen. A variety of reagents is available to assist in antibody production and purification, and various companies specialize in antibody production services. Depending on the application to be performed, different levels of purity and types of specificity are needed in a supplied primary antibody. To name just a few parameters, antibodies may be monoclonal or polyclonal, supplied as antiserum or affinity-purified solution, and validated for native protein or denatured protein detection.
An antibody that recognizes the target antigen, here Slfn2 or fragment or variant thereof, is called the "primary antibody." If this antibody is labeled with a tag, direct detection of the antigen is possible. Usually, however, the primary antibody is not labeled for direct detection. Instead a "secondary antibody" that has been labeled with a detectable tag is applied in a second step to probe for the primary antibody, which is bound to the target antigen. Thus, the antigen is detected indirectly. Another form of indirect detection involves using a primary or secondary antibody that is labeled with an affinity tag such as biotin. Then a secondary (or tertiary) probe, such as streptavidin that is labeled with the detectable enzyme or fluorophore tag, can be used to probe for the biotin tag to yield a detectable signal. Several variants of these probing and detection strategies exist. However, each one depends on a specific probe (e.g., a primary antibody) whose presence is linked directly or indirectly to some sort of measurable tag (e.g., an enzyme whose activity can produce a colored product upon reaction with its substrate).
Usually, a primary antibody without a detectable label and some sort of secondary (indirect) detection method is required in assay methods. Nevertheless, nearly any antibody can be labeled with biotin, HRP enzyme or one of several fluorophores if needed. Most primary antibodies are produced in mouse, rabbit or one of several other species. Nearly all of these are antibodies of the IgG class. Therefore, it is relatively easy and economical for manufacturers to produce and supply ready-to-use, labeled secondary antibodies for most applications and detection systems. Even so, several hundred options are available, differing in the level of purity, IgG- and species-specificity, and detection label. The choice of secondary antibody depends upon the species of animal in which the primary antibody was raised (the host species). For example, if the primary antibody is a mouse monoclonal antibody then the secondary antibody must be an anti-mouse antibody obtained from a host other than the mouse.
With biotin-binding proteins as probes, the highly specific affinity interaction between biotin and avidin or streptavidin protein is the basis for many kinds of detection and affinity-purification methods. Biotin is very small (244 Daltons), so its covalent attachment to antibodies or other probes rarely interferes with their functions. Yet its presence as a label on a probe allows efficient and specific secondary detection with either avidin or streptavidin. Both kinds of biotin-binding proteins are available in purified forms labeled with enzymatic or fluorescent tags that enable detection in many kinds of assays systems. Enzymatic labels are most commonly used as secondary antibody (or streptavidin) tags for detection in blotting and immunoassays. Enzymes provide detectable signal via their activity; reaction with a specific substrate chemical yields a colored, light-emitting, or fluorescent product. While reporter enzymes like beta-galactosidase and luciferase have been successfully used to make probes, alkaline phosphatase (AP) and horseradish peroxidase (HRP) are the two enzymes used most extensively as labels for protein detection. An array of chromogenic, fluorogenic and chemiluminescent substrates is available for use with either enzyme. Alkaline phosphatase, usually isolated from calf intestine, is a large (140 kDa) protein that catalyzes the hydrolysis of phosphate groups from a substrate molecule resulting in a colored or fluorescent product or the release of light as a byproduct of the reaction. AP has optimal enzymatic activity at a basic pH (pH 8-10) and can be inhibited by cyanides, arsenate, inorganic phosphate and divalent cation chelators, such as EDTA. As a label for Western blotting, AP offers a distinct advantage over other enzymes. Because its reaction rate remains linear, detection sensitivity can be improved by simply allowing a reaction to proceed for a longer time period.
Horseradish peroxidase is a 40 kDa protein that catalyzes the oxidation of substrates by hydrogen peroxide, resulting in a colored or fluorescent product or the release of light as a byproduct of the reaction. HRP functions optimally at a near-neutral pH and can be inhibited by cyanides, sulfides and azides. Antibody-HRP conjugates are superior to antibody-AP conjugates with respect to the specific activities of both the enzyme and antibody. In addition, its high turnover rate, good stability, low cost and wide availability of substrates makes HRP the enzyme of choice for most applications. Because of the small size of the HRP enzyme, further increases in sensitivity may be achieved by using poly-HRP conjugated secondary antibodies and may eliminate the need for using ABC type amplification systems for some researchers. Fluorescent labels for detection were historically used in a small number of cell biology applications such as flow cytometry (FC), fluorescence-activated cell sorting (FACS) and immunohistochemistry (IHC) using fluorescence microscopy. Until recently, the two most common fluorophores for labeling probes were fluorescein (fluorescein isothiocyanate, FITC) and rhodamine (tetramethyl rhodamine isothiocyanate, TRITC). Other labels include fluorescent proteins such as the various forms of green fluorescent protein (GFP) and the phycobiliproteins (allophycocyanin, phycocyanin, phycoerythrin and phycoerythrocyanin). While having the ability to produce an intense fluorescent signal for detection, fluorescent proteins can be difficult to optimize for conjugation purposes and may create steric hindrance or background signal issues in binding assays.
The use of fluorophore-conjugated probes in blotting and immunoassays requires fewer steps compared to the use of enzymatic labels because there is no substrate development step to perform. While the protocol is shorter, fluorescent detection requires special equipment and the sensitivity is not a high as that which can be obtained with enzymatic chemiluminescent systems. Although not as sensitive as enzymatic detection, fluorescent detection methods reduce chemical waste and have the added advantage of multiplex compatibility (using more than one fluorophore in the same experiment).
Alternatively or additionally, two markers may be used in order to detect proximity of two substances, e.g. the test compound or the known Slfn2 ligand and the Slfn2 protein. Papain and cathepsin L and H are known ligands for Slfn2. The markers may be, e.g. one radioactive or fluorescent marker and one scintillator (e.g. for a scintillation proximity assay) or two fluorescent markers may be used (e.g. for FRET). In one example the Slfn2 protein and the test substance could be labeled with a first and a second marker. In case the test substance is bound to the protein, and the labels are therefore in close proximity, energy could be transferred from the first to the second label, thus detecting the interacting of Slfn2 protein and test substance. This test could be designed as a competition binding test, wherein a known Slfn2 ligand carries one of the labels. Examples of suitable marker combinations include:
- radiolabels 3H, 33P, 35S or 14C, 125l combined with scintillator such as Yttrium silicate or polyvinyl-toluene, e.g. compartmented in a microparticle or
- a donor fluorescent markers such as fluorescein, Lucifer Yellow, B-phycoerythrin, 9- acridineisothiocyanate, Lucifer Yellow VS, 4-acetamido-4'-isothiocyanatostilbene- 2,2'-disulfonic acid, 7-diethylamino-3-(4'-isothiocyanatophenyl)-4-methylcoumarin, succinimdyl 1 -pyrenebutyrate, and 4-acetamido-4'-isothiocyanatostilbene-2,2'- disulfonic acid derivatives combined with a acceptor fluorescent marker such as LC- Red 610, LC -Red 640, LC-Red 670, LC -Red 705, Cy5, Cy5.5, Lissamine rhodamine B sulfonyl chloride, tetramethyl rhodamine isothiocyanate, rhodamine x isothiocyanate, erythrosine isothiocyanate, fluorescein, diethylenetriamine pentaacetate or other chelates of Lanthanide ions (e.g., Europium, or Terbium).
Detection of Slfn2 can also refer to detection of Slfn2 mRNA:
Suitable methods of detecting mRNA include e.g. Northern blot analysis, nuclease protection assays (NPA), in situ hybridization, and reverse transcription-polymerase chain reaction (RT-PCR). For the Northern blotting procedure, RNA samples may be first separated by size via electrophoresis in an agarose gel under denaturing conditions. The RNA is then transferred to a membrane, crosslinked and hybridized with a labeled probe. Nonisotopic or high specific activity radio labeled probes can be used including random- primed, nick-translated, or PCR-generated DNA probes, in vitro transcribed RNA probes, and oligonucleotides. Additionally, sequences with only partial homology (e.g., cDNA from a different species or genomic DNA fragments that might contain an exon) may be used as probes.
The Nuclease Protection Assay (NPA) is an extremely sensitive method for the detection and quantitation of specific mRNAs. The basis of the NPA is solution hybridization of an antisense probe (radio labeled or non-isotopic) to an RNA sample. After hybridization, single-stranded, unhybridized probe and RNA are degraded by nucleases. The remaining protected fragments are separated e.g. on an acrylamide gel. Solution hybridization is typically more efficient than membrane-based hybridization, and it can accommodate up to 100 g of sample RNA, compared with the 20-30 g maximum of blot hybridizations. NPAs are also less sensitive to RNA sample degradation than Northern analysis since cleavage is only detected in the region of overlap with the probe (probes are usually about 100-400 bases in length). In RT-PCR, an RNA template is copied into a complementary DNA (cDNA) using a retroviral reverse transcriptase. The cDNA is then amplified exponentially by PCR. Relative quantitative RT-PCR involves amplifying an internal control simultaneously with the gene of interest. The internal control is used to normalize the samples. Once normalized, direct comparisons of relative abundance of a specific mRNA can be made across the samples. Competitive RT-PCR is used for absolute quantitation. This technique involves designing, synthesizing, and accurately quantitating a competitor RNA that can be distinguished from the endogenous target by a small difference in size or sequence. Known amounts of the competitor RNA are added to experimental samples and RT-PCR is performed. Signals from the endogenous target are compared with signals from the competitor to determine the amount of target present in the sample.
The above methods may include nucleic acid labeling. A series of techniques are known to the skilled person allowing for labeling of DNA, RNA or oligonuleotides. These include for example Nick translational labeling, random primed DNA labeling, PCR labeling of DNA probes and oligonucleotide 375' end labeling, transcriptional labeling of RNA probes, oligonucleotide 375' end labeling and oligonucleotide tailing.
The nick translation method is based on the ability of DNase I to introduce randomly distributed nicks into DNA. DNA polymerase I synthesizes DNA complementary to the intact strand in a 5'→ 3' direction using the 3'-OH termini of the nick as a primer. The 5' → 3' exonucleolytic activity of DNA Polymerase I simultaneously removes nucleotides in the direction of synthesis. The polymerase activity sequentially replaces the removed nucleotides with isotope-labeled or hapten-labeled deoxyribonucleoside triphosphates. At low temperature (15°C), the unlabeled DNA in the reaction is thus replaced by newly synthesized labeled DNA. Common labels include digoxigenin-, biotin-, or fluorochromes such as fluorescein or tetramethylrhodamin.
The method of "random primed" DNA labeling is based on the hybridization of a mixture of all possible hexanucleotides to the DNA to be labeled. All sequence combinations are represented in the hexanucleotide primer mixture, which leads to binding of primer to the template DNA in a statistic manner. Thus an equal degree of labeling along the entire length of the template DNA is guaranteed. The complementary strand is synthesized from the 3' OH termini of the random hexanucleotide primer using Klenow enzyme, labeling grade. Modified deoxyribonucleoside triphosphates (e.g. [32P]-, [35S]-, [3H]-, [125l]-, digoxigenin- or biotin-labeled) present in the reaction are incorporated into the newly synthesized complementary DNA strand. The polymerase chain reaction (PCR) allows the amplification of minute amounts of DNA. The only prerequisite is that some sequence information of the target sequence is known for synthesizing the appropriate primers. The combination of labeling with PCR is a powerful tool for the analysis of PCR products, and also for the preparation of labeled probes from small amounts of a respective target sequence. For example digoxigenin, a steroid hapten, may be used to label DNA, RNA, or oligonucleotides for hybridization, and subsequent color- or luminescent detection. The digoxigenin is usually coupled to dUTP via an alkali-labile ester bond. The labeled dUTP can be easily incorporated by enzymatic nucleic-acid synthesis using DNA polymerases.
Oligonucleotides may enzymatically be labeled at their 3'-end with terminal transferase either by incorporation of a label such as single digoxigenin-labeled dideoxyuridine- triphosphate (DIG-ddUTP) or by the addition of a longer nucleotide tail. Terminal Transferase catalyzes the template independent addition of deoxy- and dideoxynucleoside triphosphates to the 3ΌΗ ends of double and single-stranded DNA fragments and oligonucleotides. Terminal transferase incorporates digoxigenin-, biotin-, and fluorochrome-labeled deoxy- and dideoxynucleotides as well as radioactive labeled deoxy-and dideoxynucleotides. Alternatively or additionally, oligonucleotides may be labeled at the 5'-terminus, e.g. by reacting with a phosphoramidite in a final step according to the classical solid phase phosphoramidite synthesis method. By this process a 5'-terminal amino function is created. Treatment with ammonia releases the oligonucleotide from the support and cleaves the protecting groups. In the subsequent step the digoxigenin moiety is introduced at the 5'-position. Different labels are known which may be used in the above labeling methods. Some of them including their detection are exemplarily described in the following:
Biotin-labeled compounds can be detected for example by anti-biotin antibodies or by streptavidin conjugates. Anti-biotin antibodies (e.g. monoclonal anti-biotin antibody or Fab-fragment, conjugated with alkaline phosphatase (AP)) may be used in the detection of biotin-labeled nucleic acids by enzyme immunoassay with luminescence on nylon membranes. This method of detection may be employed for detection of biotin labeled nucleic acids on membranes (e.g. Southern blots, dot blots), in cells and tissues (e.g. in situ hybridization), immunoblotting, immunohistochemistry or ELISA. Streptavidin conjugates are used for the detection of biotin-labeled substances (e.g., biotinylated antibodies) which can be used for several immunological detection systems. For this, streptavidin e.g. from Streptomyces avidinii could be coupled to alkaline phosphatase or to β-peroxidase. This method of detection may be employed with immunoblotting, immunohistochemistry or ELISA.
Probe-target hybrids may be detected with an enzyme-linked immunoassay. This immunochemical detection step is usually more sensitive than radioactive detection procedures. In this assay, the membrane may be blocked to prevent non-specific interaction of the antibody with the filter. Alkaline phosphatase-conjugated antibody, specific for digoxigenin, recognizes the digoxigenin molecule on the labeled hybrid. Addition of an alkaline phosphatase substrate allows the visualization of the hybrids.
For chemiluminescence detection, suitable substrates for alkaline phosphatase such as disodium 3-(4-methoxyspiro {1 ,2-dioxetane-3,2-(5-chloro)tricyclo [3.3.1 .13,7]decan}-4- yl)phenyl phosphate or disodium 4-chloro-3-(methoxyspiro {1 ,2-dioxetane-3,2-(5- chloro)tricyclo [3.3.1 .13,7]decan}-4-yl)phenyl phosphate belong to the group of the dioxetane phenyl phosphates. Upon dephosphorylation by alkaline phosphatase, an intermediate is formed whose decomposition results in light emission which can be recorded e.g. on X-ray film.
Colorimetric detection of DIG-labeled probes is usually performed with colorless substrates which form a redox system. Examples are like 5-bromo-4-chloro-3-indolyl- phosphate and 4-Nitro-blue-tetrazolium-chloride. 5-bromo-4-chloro-3-indolyl-phosphate is oxidized by the alkaline phosphatase to indigo by release of a phosphate group. In parallel, 4-Nitro-blue-tetrazolium-chloride is reduced to diformazan. The reaction products form a water insoluble dark blue to brownish precipitate, depending on the type of membrane. Various reporter molecules can be coupled to detecting antibodies to visualize the specific probe-target hybridization including, but not limited to, enzyme-coupled antibodies, fluorochrome-labeled antibodies (detection by fluorescent microscope and specific filters which allow visualization of the wavelength emitted by the fluorescent dye) and antibodies coupled to colloidal gold (detection by electron microscope on cryostatic sections).
Multiple simultaneous hybridizations can be performed by using combinations of digoxigenin-, biotin- and fluorochrome-labeled probes to localize different chromosomal regions or different RNA sequences in one preparation. Such multiprobe experiments are made possible by the availability of different fluorescent dyes coupled to antibodies. These include fluorescein or FITC (fluorescein isothiocyanate; yellow), rhodamine or TRITC (tetramethylrhodamine isothiocyanate; red) and AMCA (amino-methylcoumarin acetic acid; blue).
Suitable diagnostic assays comprise the detection of Slfn2 protein:
Suitable methods for detecting a protein are described herein and include e.g. detection of protein immunostaining, protein immunoprecipitation, Immunoelectrophoresis, immunoblotting, Western blotting, spectrophotometry, enzyme assays etc.. The method may require protein purification prior to the detection, which could involve protein isolation (e.g. by chromatography methods, protein extraction, protein solubilization, gel electrophoresis, and electrofocusing).
Protein immunostaining is an antibody-based method to detect a specific protein in a sample. The term immunostaining was originally used to refer to the immunohistochemical staining of tissue sections. Now however, immunostaining encompasses a broad range of techniques used in histology, cell biology, and molecular biology that utilize antibody-based staining methods. Immunohisto- or -cytochemistry of tissue sections or cells which are preserved by fixation.
While the first cases of IHC staining used fluorescent dyes, other non-fluorescent methods using enzymes such as peroxidase and alkaline phosphatase are now used more often. These enzymes are capable of catalyzing reactions that give a coloured product that is easily detectable by light microscopy. Alternatively, radioactive elements can be used as labels, and the immunoreactions can be visualized by autoradiography. Tissue preparation or fixation is essential for the preservation of cell morphology and tissue architecture. Inappropriate or prolonged fixation may significantly diminish the antibody binding capability. Many antigens can be successfully demonstrated in formalin-fixed paraffin-embedded tissue sections. Optimization of fixation methods and times, pre-treatment with blocking agents, incubating antibodies with high salt, and optimizing post-antibody wash buffers and wash times may be important for obtaining high quality immunostaining.
Western blotting allows the detection of specific proteins (native or denatured) from extracts made from cells or tissues, before or after any purification steps. Proteins are generally separated by size using gel electrophoresis before being transferred to a synthetic membrane (typically nitrocellulose or PVDF) via dry, semi-dry, or wet blotting methods. The membrane can then be probed using antibodies using methods similar to immunohistochemistry, but without a need for fixation. Detection is typically performed using peroxidase linked antibodies to catalyze a chemiluminescent reaction. Western blotting is a routine molecular biology method that can be used to semi quantitatively or quantitatively compare protein levels between extracts. The size separation prior to blotting allows the protein molecular weight to be gauged as compared with known molecular weight markers. Western blotting is an analytical technique used to detect specific proteins in a given sample of tissue homogenate or extract. It uses gel electrophoresis to separate proteins by the length of the polypeptide (denaturing conditions) or by the 3-D structure of the protein (native/ non-denaturing conditions).
The enzyme-linked immunosorbent assay or ELISA is a diagnostic method for quantitatively or semi-quantitatively determining protein concentrations from blood plasma, serum or cell/tissue extracts in a multi-well plate format (usually 96-wells per plate). Broadly, proteins in solution are adsorbed to ELISA plates. Antibodies specific for the protein of interest are used to probe the plate. Background is minimized by optimizing blocking and washing methods (as for IHC), and specificity is ensured via the presence of positive and negative controls. Detection methods are usually colorimetric or chemiluminescence based.
Electron microscopy or EM can be used to study the detailed micro architecture of tissues or cells. Immuno-EM allows the detection of specific proteins in ultrathin tissue sections. Antibodies labeled with heavy metal particles (e.g. gold) can be directly visualized using transmission electron microscopy. While powerful in detecting the sub- cellular localization of a protein, immuno-EM can be technically challenging, expensive, and require rigorous optimization of tissue fixation and processing methods.
The terms "subject" or "individual" are used interchangeably herein. As used herein, an "individual" refers to a human or a non-human animal (e.g. a mammal, avian, reptile, fish, amphibian or invertebrate; preferably an individual that can either benefit from one of the different aspects of present invention (e.g. a method of treatment or a drug identified by present methods) or that can be used as laboratory animal for the identification or characterisation of a drug or a method of treatment. The individual can e.g. be a human, a wild-animal, domestic animal or laboratory animal; examples comprise: mammal, e.g. human, non-human primate (chimpanzee, bonobo, gorilla), dog, cat, rodent (e.g. mouse, guinea pig, rat, hamster or rabbit, horse, donkey, cow, sheep, goat, pig, camel; avian, such as duck, dove, turkey, goose or chick; reptile such as: turtle, tortoise, snake, lizard, amphibian such as frog (e.g. Xenopus laevis); fish such as koy or zebrafish; invertebrate such as a worm (e.g. c.elegans) or an insect (such as a fly, e.g. drosophila melanogaster). The term individual also comprises the different morphological developmental stages of avian, fish, reptile or insects, such as egg, pupa, larva or imago. The individual may be an individual which is suspected to experience pain. The individual may be diagnosed and/or prognosed to experience pain. The individual as mentioned in the method above may also be an individual which is experiencing pain. The individual may be retested for experiencing pain and may be diagnosed and/or prognosed to still experiencing pain.
The term "sample", as used in the context of the different aspects of present invention, preferably refers to a biologically sample. The term "sample" or "sample of interest" are used interchangeably herein, referring to a small part intended to represent the whole of a tissue, an organ or an individual. Upon analysis a sample provides information about the tissue status or the health or diseased status of an organ or individual. Examples of samples include but are not limited to fluid samples such as cerebrospinal fluid, blood, serum, plasma, synovial fluid, urine, saliva, and lymphatic fluid, or solid samples such as biopsy samples, tissue, and tissue-extracts, e.g. taken from nervous tissue (e.g from the spinal cord), skin, muscle, cartilage, bone, synovium, perichondrium, capsule, and connective tissue. Further examples of samples are cell cultures or tissue cultures such as but not limited to cultures of neural cells.
Analysis of a sample may be accomplished on a visual or chemical basis. Visual analysis includes but is not limited to microscopic imaging or radiographic scanning of a tissue, organ or individual allowing for morphological evaluation of a sample. Chemical analysis includes but is not limited to the detection of the presence or absence of specific indicators or alterations in their amount or level. For example, a tissue sample may be removed from a subject by conventional biopsy techniques or a blood sample may be taken from a subject by conventional blood collection techniques. The sample, e.g. tissue or blood sample, may be obtained from a subject prior to initiation of the therapeutic treatment, during the therapeutic treatment, and/or after the therapeutic treatment, e.g. with a pain reducing compound. It is preferred that the sample is a body fluid sample, a tissue sample, a cell colony sample, a single cell sample or a cell culture sample. More preferably, the tissue sample is a section or an explant sample, e.g. an explant sample of dorsal root ganglia or spinal cord. The term "body fluid sample" refers to a liquid sample derived from the body of a subject. Said body fluid sample may be a blood, urine, cerebrospinal fluid, cerumen (earwax), endolymph, perilymph, gastric juice, mucus, peritoneal fluid, pleural fluid, saliva, or sebum (skin oil) sample including components or fractions thereof. Said body fluid samples may be mixed or pooled. Thus, a body fluid sample may be a mixture of a blood sample and anurine sample or a mixture of a blood sample and cerebrospinal fluid sample. A "body fluid sample" may be provided by removing a body liquid from a subject, but may also be provided by using previously isolated body fluid sample material. Preferably, the blood sample of a subject is whole blood or a blood fraction such as serum or plasma. It is also particularly preferred to use blood cells also known as hemopoietic cells. It is preferred that the tissue sample has a weight of between 0.1 and 500 mg, more preferably of between 0.5 and 250 mg, and most preferably of between 1 and 50 mg, i.e. 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, or 500 mg. It is also preferred that the cell sample (e.g. cell colony sample or cell culture sample) consists of between 100 and 1000 cells, more preferably of between 200 and 800 cells, and most preferably of between 400 and 600 cells.
It is further preferred that the body fluid sample has a volume of between 0.1 and 20 ml, more preferably of between 0.5 and 10 ml, more preferably between 1 and 8 ml and most preferably between 2 and 5 ml, i.e. 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 ml. More preferably, the blood sample has a volume of between 0.1 and 20 ml, more preferably of between 0.5 and 10 ml, and most preferably of between 1 and 5 ml, i.e. 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 ml.
The term "reference", especially in the context of "reference individual", "reference sample" or "reference value" in the context of present invention refers to a comparison or standard that is characteristic or representative for a certain (health) status, disease etc. Thus, a reference value, is a standard value for a certain parameter (e.g. expression level of a certain indicator/biomarker molecule) that is typical for a certain status (e.g. a disease status or health status), a reference individual is an individual that has been selected for comparison and has a certain health state or disease, a reference sample can e.g. be a sample from a reference individual or an artificial sample with a characteristic level of a certain indicator or biomarker typical for a disease state or health state. The term "reference sample" as used herein, refers to a sample which is analysed in a substantially identical manner as the sample of interest and whose information is compared to that of the sample of interest. A reference sample thereby provides a standard allowing for the evaluation of the information obtained from the sample of interest.
A reference sample may be derived from a healthy or normal tissue, organ or individual, thereby providing a standard of a healthy status of a tissue, organ or individual. Differences between the status of the normal reference sample and the status of the sample of interest may be indicative of the risk of disease development or the presence or further progression of such disease or disorder.
A reference sample may be derived from an abnormal or diseased tissue, organ or individual thereby providing a standard of a diseased status of a tissue, organ or individual. Differences between the status of the abnormal reference sample and the status of the sample of interest may be indicative of a lowered risk of disease development or the absence or bettering of such disease or disorder. A reference sample may also be derived from the same tissue, organ, or individual as the sample of interest but has been taken at an earlier time point. Differences between the status of the earlier taken reference sample and the status of the sample of interest may be indicative of the progression of the disease, i.e. a bettering or worsening of the disease over time. A reference sample was taken at an earlier or later time point in case a period of time has lapsed between taking of the reference sample and taking of the sample of interest. Such period of time may represent years (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 years), months (1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 months), weeks (e.g. 1 , 2, 3, 4, 5, 6, 7, 8 weeks), days (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500 days), hours (1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 hours), minutes (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60 minutes), or seconds (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60 seconds).
The reference sample representative for a status or stage of pain may be from a control subject known to experience pain. Preferably, the control sample is from a control subject known to experience algesia, particularly hyperalgesia. The control subject may be a mammal such as a human, rodent (e.g. rat, hamster, or mouse) or monkey, or may be another animal than a mammal such as an avian. Preferably, both the sample or value and the reference sample or value are from subjects of the same species (e.g. human), more preferably of the same gender (e.g. female or male) and/or of a similar age or phase of life (e.g. infant, young child, juvenile, adult, or elderly). The reference or reference sample in the different aspects and embodiments of present invention is preferably derived from a healthy individual, a diseased individual, or from the same individual as the sample of interest. Where the reference (e.g. reference value) or reference sample was taken from the same individual as the sample of interest, the reference (e.g. reference value) or reference sample was preferably taken at an earlier or later time point then the sample of interest. The time period which has lapsed between taking of the reference (e.g. reference value) or reference sample and taking of the reference (e.g. reference value) or sample or value of interest preferably represents years (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 years), months (1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 months), weeks (e.g. 1 , 2, 3, 4, 5, 6, 7, 8 weeks), days (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500 days), hours (1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12 hours), minutes (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60 minutes), or seconds (e.g. 1 , 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60 seconds). Alternatively or additionally, the reference sample is a reference sample with a level of Slfn2 representative for a healthy individual or representative for the presence or absence of an altered tissue status or disease or representative for an increased or decreased risk of developing an altered tissue status or disease. In embodiments, wherein the reference or reference sample is derived from a healthy individual or an individual with a decreased risk of developing an altered tissue status or a disease or with a level of Slfn2 representative of the absence of an altered tissue status or disease, an elevated level of Slfn2 in the reference or value or sample of interest in comparison to said reference (value) or reference sample indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual. In embodiments, wherein the reference is derived from a diseased individual or an individual with an increased risk of developing an altered tissue status or a disease or a value representative of the presence of an altered tissue status or disease, a similar level of Slfn2 indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual. In embodiments, wherein the reference (value) or reference sample is (from) the same individual as the individual of interest at an earlier time point, an elevated level of Slfn2 in the individual/value/sample of interest indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual. In embodiments, wherein the reference (value) or reference sample is (from) the same individual as the individual/sample of interest at an earlier time point, a lowered level of Slfn2 in the sample of interest indicates (a) an alteration of the tissue status or an improvement or absence of a disease and/or (b) a decreased risk to develop a deteriorated tissue status or a disease and/or (c) a declined progression of the deteriorated tissue status or the disease.
In embodiments, wherein the reference (value) or reference sample is (from) the same individual as the sample/value of interest at an earlier time point, a similar level of Slfn2 in the sample of interest indicates (a) a similar risk to develop a deteriorated tissue status or a disease and/or (b) a stagnation in the progression of a deteriorated tissue status or a disease, and/or (c) a persistence of the deteriorated tissue status or the disease in the individual. In embodiments, wherein the reference (value) or reference sample is derived from a healthy individual or from an individual with a decreased risk of developing an altered tissue status or disease or comprises a level of Slfn2 representative of a healthy individual or of a status of disease-absence or for a decreased risk of developing an altered tissue status or disease, wherein an elevated level of Slfn2 indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
In embodiments, wherein the reference (value) or reference sample is derived from a diseased individual or from an individual with an increased risk of developing an altered tissue status or disease or comprises a level or amount of Slfn2 representative for a diseased individual or for a status of disease-presence or for an increased risk of developing an altered tissue status or disease, wherein a similar level of Slfn2 indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
In embodiments, wherein the reference (value) or sample is derived from the same individual as sample of interest and was taken at an earlier time point, an elevated level of Slfn2 in the sample of interest indicates (a) the presence of a deteriorated tissue status or a disease and/or (b) an increased risk to develop a deteriorated tissue status or a disease and/or (c) the progression of a deteriorated tissue status or a disease in the individual.
In embodiments, wherein the reference (value) or reference sample is derived from the same individual as sample of interest and was taken at an earlier time point, a lowered level of Slfn2 in the sample of interest indicates (a) an alteration of the tissue status or an improvement or absence of a disease and/or (b) a decreased risk to develop a deteriorated tissue status or a disease and/or (c) a declined progression of the deteriorated tissue status or the disease. In embodiments, wherein the reference sample is derived from the same individual as sample of interest and was taken at an earlier time point, a similar level of Slfn2 in the sample of interest indicates (a) a similar risk to develop a deteriorated tissue status or a disease and/or (b) a stagnation in the progression of a deteriorated tissue status or a disease, and/or (c) a persistence of the deteriorated tissue status or the disease in the individual.
The terms "lowered" or "decreased", especially in the context of the level of Slfn2 refer to the level of Slfn2 in the sample being reduced in comparison to the reference or reference sample. The terms "elevated" or "increased", especially in the context of the level of Slfn2 refer to the level of Slfn2 in the sample being higher in comparison to the reference or reference sample. E.g. a miRNA that is detectable in higher amounts in cerebrospinal fluid or spinal cord tissue of one individual suffering from pain than in cerebrospinal fluid or spinal cord tissue of individuals not suffering from pain, has an elevated level. For Slfn2, an elevated level in a patient sample (such as cerebrospinal fluid or blood) indicates the presence of a pain-associated disease or pain-associated tissue state or pain or an increased susceptibility or increased probability to develop a pain-associated disease or pain-associated tissue state or pain, especially the presence or an increased susceptibility to neuropathic algesia or neuropathic pain. As used herein, "treat", "treating" or "treatment" of a disease or disorder means accomplishing one or more of the following: (a) reducing the severity of the disorder; (b) limiting or preventing development of symptoms characteristic of the disorder(s) being treated; (c) inhibiting worsening of symptoms characteristic of the disorder(s) being treated; (d) limiting or preventing recurrence of the disorder(s) in an individual that have previously had the disorder(s); and (e) limiting or preventing recurrence of symptoms in individuals that were previously symptomatic for the disorder(s). As used herein, "prevent", "preventing", "prevention", or "prophylaxis" of a disease or disorder means preventing that such disease or disorder occurs in patient.
The terms "pharmaceutical", "medicament" and "drug" are used interchangeably herein referring to a substance and/or a combination of substances being used for the identification, prevention or treatment of a tissue status or disease.
The terms "specifically binds", "specific binding" or the like, mean that an antibody or antigen-binding fragment thereof forms a complex with an antigen that is relatively stable under physiologic conditions. Specific binding can be characterized by an equilibrium dissociation constant of at least about 1 x10"6 M or less (e.g., a smaller KD denotes a tighter binding). Methods for determining whether two molecules specifically bind are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like. An isolated antibody that specifically binds murineSlfn2 may, however, exhibit cross-reactivity to other antigens such as Slfn2 or Slfn12L molecules from other species. Moreover, multi-specific antibodies (e.g., bispecifics) that bind to Slfn2 and one or more additional antigens are nonetheless considered antibodies that "specifically bind" Slfn2, as used herein.
As used herein, a "patient" means any mammal, reptile or bird that may benefit from a treatment with the compounds described herein. Preferably, a "patient" is selected from the group consisting of laboratory animals (e.g. mouse or rat), domestic animals (including e.g. guinea pig, rabbit, chicken, turkey, pig, sheep, goat, camel, cow, horse, donkey, cat, or dog), or primates including chimpanzees and human beings. It is particularly preferred that the "patient" is a human being. The present invention will now be further described. In the following passages, different aspects of the invention are described in more detail, wherein this description and the terms used therein are to be understood in the context of the whole application, i.e. in light of the previous passages (e.g. the definitions and methods) and the consecutive passages (e.g. the below listed examples and embodiments). Each aspect so defined may be combined with any other aspect or aspects unless clearly indicated to the contrary. In particular, any feature indicated as being preferred or advantageous may be combined with any other feature or features indicated as being preferred or advantageous, unless clearly indicated to the contrary.
In a first aspect, present invention relates to Slfn2 for use as an indicator of a pain- related tissue status or a pain related disease or as an indicator of the risk for developing a pain-related tissue status or disease.
The surprising finding of the inventors that the presence or absence of Slfn2 correlates with pain, especially neuropathic pain in mice leads to the conclusion that Slfn2 can be used as indicator/biomarker for tissue statuses or diseases that correlate with pain and for pain itself. Thus, Slfn2 is indicative of a pain-related tissue status or a pain-related disease. The presence, absence or altered amounts of Slfn2 are thus a sign for the presence, absence or progression of a pain-related tissue status or disease.
Accordingly, a second aspect of present invention relates to Slfn2 for use in the diagnosis or prognosis of a pain-related tissue status or of a pain-related disease.
Due to the different expression levels of Slfn2 related to different pain-states, Slfn2 is used as an indicator of a tissue status or a disease in the context of present invention.
According to a preferred embodiment of the first and second aspect of present invention, the level of Slfn2 is indicative of a pain-related tissue status or the presence of a pain-related disease and/or the risk of developing a pain-related tissue status or a pain-related disease and/or the progression or a stage of a pain-related tissue status or a pain-related disease in an individual. The level of Slfn2 in the first and second aspect of present invention is the expression level of Slfn2 or the level or the amount (number) of Slfn2 gene (copies), wherein the level can be the level of Slfn2 in an individual (wherein Slfn2 needs to be determined by a method able to directly determine and quantify Slfn present in an individual without a sample needing to be taken) or in a sample of an individual.
The sample to be used in the first and second aspect of present invention can be any suitable sample, e.g. such as herein defined wherein cerebrospinal fluid is one of the preferred embodiments.
In one embodiment of the first pr second aspect of present invention, it is suitable to compare the level of Slfn2 to the level of Slfn2 in one or more references or reference samples, wherein the reference(s) or reference sample(s) can be any suitable reference(s) or reference sample(s), e.g. such as herein defined.
According to one preferred embodiment of the first and second aspect of the invention, the reference is selected from the group consisting of
a) an individual not having a pain-related tissue status or disease,
b) an individual having a pain-related tissue status or disease (especially one that is known to be associated with a certain level of Slfn2) or
c) the same individual as the individual to be tested at an earlier or later time point (especially suitable for a staging or monitoring of the proceeding of a pain-related tissue status or a pain-related disease, such as pain) or
d) a value representative for the level of slfn2
(i) in absence of the a pain-related tissue status or disease,
(ii) in presence of a pain-related altered tissue status or disease or e) a value representative for an increased or decreased risk of developing a pain- related altered tissue status or disease.
According to another embodiment of the first and second aspect of present invention, the reference is (derived from) an individual with a decreased
risk of developing a pain-related tissue status or disease or is a value representative of the absence of a pain-related tissue status or disease. According to another preferred embodiment of the first and second aspect of present invention, the reference is a sample that is selected from the group consisting of a) a reference sample derived from an individual not having a pain-related tissue status or disease,
b) a reference sample from an individual having a pain-related tissue status or disease, c) a reference sample from the same individual as the sample to be tested taken at an earlier or later time point or
d) a reference sample with a level or amount of slfn2 representative for
(i) an individual having or not having a pain-related tissue status or disease or for
(ii) the absence or presence of the pain-related disease or altered tissue status or for
(iii) an increased or decreased risk of developing a pain-related tissue status or disease.
According to another embodiment of the first and second aspect of present invention, the reference sample is derived from an individual with a decreased risk of developing a pain-related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual not having a pain-related tissue status or disease or for a status of disease-absence or for a decreased risk of developing a pain-related tissue status or disease, wherein an elevated level of Slfn2 in the tested sample indicates
a. the presence of a pain-related tissue status or disease and/or b. an increased risk of developing a pain-related tissue status or disease and/or
c. the progression of a pain-related tissue status or disease
in the individual of interest.
According to another preferred embodiment of the first and second aspect the reference is the same individual at an earlier time point or the sample is derived from the same individual as the tested sample and was taken at an earlier time point, wherein
(a) an elevated level of Slfn2 in the tested sample indicates
(i) the presence of a pain-related tissue status or disease and/or (ii) an increased risk of developing a pain-related tissue status or disease and/or
(iii) the progression of the pain-related tissue status or disease (b) a lowered level of Slfn2 in the tested sample indicates
(i) an improvement or the absence of a pain-related tissue status or an improvement or absence of a pain-related disease and/or
(ii) a decreased risk to develop a pain-related tissue status or disease and/or
(iii) a declined progression of the pain-related tissue status or disease (c) a similar level of Slfn2 in the tested sample indicates
According to another embodiment of the first and second aspect of present invention the reference is the same individual as the individual of interest at an earlier time point or the sample is derived from the same individual as the individual of interest, wherein the sample is derived from the same individual as the tested sample and was taken at an earlier time point, and wherein
(a) an elevated level of Slfn2 in the tested sample indicates
(iv) the presence of a pain-related tissue status or disease and/or
(v) an increased risk of developing a pain-related tissue status or disease and/or
(vi) the progression of the pain-related tissue status or disease
(b) a lowered level of Slfn2 in the tested sample indicates
(iv) an improvement or the absence of a pain-related tissue status or an improvement or absence of a pain-related disease and/or
(v) a decreased risk to develop a pain-related tissue status or disease and/or
(vi) a declined progression of the pain-related tissue status or disease
(c) a similar level of Slfn2 in the tested sample indicates
(i) a similar or persistent risk to develop a pain-related tissue status or disease and/or
(ii) a stagnation in the progression of a pain-related tissue status or disease and/or
(iii) the persistence of the a pain-related tissue status or disease in the individual. Comparison of the Slfn2 level in the individual of interest (i.e. the individual to be diagnosed or prognosed) with the level of the reference (e.g. reference value or reference individual) or reference sample by means of one of the different embodiments of the first or second aspect of present invention thus serves as basis for an objective and quantifiable diagnosis or prognosis of the presence or absence or staging of a pain- related tissue state or a pain-related disease, such as pain.
The sample, e.g. the reference or tested sample of the first, second, third and fourth aspect of present invention can be any suitable sample, e.g. such as disclosed herein e.g. tissue such as extraneural or neural tissue or one or more extraneural or neural cells and/or body fluid such as cerebrospinal fluid. It can also be suitable to take samples of different origin such as a tissue sample and a body fluid sample. The Slfn2 according to one of the embodiments of the first and second aspect of present invention can e.g. a Slfn2 gene or gene product, preferably a Slfn2 mRNA, or Slfn2 protein or a variant (such as a functionally active variant) thereof.
According to one embodiment of the first and second aspect of present invention Slfn2 is a gene and the gene
a. is a naturally occurring or non-naturally occurring Slfn2 gene variant, or
b. codes for a Slfn2 protein or a functionally active variant thereof that has
i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9,
11. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
in. an amino acid sequence that has at least 80%, at least 90% at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
IV. an amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to SEQ ID NO:2, and/or v. an amino acid sequence with at least 50% identity with the amino acid sequence according to SEQ ID NO:2 and/or vi. an amino acid sequence with one of the following sequence motifs: YxxlxV, YxxlxVxxW, YxxlxVxxWS or YxxlxVxxWSxS with "x" standing for any amino acid and/or
vii. carries a variant AAA domain, preferably with 50, 60, 70, 80, 90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2.
According to another embodiment of the first and second aspect of present invention Slfn2 is a gene product, wherein the gene product
a) is a naturally occurring Slfn2 protein variant, preferably a homolog or
ortholog in same or different species, or a non-naturally occurring Slfn2 protein variant,
b) is a Slfn2 protein or a functionally active variant thereof having
i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
iii. an amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
iv. an amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to SEQ ID NO:2, and/or
v. an amino acid sequence with at least 50% identity with the amino acid sequence according to SEQ ID NO:2 and/or vi. an amino acid sequence with one of the following sequence motifs: YxxlxV, YxxlxVxxW, YxxlxVxxWS or YxxlxVxxWSxS with "x" standing for any amino acid and/or vii. carries a variant AAA domain, preferably with 50, 60, 70, 80,
90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2, c) is a Slfn2 nucleic acid encoding a protein as defined in b), or
d) is a Slfn2 nucleic acid having
(i) a nucleic acid sequence according to SEQ ID NO: 1 , 6 or 8,
(ii) a nucleic acid sequence that is a fragment of the nucleic acid
sequence according to (i), or
(iii) a nucleic acid sequence that has at least 80%, at least 90%, at least
95% or at least 99% sequence identity to the nucleic acid sequence according to (i) or (ii).
The first or second aspects of present invention are e.g. suitable for diagnosing and/or prognosing pain in an in vitro or in vivo test model (i.e. a test animal, such as a non- human test animal, e.g. in the context of clinical studies) or in a patient (such as a human or non-human animal).
Another embodiment of the first and second aspect of present invention comprises determining the expression level of Slfn2 and preferably comprising determining the expression level of Slfn2 on the gene, mRNA or protein level.
According to yet another embodiment of the first and second aspect of present invention, the tissue status is a pain-associated tissue status, such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue, the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
According to another embodiment of the first and second aspect of present invention the pain-related or pain-associated disease is a pain, e.g.
(i) acute pain, subacute pain, or chronic pain,
(ii) nociceptive pain, neuropathic pain, inflammatory pain, or a mixture thereof, (iii) disease-associated pain, or
(iv) algesia.
According to another embodiment of the first and second aspect of present invention,
(i) the acute pain is somatogenetic (organic) pain,
(ii) the chronic pain is somatogenetic (organic) pain or psychogenic
(psychosomatic) pain,
(iii) the nociceptive pain is superficial somatic pain or deep pain, preferably deep somatic pain or visceral pain,
(iv) the neuropathic pain is central neuropathic pain or peripheral neuropathic pain,
(v) the mixed pain is a mixture of nociceptive pain and neuropathic pain,
(vi) the disease-associated pain is pain associated with depression, migraine, epilepsy, cancer, or diabetes, or
(vii) the algesia is hyperalgesia.
According to another embodiment of the first and second aspect of present invention the pain-related or pain-associated disease is characterized by an altered level of pain- related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
According to another embodiment of the first and second aspect of present invention the diagnosis comprises
(i) determining the presence or occurrence of pain,
(ii) monitoring the course of pain,
(iii) staging of pain,
(iv) classification of a subject with pain.
According to another embodiment of the first and second aspect of present invention, the prognosis comprises predicting or estimating the occurrence or reoccurrence, preferably the severity of occurrence or reoccurrence, of pain. In a third aspect, present invention relates to a method of identifying
(i) a pain-related tissue status or the presence of a pain-related disease and/or
(ii) the risk of developing a pain-related tissue status or a pain-related disease and/or
(iii) the progression or a stage of a pain-related tissue status or a pain-related disease
in an individual, the method comprising detecting the level of Slfn2.
In a fourth aspect, present invention relates to a method of identifying
(ii) a pain-related tissue status or the presence of a pain-related disease and/or
(iii) the risk of developing a pain-related tissue status or a pain-related disease and/or
(iv) the progression or a stage of a pain-related tissue status or a pain- related disease
in an individual, the method comprising detecting the level of Slfn2.
The individual of the fourth aspect of present invention can be any suitable individual such as herein defined.
According to one embodiment, the fourth aspect of present invention further comprises comparing the level of Slfn2 (e.g. the level of Slfn2 in the individual of interest (i.e. the test animal or the patient)) to the level of Slfn2 in one or more references or reference samples. The level of Slfn2 can e.g. be the level of Slfn2 in an individual (wherein Slfn2 needs to be determined by a method able to directly determine and quantify Slfn present in an individual without a sample needing to be taken) or in a sample of an individual. According to another embodiment of the fourth aspect of present invention, the level of Slfn2 is the expression level of Slfn2 or the level or the amount (number) of Slfn2 gene (copies).
According to another embodiment of the fourth aspect of present invention, the reference is selected from the group consisting of an individual not having a pain-related tissue status or disease, an individual having a pain-related tissue status or disease or the same individual as the individual to be tested at an earlier or later time point or a value representative for the level of slfn2 in absence of the a pain-related tissue status or disease, in presence of a pain-related altered tissue status or disease or for an increased or decreased risk of developing a pain-related altered tissue status or disease.
According to another embodiment of the fourth aspect of present invention, the reference sample is selected from the group consisting of a reference sample derived from an not having a pain-related tissue status or disease individual, a reference sample from an individual having a pain-related tissue status or disease, and a reference sample from the same individual as the sample to be tested taken at an earlier or later time point or a reference sample with a level or amount of slfn2 representative for an individual having or not having a pain-related tissue status or disease or for the absence or presence of the pain-related disease or altered tissue status or representative for an increased or decreased risk of developing a pain-related tissue status or disease.
According to another embodiment of the fourth aspect of present invention, the reference is an individual not having a pain-related tissue status or disease or an individual with a decreased risk of developing a pain-related tissue status or disease or a value representative of the absence of a pain-related tissue status or disease, or the Slfn2 or method according to embodiment 8, wherein the reference sample is derived from an individual not having a pain-related tissue status or disease or from an individual with a decreased risk of developing a pain-related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual not having a pain-related tissue status or disease or for a status of disease-absence or for a decreased risk of developing a pain-related tissue status or disease, wherein an elevated level of Slfn2 in the tested sample indicates
a. the presence of a pain-related tissue status or disease and/or b. an increased risk of developing a pain-related tissue status or disease and/or
c. the progression of a pain-related tissue status or disease
in the individual. According to another embodiment of the fourth aspect of present invention, the reference is an individual having a pain-related tissue status or disease or an individual with an increased risk of developing a pain-related tissue status or a disease or a value representative of the presence of a pain-related tissue status or disease, or Slfn2 or method according to embodiment 8, wherein the reference sample is derived from an individual having a pain-related tissue status or disease or from an individual with an increased risk of developing a pain-related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual having a pain-related tissue status or disease or for a status of disease-presence or for an increased risk of developing a pain-related tissue status or disease, wherein a similar or higher level of Slfn2 in the tested sample indicates
a. the presence of the pain-related tissue status or disease and/or b. an increased risk of developing a pain-related tissue status or disease and/or
c. the progression of a pain-related tissue status or disease
in the individual.
According to another embodiment of the fourth aspect of present invention, the reference is the same individual at an earlier time point or Slfn2 or method of embodiment 8, wherein the sample is derived from the same individual as the tested sample and was taken at an earlier time point, wherein
(a) an elevated level of Slfn2 in the tested sample indicates
(vii) the presence of a pain-related tissue status or disease and/or
(viii) an increased risk of developing a pain-related tissue status or disease and/or
(ix) the progression of the pain-related tissue status or disease
(b) a lowered level of Slfn2 in the tested sample indicates
(vii) an improvement or the absence of a pain-related tissue status or an improvement or absence of a pain-related disease and/or
(viii) a decreased risk to develop a pain-related tissue status or disease and/or
(ix) a declined progression of the pain-related tissue status or disease
(c) a similar level of Slfn2 in the tested sample indicates (iv) a similar or persistent risk to develop a pain-related tissue status or disease and/or
(v) a stagnation in the progression of a pain-related tissue status or disease and/or
(vi) the persistence of the a pain-related tissue status or disease in the individual.
According to another embodiment of the fourth aspect of present invention, the tested sample is tissue and/or body fluid, wherein the tissue sample can e.g. be extraneural or neural tissue or an extraneural or a neural cell, and wherein the body fluid sample can e.g. be cerebrospinal fluid.
According to another embodiment of the fourth aspect of present invention, the pain- related disease is pain or a pain-associated disease, preferably a pain such as defined above and below.
According to another embodiment of the fourth aspect of present invention, Slfn2 is a Slfn2 gene or gene product, preferably a Slfn2 mRNA, or Slfn2 protein or a functionally active variant thereof.
According to another embodiment of the fourth aspect of present invention, Slfn2 is a gene, wherein the gene
a. is a naturally occurring or non-naturally occurring Slfn2 gene variant, or
b. codes for a Slfn2 protein or a functionally active variant thereof that has
i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
iii. an amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
iv. an amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to
SEQ ID NO:2, and/or
v. an amino acid sequence with at least 50% identity with the amino acid sequence according to SEQ ID NO:2 and/or vi. an amino acid sequence with one of the following sequence motifs: YxxlxV, YxxlxVxxW, YxxlxVxxWS or YxxlxVxxWSxS with "x" standing for any amino acid and/or
vii. carries a variant AAA domain, preferably with 50, 60, 70, 80, 90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2.
According to another embodiment of the fourth aspect of present invention, Slfn2 is a gene product, wherein the gene product
a) is a naturally occurring Slfn2 protein variant, preferably a homolog or
ortholog in same or different species, or a non-naturally occurring Slfn2 protein variant,
b) is a Slfn2 protein or a functionally active variant thereof having
i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
iii. an amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
iv. an amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to SEQ ID NO:2, and/or
v. an amino acid sequence with at least 50% identity with the amino acid sequence according to SEQ ID NO:2 and/or vi. an amino acid sequence with one of the following sequence motifs: YxxlxV, YxxlxVxxW, YxxlxVxxWS or YxxlxVxxWSxS with "x" standing for any amino acid and/or vii. carries a variant AAA domain, preferably with 50, 60, 70, 80,
90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2, c) is a Slfn2 nucleic acid encoding a protein as defined in b), or
d) is a Slfn2 nucleic acid having
(i) a nucleic acid sequence according to SEQ ID NO: 1 , 6 or 8,
(ii) a nucleic acid sequence that is a fragment of the nucleic acid
sequence according to (i), or
(iii) a nucleic acid sequence that has at least 80%, at least 90%, at least
95% or at least 99% sequence identity to the nucleic acid sequence according to (i) or (ii).
The fourth aspect and its different embodiments are especially suitable for diagnosing and/or prognosing pain in an in vitro or in vivo test model (i.e. a test animal, such as a non-human test animal, e.g. in the context of clinical studies) or in a patient (such as a human or non-human animal).
In another embodiment, the fourth aspect of present invention comprises determining the expression level of Slfn2 and preferably comprises determining the expression level of Slfn2 on the gene, mRNA or protein level.
According to another embodiment of the fourth aspect of present invention, the tissue status is a pain-associated tissue status, such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue, the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
According to another embodiment of the fourth aspect of present invention, the pain- related or pain-associated disease is a pain.
According to another embodiment of the fourth aspect of present invention, the pain is (i) acute pain, subacute pain, or chronic pain,
(ii) nociceptive pain, neuropathic pain, inflammatory pain, or a mixture thereof,
(iii) disease-associated pain, or
(iv) algesia.
According to another embodiment of the fourth aspect of present invention
(i) the acute pain is somatogenetic (organic) pain,
(ii) the chronic pain is somatogenetic (organic) pain or psychogenic
(psychosomatic) pain,
(iii) the nociceptive pain is superficial somatic pain or deep pain, preferably deep somatic pain or visceral pain,
(iv) the neuropathic pain is central neuropathic pain or peripheral neuropathic pain,
(v) the mixed pain is a mixture of nociceptive pain and neuropathic pain,
(vi) the disease-associated pain is pain associated with depression, migraine, epilepsy, cancer, or diabetes, or
(vii) the algesia is hyperalgesia.
According to another embodiment of the fourth aspect of present invention, the disease is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
According to another embodiment of the fourth aspect of present invention, the method relates to diagnosis, wherein the diagnosis comprises
(i) determining the presence or occurrence of pain,
(ii) monitoring the course of pain,
(iii) staging of pain,
(iv) classification of a subject with pain.
According to another embodiment of the fourth aspect of present invention, the method relates to prognosis, wherein the prognosis comprises predicting or estimating the occurrence or reoccurrence, preferably the severity of occurrence or reoccurrence, of pain. In a fifth aspect, present invention relates to a kit for use in a method according to the fourth aspect and any of its embodiments, wherein the kit comprises one or more means of detecting Slfn2.
In a sixth aspect, present invention relates to a kit for detecting Slfn2 comprising
(i) a means for detecting Slfn2 and/or
(ii) a data carrier comprising instructions for a method according to the fourth aspect of present invention and any one its embodiments and/or
(iii) a container.
A kit is an article of manufacture that comprises at least the components as defined above and optionally one or more further components, the term kit in the context of present invention is understood as herein defined.
The kit according to the fifth and sixth aspect of present invention comprises at least one or more means for detecting Slfn2, it can in addition, comprise one or more means for detecting another indicator of a pain-related tissue status or a pain-related disease such as pain, e.g. as listed above.
According to one embodiment of the fifth and sixth aspect, the kit comprises a means for detecting Slfn2 or a means for detecting Slfn2 and a data carrier comprising instructions for a method according to the fourth aspect of present invention and any one its embodiments, it can also comprise a means for detecting Slfn2 and a container, it can also comprise a means for detecting Slfn2 and a means for detecting another indicator of a pain-related tissue status or pain-related disease such as pain, it can also comprise a means for detecting Slfn2 and a data carrier comprising instructions for a method according to the fourth aspect of present invention and any one its embodiments and a container it can also comprise a means for detecting Slfn2 and a data carrier comprising instructions for a method according to the fourth aspect of present invention and any one its embodiments and another indicator of a pain-related tissue status or pain-related disease such as pain, it can also comprise a means for detecting Slfn2 and another indicator of a pain-related tissue status or pain-related disease such as pain and a container. According to one embodiment of the fifth and sixth aspect of present invention, the means for detecting Slfn2 is a means for determining the expression level of Slfn2, preferably on the gene, protein or RNA level. The term means of detecting Slfn2 or means of detection of Slfn2 is understood as herein defined.
According to another embodiment of the fifth and sixth aspect of present invention, the means for detecting Slfn2 is selected from the group consisting of nucleic acid, preferably DNA, RNA or PNA, peptide and protein, preferably monoclonal or polyclonal antibody or protein scaffold. The kit according to the fifth and sixth aspect of present invention can also comprise more than one means, e.g. 1 , 2, 3, 4, 5, 6, 7, 8, 9, or 10 means of detection.
According to another embodiment of the fifth and sixth aspect of present invention, the kit comprises
(a) a container, and/or
(b) a data carrier, wherein the data carrier comprises information such as
(1 ) instructions concerning methods for identifying the risk for developing
and/or identifying the presence and/or monitoring progression of a pain- related tissue status or disease
(2) instructions for use of the means for detecting Slfn2, preferably in a sample, more preferably in a sample from an individual and/or of the kit,
(3) quality information such as information about the lot/batch number of the means for detecting Slfn2 and/or of the kit, the manufacturing or assembly site or the expiry or sell-by date, information concerning the correct storage or handling of the kit,
(4) information concerning the composition of the buffer(s), diluent(s), reagent(s) for detecting Slfn2 and/or of the means for detecting Slfn2,
(5) information concerning the interpretation of information obtained when performing the above-mentioned methods identifying and/or monitoring progression of a pain-related tissue status or disease,
(6) a warning concerning possible misinterpretations or wrong results when applying unsuitable methods and/or unsuitable means, and/or (7) a warning concerning possible misinterpretations or wrong results when using unsuitable reagent(s) and/or buffer(s).
The data carrier can thus comprise information according to (1 ), (2), (3), (4), (5), (6) and/or (7). This means that the data carrier comprises at least information according to (1 ) or information according to (2) or information according to (3) or information according to (4) or information according to (5) or information according to (6) or information according to (7); the data carrier can also comprise (1 ) and (2) or (1 ) and (3) or (1 ) and (4) or (1 ) and (5) or (1 ) and (6) or (1 ) and (7) or (2) and (3) or (2) and (6) or (2) and (5) or (2) and (6) or (2) and (7) or (3) and (4) or (3) and (5) or (3) and (6) or (3) and (7) or (6) and (5) or (4) and (6) or (4) and (7) or (5) and (6) or (5) and (7) or (6) and (7); the data carrier can also comprise (1 ) and (2) and (3) or (1 ) and (2) and (4) or (1 ) and (2) and (5) or (1 ) and (2) and (6) or (1 ) and (2) and (7) or (1 ) and (3) and (4) or (1 ) and (3) and (5) or (1 ) and (3) and (6) or (1 ) and (3) and (7) or (1 ) and (4) and (5) or (1 ) and (4) and (6) or (1 ) and (4) and (7) or (1 ) and (5) and (6) or (1 ) and (5) and (7) or (1 ) and (6) and (7) or (2) and (3) and (4) or (2) and (3) and (5) or (2) and (3) and (6) or (2) and (3) and (7) or (2) and (4) and (5) or (2) and (4) and (6) or (2) and (4) and (7) or (2) and (5) and (6) or (2) and (5) and (7) or or (2) and (6) and (7) or (3) and (4) and (5) or (3) and (4) and (6) or (3) and (4) and (7) or (4) and (5) and (6) or (4) and (5) and (7) or (5) and (6) and (7); the data carrier can also comprise (1 ) and (2) and (3) and (4) or (1 ) and (2) and (3) and (5) or (1 ) and (2) and (3) and (6) or (1 ) and (2) and (3) and (7) or (2) and (3) and (4) and (5) or (2) and (3) and (4) and (6) or (2) and (3) and (4) and (7) or (3) and (4) and (5) and (6) or (3) and (4) and (5) and (7) or (4) and (5) and (6) and (7); the data carrier can also comprise (1 ) and (2) and (3) and (4) and (5) or (1 ) and (2) and (3) and (4) and (6) or (1 ) and (2) and (3) and (4) and (7) or (2) and (3) and (4) and (5) and (6) or (2) and (3) and (4) and (5) and (7) or (3) and (4) and (5) and (6) and (7). (insert commas to delineate groups)
According to one embodiment of the fifth and sixth aspect of present invention the kit is for diagnosing and/or prognosing pain by determination of Slfn2 and preferably determining the level of Slfn2 in a sample. The sample can be any suitable sample, e.g such as defined herein, e.g. in the definitions or above under the first and second aspect of present invention. According to another embodiment of the fifth and sixth aspect of present invention the kit comprises a means as defined below for the eighth aspect of present invention.
According to aseventh aspect, present invention refers to a means of detecting Slfn2 for use in the diagnosis or prognosis of a tissue status or a disease.
According to another embodiment of the seventh aspect of present invention, the means comprises one or more nucleic acids or derivatives for detecting a Slfn2 gene or gene product or a functionally active variant thereof, wherein the nucleic acid or derivative is preferably selected from the group consisting of a nucleic acid probe, a polyamide or peptide nucleic acid (PNA), microRNA (miRNA), small interfering RNA (siRNA), primers for polymerase chain reaction (PCR), primers for reverse transcription (RT) reaction, and primers for DNA sequencing. According to another embodiment of the seventh aspect of present invention the means comprises a peptide, polypeptide or protein for detecting a Slfn2 gene or gene product or functionally active variant thereof, wherein the protein or polypeptide is preferably a protein ligand (such as an antibody) a fragment or derivate thereof, a protein scaffold (such as a darpin or an anticalin) or wherein the polypeptide or peptide is a probe, preferably a mass spectrometry probe.
According to another embodiment of the seventh aspect of present invention, the tissue status is a pain-associated tissue status, such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue, the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
According to another embodiment of the seventh aspect of present invention, the disease is a pain wherein the pain is preferably
(1 ) acute pain, subacute pain, or chronic pain,
(ii) nociceptive pain, neuropathic pain, inflammatory pain, or a mixture thereof, (iii) disease-associated pain, or
(iv) algesia.
According to another embodiment of the seventh aspect of present invention
(i) the acute pain is somatogenetic (organic) pain,
(ii) the chronic pain is somatogenetic (organic) pain or psychogenic
(psychosomatic) pain,
(iii) the nociceptive pain is superficial somatic pain or deep pain, preferably deep somatic pain or visceral pain,
(iv) the neuropathic pain is central neuropathic pain or peripheral neuropathic pain,
(v) the mixed pain is a mixture of nociceptive pain and neuropathic pain,
(vi) the disease-associated pain is pain associated with depression, migraine, epilepsy, cancer, or diabetes, or
(vii) the algesia is hyperalgesia.
According to another embodiment of the seventh aspect of present invention, the pain- associated or pain-related disease is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
According to another embodiment of the seventh aspect of present invention, the means is for use in the diagnosis or prognosis or a pain-related tissue status or a pain- related disease, preferably in a method of the third aspect of present invention.
According to yet another preferred embodiment of the seventh aspect of present invention, the diagnosis comprises
(i) determining the presence or occurrence of pain,
(ii) monitoring the course of pain,
(iii) staging of pain,
(iv) classification of a subject with pain. According to yet another preferred embodiment of the seventh aspect of present invention, the prognosis comprises predicting or estimating the occurrence or reoccurrence, preferably the severity of occurrence or reoccurrence, of pain. In the following, preferred embodiments of present invention are listed but are not to be understood as limiting present invention:
1 . Slfn2 for use as an indicator of a pain-related tissue status or a pain related disease or as an indicator of the risk for developing a pain-related tissue status or disease.
2. Slfn2 for use in the diagnosis or prognosis of a pain-related tissue status or of a pain- related disease.
3. Slfn2 according to embodiment 1 or 2, wherein the level of Slfn2 is indicative of
(iv) a pain-related tissue status or the presence of a pain-related disease and/or
(v) the risk of developing a pain-related tissue status or a pain-related disease and/or
(vi)the progression or a stage of a pain-related tissue status or a pain-related disease
in an individual.
4. A method of identifying
(v) a pain-related tissue status or the presence of a pain-related disease and/or
(vi) the risk of developing a pain-related tissue status or a pain-related disease and/or
(vii) the progression or a stage of a pain-related tissue status or a pain- related disease
in an individual, the method comprising detecting the level of Slfn2. 5. Slfn2 according to embodiment 3 or the method according embodiment 5, wherein the level of Slfn2 is the level of Slfn2 in an individual or in a sample of an individual.
6. Slfn2 or method according embodiment 5, further comprising comparing the level of Slfn2 to the level of Slfn2 in one or more references or reference samples.
7. Slfn2 or method according to embodiment 6, wherein the reference is selected from the group consisting of an individual not having a pain-related tissue status or disease, an individual having a pain-related tissue status or disease or the same individual as the individual to be tested at an earlier or later time point or a value representative for the level of slfn2 in absence of the a pain-related tissue status or disease, in presence of a pain-related altered tissue status or disease or for an increased or decreased risk of developing a pain-related altered tissue status or disease. 8. Slfn2 or method of embodiment 6, wherein the reference sample is selected from the group consisting of a reference sample derived from an not having a pain-related tissue status or disease individual, a reference sample from an individual having a pain-related tissue status or disease, and a reference sample from the same individual as the sample to be tested taken at an earlier or later time point or a reference sample with a level or amount of slfn2 representative for an individual having or not having a pain- related tissue status or disease or for the absence or presence of the pain-related disease or altered tissue status or representative for an increased or decreased risk of developing a pain-related tissue status or disease. 9. Slfn2 or method of embodiment 7 wherein the reference is an individual not having a pain-related tissue status or disease or an individual with a decreased risk of developing a pain-related tissue status or disease or a value representative of the absence of a pain-related tissue status or disease, or the Slfn2 or method according to embodiment
8. wherein the reference sample is derived from an individual not having a pain-related tissue status or disease or from an individual with a decreased risk of developing a pain- related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual not having a pain-related tissue status or disease or for a status of disease-absence or for a decreased risk of developing a pain-related tissue status or disease, wherein an elevated level of Slfn2 in the tested sample indicates
a. the presence of a pain-related tissue status or disease and/or b. an increased risk of developing a pain-related tissue status or disease and/or
c. the progression of a pain-related tissue status or disease
in the individual.
10. Slfn2 or method of embodiment 7, wherein the reference is an individual having a pain-related tissue status or disease or an individual with an increased risk of developing a pain-related tissue status or a disease or a value representative of the presence of a pain-related tissue status or disease, or Slfn2 or method according to embodiment 8, wherein the reference sample is derived from an individual having a pain-related tissue status or disease or from an individual with an increased risk of developing a pain-related tissue status or disease or wherein the reference sample comprises a level or amount of Slfn2 representative for an individual having a pain- related tissue status or disease or for a status of disease-presence or for an increased risk of developing a pain-related tissue status or disease, wherein a similar or higher level of Slfn2 in the tested sample indicates
d. the presence of the pain-related tissue status or disease and/or e. an increased risk of developing a pain-related tissue status or disease and/or
f. the progression of a pain-related tissue status or disease
in the individual.
1 1 . Slfn2 or method of embodiment 7, wherein the reference is the same individual at an earlier time point or Slfn2 or method of embodiment 8, wherein the sample is derived from the same individual as the tested sample and was taken at an earlier time point, wherein
(a) an elevated level of Slfn2 in the tested sample indicates
(x) the presence of a pain-related tissue status or disease and/or
(xi) an increased risk of developing a pain-related tissue status or disease and/or
(xii) the progression of the pain-related tissue status or disease (b) a lowered level of Slfn2 in the tested sample indicates
(x) an improvement or the absence of a pain-related tissue status or an improvement or absence of a pain-related disease and/or
(xi) a decreased risk to develop a pain-related tissue status or disease and/or
(xii) a declined progression of the pain-related tissue status or disease
(c) a similar level of Slfn2 in the tested sample indicates
(vii) a similar or persistent risk to develop a pain-related tissue status or disease and/or
(viii) a stagnation in the progression of a pain-related tissue status or disease and/or
(ix) the persistence of the a pain-related tissue status or disease in the individual.
12. The Slfn2 or the method of any of embodiments 8 - 12, wherein the tested sample tissue and/or body fluid.
13. The Slfn2 or the method of embodiment 12, wherein the tissue sample is extraneural or neural tissue or an extraneural or a neural cell, and wherein the body fluid sample is cerebrospinal fluid.
14. The Slfn2 according to one of embodiments 1 to 3 or 5-13 or the method according to one of the embodiments 4-13 , wherein the pain-related disease is pain or a pain- associated disease, preferably a pain according to one of the embodiments 34-36.
15. The Slfn2 according to one of the embodiments 1 -3 or 5-14 or the method according to one of the embodiments 4-14, wherein Slfn2 is a Slfn2 gene or gene product, preferably a Slfn2 mRNA, or Slfn2 protein or a functionally active variant thereof. 16. The Slfn2 or method according to embodiment 15, wherein Slfn2 is a gene and wherein the gene
a. is a naturally occurring or non-naturally occurring Slfn2 gene variant, or b. codes for a Slfn2 protein or a functionally active variant thereof that has
i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
iii. an amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or
iv. an amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to SEQ ID NO:2, and/or
v. an amino acid sequence with at least 50% identity with the amino acid sequence according to SEQ ID NO:2 and/or vi. an amino acid sequence with one of the following sequence motifs: YxxlxV, YxxlxVxxW, YxxlxVxxWS or YxxlxVxxWSxS with "x" standing for any amino acid and/or
vii. carries a variant AAA domain, preferably with 50, 60, 70, 80, 90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2.
17. The Slfn2 or method according to embodiment 16, wherein Slfn2 is a gene product and wherein the gene product
a) is a naturally occurring Slfn2 protein variant, preferably a homolog or
ortholog in same or different species, or a non-naturally occurring Slfn2 protein variant,
b) is a Slfn2 protein or a functionally active variant thereof having
i. an amino acid sequence according to SEQ ID NO: 2, 7 or 9, ii. an amino acid sequence that is a fragment of the amino acid sequence according to (i) and/ or
iii. an amino acid sequence that has at least 80%, at least 90%, at least 95% or at least 99% sequence identity to the amino acid sequence according to (i) or (ii) and/or iv. an amino acid sequence with at least 95% or at least 99% or 100% identity to at least 10, 1 1 , 12, 13, 14 or 15 consecutive amino acids of the Slfn2 protein according to SEQ ID NO:2, and/or
v. an amino acid sequence with at least 50% identity with the amino acid sequence according to SEQ ID NO:2 and/or vi. an amino acid sequence with one of the following sequence motifs: YxxlxV, YxxlxVxxW, YxxlxVxxWS or YxxlxVxxWSxS with "x" standing for any amino acid and/or
vii. carries a variant AAA domain, preferably with 50, 60, 70, 80,
90, 95 or more sequence identity with the variant AAA domain of the Slfn2 protein according to SEQ ID NO:2, c) is a Slfn2 nucleic acid encoding a protein as defined in b), or
d) is a Slfn2 nucleic acid having
(i) a nucleic acid sequence according to SEQ ID NO: 1 , 6 or 8,
(ii) a nucleic acid sequence that is a fragment of the nucleic acid
sequence according to (i), or
(iii) a nucleic acid sequence that has at least 80%, at least 90%, at least
95% or at least 99% sequence identity to the nucleic acid sequence according to (i) or (ii).
18. The Slfn2 according to one of the embodiments 1 -3 or 5-17 or the method according to one of the embodiments 5-17 for diagnosing and/or prognosing pain in an in vitro or in vivo test model.
19. The Slfn2 according to one of the embodiments 1 -3 or 5-18 or the method according to one of the embodiments 4-18 comprising determining the expression level of Slfn2 and preferably comprising determining the expression level of Slfn2 on the gene, mRNA or protein level.
20. A kit for use in a method according to any of embodiments 4 to 19, comprising one or more means of detecting Slfn2.
21 . A kit for detecting Slfn2 comprising (i) a means for detecting Slfn2,
(ii) a data carrier comprising instructions for a method according to one of the embodiments 4-19 and optionally
(iii) a container.
22. The kit according to embodiment 20 or 21 wherein the means for detecting Slfn2 is a means for determining the expression level of Slfn2, preferably on the gene, protein or RNA level. 23. The kit according to one of the embodiments 20 -22, wherein the means for detecting Slfn2 is selected from the group consisting of nucleic acid, preferably DNA, RNA or PNA, peptide and protein, preferably monoclonal or polyclonal antibody or protein scaffold.
24. The kit according to any of embodiments 20-23, wherein the kit comprises
(a) a container, and/or
(b) a data carrier, wherein the data carrier comprises information such as
(i) instructions concerning methods for identifying the risk for developing
and/or identifying the presence and/or monitoring progression of a pain- related tissue status or disease
(ii) instructions for use of the means for detecting Slfn2, preferably in a sample, more preferably in a sample from an individual and/or of the kit,
(iii) quality information such as information about the lot/batch number of the means for detecting Slfn2 and/or of the kit, the manufacturing or assembly site or the expiry or sell-by date, information concerning the correct storage or handling of the kit,
(iv) information concerning the composition of the buffer(s), diluent(s), reagent(s) for detecting Slfn2 and/or of the means for detecting Slfn2,
(v) information concerning the interpretation of information obtained when performing the above-mentioned methods identifying and/or monitoring progression of a pain-related tissue status or disease,
(vi) a warning concerning possible misinterpretations or wrong results when applying unsuitable methods and/or unsuitable means, and/or (vii) a warning concerning possible misinterpretations or wrong results when using unsuitable reagent(s) and/or buffer(s).
26. The kit according to any of embodiments 20-24, wherein the kit is for diagnosing and/or prognosing pain by determination of Slfn2 and preferably the level of Slfn2 in a sample.
27. The kit according to any of embodiments 20-25, wherein the kit comprises a means according to any of the embodiments 27-39.
28. A means of detecting Slfn2 for use in the diagnosis or prognosis of a tissue status or a disease.
29. The means according to embodiment 28 comprising one or more nucleic acids or derivatives for detecting a Slfn2 gene or gene product or functionally active variant thereof.
30. The means according to embodiment 28, wherein the nucleic acid or derivative is selected from the group consisting of a nucleic acid probe, a polyamide or peptide nucleic acid (PNA), microRNA (miRNA), small interfering RNA (siRNA), primers for polymerase chain reaction (PCR), primers for reverse transcription (RT) reaction, and primers for DNA sequencing.
31 . The means according to embodiment 28 comprising a peptide, polypeptide or protein for detecting a Slfn2 gene or gene product or functionally active variant thereof.
32. The means according to embodiment 31 , wherein the protein or polypeptide is a protein ligand, preferably an antibody, a fragment or derivate thereof, a protein scaffold, such as a darpin or an anticalin, or wherein the polypeptide or peptide is a probe, preferably a mass spectrometry probe.
33. The means according to any of embodiments 28-32, wherein said means comprises
(i) a biochip, or
(ii) a set of beads. 34. The means according to one of embodiments 28-33, wherein the diagnosis comprises one of the methods according to embodiments 4-19. 35. Slfn2 according to any of embodiments 1 -3 or 5-19 or method according to any of embodiments 4 to 19, kit according to any of embodiments 20-27 or means according to any of embodiments 28-34, wherein the tissue status is a pain-associated tissue status, such as a tissue damage, trauma, inflammation, infarction, necrosis, oedema, neoplasia of any tissue, the tissue preferably being selected from the group consisting of bone, cartilage, muscle, skin, neural tissue, such as a nerve, connective tissue such as perichondrium, periosteum, tendon, capsule, skin and and/or wherein the tissue status is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue. 36. The Slfn2 according to any of embodiments 1 -3 or 5-19 or method according to any of embodiments 4 to 19, kit according to any of embodiments 20-27 or means according to any of embodiments 28-35, wherein the disease is a pain.
37. The Slfn2, method, kit or means according to embodiment 36, wherein the pain is (i) acute pain, subacute pain, or chronic pain,
(ϋ) nociceptive pain, neuropathic pain, inflammatory pain, or a mixture thereof, (iii) disease-associated pain, or
(iv) algesia.
38 The Slfn2, method, kit or means according to embodiment 37, wherein
(i) the acute pain is somatogenetic (organic) pain,
(ii) the chronic pain is somatogenetic (organic) pain or psychogenic
(psychosomatic) pain,
(iii) the nociceptive pain is superficial somatic pain or deep pain, preferably deep somatic pain or visceral pain,
the neuropathic pain is central neuropathic pain or peripheral neuropath pain
(v) the mixed pain is a mixture of nociceptive pain and neuropathic pain (vi) the disease-associated pain is pain associated with depression, migraine, epilepsy, cancer, or diabetes, or
(vii) the algesia is hyperalgesia. 39. The Slfn2 according to any of embodiments 1 -3 or 5-19 or method according to any of embodiments 4 -19, kit according to any of embodiments 20-27 or means according to any of embodiments 28-33, wherein the disease is characterized by an altered level of pain-related regulatory molecules or signaling molecules, indicators of pain and/or degenerative molecules present in the tissue.
40. The Slfn2 according to any of embodiments 1 -3, 5-19 or 35-39, method according to any of embodiments 4 -19 or 35-39, kit according to any of embodiments 20-27 or 35-39 or means according to any of embodiments 28-39, wherein diagnosis comprises
(i) determining the presence or occurrence of pain,
(ii) monitoring the course of pain,
(iii) staging of pain,
(iv) classification of a subject with pain.
41 . The Slfn2 according to any of embodiments 1 -3, 5-19 or 35-39, method according to any of embodiments 4 -19 or 35-39, kit according to any of embodiments 20-27 or 35-39 or means according to any of embodiments 28-39, wherein the prognosis comprises predicting or estimating the occurrence or reoccurrence, preferably the severity of occurrence or reoccurrence, of pain.
LITERATURE:
Schwarz, D.A., Katayama, CD. and Hedrick, S.M., Immunity Vol.9 (1998), p.657-668, Schlafen, a New Family of Growth Regulatory Genes that Affect Thymocyte
Development.
Wern-Joo Sohn, et al., Molecular Immunology 44 (2007), 3273-3282, Novel
transcriptional regulation of the schlafen-2 gene in macrophages in response to TLR- triggered stimulation.
Katsoulidis, E., et al., Journal of Biological Chemistry Vol.284, Nr. 37 (2009), p.25051 - 25064, Role of Schlafen 2 (SLFN2) in the Generation of Interferon alpha-induced Growth Inhibitory Responses.
Horton, M.R. and Powell, J.D., Nature Immunology Vol. 1 1 , Nr.4 (2010), p.281 -282, Quieting T cells with Slfn2.
Berger, M. et al., Nature Immunology Vol.1 1 , Nr.4 (2010), p.335-343, A Slfn2 mutation causes lymphoid and myeloid immunodeficiency due to loss of immune cell quiescence.
Bourquin AF et al (2006) Assessment and analysis of mechanical allodynia-like behavior induced by spared nerve injury (SNI) in the mouse. Pain 122:p.1 -14. Storey JD. (2002) A direct approach to false discovery rates. Journal of the Royal Statistical Society, Series B, 64: 479-498.
If not indicated otherwise, standard laboratory methods in the context of present invention were or can be performed according to the following standard literature:
Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual. Second edition. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. 545 pp. Current Protocols in Molecular Biology; regularly updated, e.g. Volume 2000; Wiley & Sons, Inc; Editors: Fred M. Ausubel, Roger Brent, Robert Eg. Kingston, David D. Moore, J.G. Seidman, John A. Smith, Kevin Struhl. Current Protocols in Human Genetics; regularly updated; Wiley & Sons, Inc; Editors: Nicholas C. Dracopoli, Honathan L. Haines, Bruce R. Korf, Cynthia C. Morton, Christine E. Seidman, J.G. Seigman, Douglas R. Smith.
Current Protocols in Protein Science; regularly updated; Wiley & Sons, Inc; Editors: John E. Coligan, Ben M. Dunn, Hidde L. Ploegh, David W. Speicher, Paul T. Wingfield.
Molecular Biology of the Cell; third edition; Alberts, B., Bray, D., Lewis, J., Raff, M., Roberts, K., Watson, J.D.; Garland Publishing, Inc. New York & London, 1994; Short Protocols in Molecular Biology, 5th edition, by Frederick M. Ansubel (Editor), Roger Brent (Editor), Robert E. Kingston (Editor), David D. Moore (Editor), J.G.
Seidman (Editor), John A. Smith (Editor), Kevin Struhl (Editor), October 2002, John Wiley & Sons, Inc., New York". Transgenic Animal Technology A Laboratory Handboook. C.A. Pinkert, editor;
Academic Press Inc., San Diego, California, 1994 (ISBN: 0125571658).
Gene targeting: A Practical Approach, 2nd Ed., Joyner AL, ed. 2000. IRL Press at Oxford University Press, New York.
Manipulating the Mouse Embryo: A Laboratory Manual. Nagy, A, Gertsenstein, M., Vintersten, K., Behringer, R., 2003, Cold Spring Harbor Press, New York.
Remington's Pharmaceutical Sciences, 17th Edition, 1985 (for physiologically tolerable salts (anorganic or organic), see esp. p. 1418). LEGEND TO THE FIGURES
Figure 1 : ClustalW 2.1 (available by EMBL-EBI (EMBL-European Bioinformatics
Institute)) sequence alignment of murine, rat and homo sapiens Slfn2/12L amino acid sequences: sequence 1 is murine (mus musculus) Slfn2 according to
AF_099973.1/AAC838261 (SEQ ID NO:2), sequence 2 is rat (rattus norwegicus) Slfn2 according to NP_001 100501/NM_001 107031 .1 (SEQ ID NO:7), sequence 3 is homo sapiens SLFN12L according to NM_001 195790.1/NP_001 182719.1 (SEQ ID NO:9). The identity score with ClustalW is 83% between mSlfn2 and ratSlfn2, 50% between mSlfn2 and hsSLFN12L and 50% between rat Slfn2 and hsSLFN12L. The conserved variant AAA domain is shown underlined. The isoleucine at position 135 of murine Slfn2 (corresponding to position 135 in rat Slfn2 and position 123 in hsSLFN12L), the mutation of which gives rise to the "electra" phenotype in mice (see description for details) is marked bolt and underlined. As can be gained from the sequence comparison, this position is conserved among the three different species hinting to an equal importance of said amino acid or surrounding protein region within the respective rat and homo sapiens proteins. Figure 2: Sequence alignment of murine Slfn2 (SEQ ID NO:2, sequence 1 of
comparison) and rat Slfn2 (SEQ ID NO:7, sequence 2 of comparison) using the
EMBOSS needle 6.3.1 program (working on Needleman-Wunsch algorithm) of the EMBL EBI database . The divergent AAA domains (from position 231 to 355 of murine Slfn2 and from position 231 to 361 of rat Slfn2) as outlined in the NCBI database (see respective accession number) have been marked underlined.
Figure 3: Sequence alignment of murine Slfn2 (SEQ ID NO:2, sequence 1 of
comparison) and homo sapiens SLFN12L (SEQ ID NO:9, sequence 2 of comparison) using the EMBOSS needle 6.3.1 program (working on Needleman-Wunsch algorithm) of the EMBL EBI database. The divergent AAA domains (from position 231 to 355 of murine Slfn2 and from position 218 to position 349 for hsSLFN12L) as outlined in the NCBI database (see respective accession number) have been marked underlined.
Figure 4: Promoter structure of murine Slfn2 according to Wern-Joo et al, 2009. Figure 5: Figure 5 shows for every individual mouse its neuropathic pain phenotype (mechanical hypersensitivity, X-axis) and the corresponding gene expression (signal intensity, Y-axis) in the L5 DRG. Mouse data are colour-coded depending on the used strain. A Pearson correlation analysis has been performed and revealed a significant positive correlation of the two parameters pain phenotype and Slfn2 gene expression. This means for individual mice that the higher the L5 DRG expression of Slfn2 in Chung-operated neuropathic mice was, the more pronounced the mechanical hyperalgesia as exhibited in the behavioral test.
This significant correlation indicates a causal relationship of Slfn2 gene expression for the induction of the neuropathic pain phenotype.
Figure 6: shows exemplary intensity data for Slfn2 of L5 DRG (3d p.o.).
EXAMPLES
The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the methods and compositions of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers used but some experimental errors and deviations should be accounted for. Unless indicated otherwise, molecular weight is average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric. Identification of Slfn2 as protein involved in algesia.
Example 1
In order to identify new targets for pain therapy, a correlational analysis for identifying genes whose regulation contributes to chronic neuropathic pain was carried out (see also Persson et al., 2009, Molecular Pain 5:7). In summary, RNA samples of dorsal root ganglia (DRGs) of inbred mouse strains AKR/J (AKR), C57BL/6J (C57/B6) and CBA J (CBA) were examined. Inbred mouse strains obtained from The Jackson Laboratory (Bar Harbor, ME, USA). The spinal nerve at position L5 of Chung-operated (Chung model of neuropathic pain (Kim and Chung, 1992, Pain 50: 355-363) and of corresponding sham-operated control animals were subjected to axotomy.
Surgical nerve lesion and sham surgical procedures:
In anesthetized mice, the left sciatic nerve was exposed at its trifurcation and the two major branches (tibial and common peroneal nerves) were ligated and transected distal to the ligation. Sham surgery was identical, the nerves were exposed but not ligated and transected (see Bourquin et al. 2006).
Manifestation of pain phenotype:
The manifestation of the pain phenotype "mechanic hyperalgesia" was determined by means of the paw withdrawal threshold at all mice before removal of DRGs (Persson et al., supra, particularly section "Behavioral testing"). The three mouse strains differ in their phenotypes. In CBA mice, C57/B6 mice and AKR mice, the phenotype is manifested at a low, middle and high level, respectively. Determination of paw withdrawal threshold:
Paw withdrawal thresholds (PWTs) were assessed using a dynamic plantar aesthesiometer (see Szabo et al. 2005). After acclimation in a compartment with metal mesh floor, the stimulator was positioned under the animal's hindpaw, a straight metal filament driven by an electrodynamic actuator touched the plantar surface and exerted an increasing upward force until the animal removed the paw (paw withdrawal threshold, PWT). PWTs were assessed for hindpaws of the ipsilateral, operated side and of the contralateral side. RNA isolation from dorsal root ganglia:
In order to carry out gene expression experiments, a method for isolating total RNA of murine DRGs was developed (Persson et al., supra, particularly section "RNA extraction for TaqMan and microarray analysis"), wherein the method provided for RNA in a sufficient amount (> 300 ng) and quality. Total RNA from DRGs was isolated with the PicoPure RNA Isolation Kit (Arcturus) following the manufacturer's instructions. RNA quality was assessed using the 2100 Bioanalyzer and RNA 6000 Nano LabChip kit (Agilent). After having extracted RNA from L5 DRGs of the three mouse strains, either Chung-operated or sham-operated control animals, the RNA probes were hybridized on Affymetrix microarrays (MOE430 2.0).
Affymetrix GeneChip™ Microarray-Analysis:
Samples were profiled with Affymetrix microarrays (MOE430 2.0). At least five animals of each group were tested. First-strand cDNA synthesis was performed using 500ng total RNA with a 100pM T7-(dT)24 oligomer (GGCCAGTGAATTGTAATACGACTCACTATAGGGAGGCGG-dT24) (SEQ ID NO: 15) according to Baugh, L.R, Hill, A.A., Brown, E.L. and Hunter, CP. (2001 ) Nucleic Acids Res. 29, e29 and Superscript II Reverse Transcriptase following the manufacturer's instructions. Double-stranded cDNA was synthesized and then extracted using phenol- chloroform followed by an ethanol precipitation step. An in vitro transcription reaction was performed with the doublestranded cDNA sample using the BioArray High Yield RNA Transcription Labeling kit (Enzo) according to the manufacturer's instructions. Transcription reactions were incubated at 37°C for 16h. cRNA was purified using the RNeasy Mini kit (Qiagen) protocol for RNA cleanup and quantified by a spectrophotometer. The biotin-labeled cRNA was fragmented using a RNA fragmentation buffer (200mM Tris-acetate, 500mM KOAc, 150mM MgOAc, pH 8.1 ). Hybridization and staining of mouse MOE430_2 GeneChips™ (Affymetrix) was performed according to the manufacturer's instructions. The microarrays were scanned using a GeneChip 3000 Scanner, and the scanned data were imported and analyzed using Resolver v5.1 expression data analysis software (Rosetta Biosoftware).
The Affymetrix gene expression data were statistically analyzed and filtered prior to a correlation analysis. The following filter criteria were used: - Abs. fold-change in at least 60 % of all Chung-operated animals > 1 .5 or
- in at least 20 % of all Chung-operated animals > 2.0 (each with respect to the mean value of all sham-operated control animals) and
- gene expression intensity in at least 5 animals > 50 (background level). Phenotype data of the individual mice of the three strains and their gene expression data (expressed as log ratio (Chung-operated vs. sham-operated)) or expression intensity of Chung-operated animals were used for correlation analysis.
For each gene which fulfilled the above filter criteria, a Pearson correlation coefficient of gene expression data and phenotype data (mechanic hypersensitivity) was calculated (Persson et al., supra, particularly section "Correlational analysis"). In order to determine the significance of correlation coefficients of the single genes, a "false discovery rate" (FDR) was introduced (Storey, J.D. (2002) J. R. Statist. Soc. 64, part 3, 479-498). Pearson correlation coefficients of genes having an FDR > 0.05 were regarded as significant. Using the log ratio data (Chung-operated vs. sham-operated) and expression intensity 74 sequences and 1 14 genes, respectively, were identified. Slfn2 was one of the genes identified to exhibit the best correlation data. The data for these sequences/genes, especially Slfn2, showed a significant correlation of gene expression and phenotype data (FDR < 0.05). Slfn2 was not previously known to be involved in pain and hyperalgesia.
Correlational analysis: For correlational analysis, the "pain phenotype" was defined for each nerve-transsected animal (Chung animal) as C1 - S1, where C1 = Infipsilateral PWT / contralateral PWT) and
S1 = meanaii sham animais within same strain ln(ipsilateral PWT / contralateral PWT).
Two measures of differential transcriptional regulation were defined for each Chung animal and each measured gene based on its intensity expression data. The "raw intensity measure" was taken as the intensity measure computed by the Resolver expression data analysis software (v5.1 ) for the respective gene and animal. The "log ratio measure" was computed for a specific gene and Chung animal as ln(C2 / S2), where C2 = Chung expression intensity and S2 = meanaii sham animals within same strain Sham expression intensity.
Before correlations were computed, the set of genes was filtered to exclude genes that were expressed below noise level and without significant Chung vs. sham regulation.
Eligible genes must be regulated in at least 60% of Chung animals with an absolute fold-change =>1 .5 or in at least 20% of Chung animals with an absolute fold-change
=>2.0. Also, corresponding gene expression had to be detectable ("present") in at least five animals as defined by a respective intensity p-value< 0.001 .
Pearson correlation coefficients for each gene between the pain phenotypic scores and one of the defined measures of transcriptional regulation were computed using the R software package (http://www.r-project.org/). Based on these, p-values of statistical significance and corresponding false-discovery rates (FDRs) were generated following the method of Storey et al. (2002). Genes with FDR< 0.05 under "log ratio measure" or
"intensity measure" were considered significantly correlated. Slfn2:
For the Slfn2 gene which was among the genes with the best correlation of expression and pain phenotype, the correlation analysis of log ratio data of L5 DRG signal intensities and mechanic hypersensitivity yielded a Pearson correlation coefficient of 0.75 (p value of 1 .02* 10"5) and an FDR of 0.029.

Claims

Claims 1 . Slfn2 for use as an indicator of a pain-related tissue status or a pain related disease or as an indicator of the risk for developing a pain-related tissue status or disease.
2. Slfn2 for use in the diagnosis or prognosis of a pain-related tissue status or of a pain- related disease.
3. Slfn2 according to claim 1 or 2, wherein the level of Slfn2 is indicative of
(vii) a pain-related tissue status or the presence of a pain-related disease and/or
(viii) the risk of developing a pain-related tissue status or a pain-related disease and/or
(ix) the progression or a stage of a pain-related tissue status or a pain-related disease
in an individual.
4. A method of identifying
c. a pain-related tissue status or the presence of a pain-related disease and/or
d. the risk of developing a pain-related tissue status or a pain-related disease and/or
e. the progression or a stage of a pain-related tissue status or a pain- related disease
in an individual, the method comprising detecting the level of Slfn2.
5. A kit for use in a method according to claim 4, comprising one or more means of detecting Slfn2.
6. A kit for detecting Slfn2 comprising
(i) a means for detecting Slfn2,
(iii) a data carrier comprising instructions for the method of claim 4 and/or (iii) a container.
7. The kit according to claim 6, wherein the means for detecting Slfn2 is a means for determining the expression level of Slfn2, preferably on the gene, protein or RNA level.
8. The kit according to any of claims 6 or 7, wherein the kit comprises
(a) a container, and/or
(b) a data carrier, wherein the data carrier comprises information such as
(i) instructions concerning methods for identifying the risk for developing
and/or identifying the presence and/or monitoring progression of a pain- related tissue status or disease
(ii) instructions for use of the means for detecting Slfn2, preferably in a sample, more preferably in a sample from an individual and/or of the kit,
(iii) quality information such as information about the lot/batch number of the means for detecting Slfn2 and/or of the kit, the manufacturing or assembly site or the expiry or sell-by date, information concerning the correct storage or handling of the kit,
(iv) information concerning the composition of the buffer(s), diluent(s), reagent(s) for detecting Slfn2 and/or of the means for detecting Slfn2,
(v) information concerning the interpretation of information obtained when performing the above-mentioned methods identifying and/or monitoring progression of a pain-related tissue status or disease,
(vi) a warning concerning possible misinterpretations or wrong results when applying unsuitable methods and/or unsuitable means, and/or
(viii) a warning concerning possible misinterpretations or wrong results when using unsuitable reagent(s) and/or buffer(s).
9. The kit according to any of claims 6 to 8, wherein the kit is for diagnosing and/or prognosing pain by determination of Slfn2 and preferably the level of Slfn2 in a sample.
10. The kit according to any of claims 6 to 9, wherein the kit comprises a means according to any of the claims 1 1 to 15.
1 1 . A means of detecting Slfn2 for use in the diagnosis or prognosis of a tissue status or a disease.
12. The means according to claim 1 1 comprising one or more nucleic acids or derivatives for detecting a Slfn2 gene or gene product or functionally active variant thereof, wherein the nucleic acid or derivative is selected from the group consisting of a nucleic acid probe, a polyamide or peptide nucleic acid (PNA), microRNA (miRNA), small interfering RNA (siRNA), primers for polymerase chain reaction (PCR), primers for reverse transcription (RT) reaction, and primers for DNA sequencing.
13. The means according to claim 1 1 or 12 comprising a peptide, polypeptide or protein for detecting a Slfn2 gene or gene product or functionally active variant thereof, wherein the protein or polypeptide is a protein ligand, preferably an antibody, a fragment or derivate thereof, a protein scaffold, such as a darpin or an anticalin, or wherein the polypeptide or peptide is a probe, preferably a mass spectrometry probe.
14. The means according to any of claims 1 1 to 13 , wherein said means comprises
(i) a biochip, or
(ii) a set of beads.
15. The means according to one of claims 1 1 to 14, wherein the diagnosis comprises the method according to claim 4.
PCT/EP2012/060932 2011-06-10 2012-06-08 Methods and uses relating to the diagnosis or prognosis of pain-related tissue states or pain-related diseases such as pain WO2012168453A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP11305732 2011-06-10
EP11305732.7 2011-06-10

Publications (1)

Publication Number Publication Date
WO2012168453A1 true WO2012168453A1 (en) 2012-12-13

Family

ID=46354199

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/060932 WO2012168453A1 (en) 2011-06-10 2012-06-08 Methods and uses relating to the diagnosis or prognosis of pain-related tissue states or pain-related diseases such as pain

Country Status (1)

Country Link
WO (1) WO2012168453A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103784922A (en) * 2014-01-15 2014-05-14 冉兆勇 Traditional Chinese medicine egg boiling powder for treating cervical vertebra, lumbar vertebra and osteoproliferation

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995011910A1 (en) 1993-10-27 1995-05-04 Ribozyme Pharmaceuticals, Inc. 2'-amido and 2'-peptido modified oligonucleotides
US5582981A (en) 1991-08-14 1996-12-10 Gilead Sciences, Inc. Method for identifying an oligonucleotide aptamer specific for a target
WO1997029116A1 (en) 1996-02-06 1997-08-14 Cruachem Limited Sulphur containing dinucleotide phosphoramidites
WO1998037240A1 (en) 1997-02-24 1998-08-27 Tm Technologies, Inc. Anti-sense oligonucleotide pharmacological agents
WO2002031116A2 (en) * 2000-10-11 2002-04-18 Digital Gene Technologies, Inc. Gene expression modulated in spontaneous inflammatory bowel disease
EP2105742A1 (en) * 2008-03-26 2009-09-30 Sanofi-Aventis Use of cathepsin C
US20100048408A1 (en) * 2006-09-06 2010-02-25 Nippon Zoki Pharmaceutical Co., Ltd. Method for study, determination or evaluation by gene expression analysis
EP2320236A1 (en) * 2009-11-06 2011-05-11 Sanofi-aventis Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5582981A (en) 1991-08-14 1996-12-10 Gilead Sciences, Inc. Method for identifying an oligonucleotide aptamer specific for a target
WO1995011910A1 (en) 1993-10-27 1995-05-04 Ribozyme Pharmaceuticals, Inc. 2'-amido and 2'-peptido modified oligonucleotides
WO1997029116A1 (en) 1996-02-06 1997-08-14 Cruachem Limited Sulphur containing dinucleotide phosphoramidites
WO1998037240A1 (en) 1997-02-24 1998-08-27 Tm Technologies, Inc. Anti-sense oligonucleotide pharmacological agents
WO2002031116A2 (en) * 2000-10-11 2002-04-18 Digital Gene Technologies, Inc. Gene expression modulated in spontaneous inflammatory bowel disease
US20100048408A1 (en) * 2006-09-06 2010-02-25 Nippon Zoki Pharmaceutical Co., Ltd. Method for study, determination or evaluation by gene expression analysis
EP2105742A1 (en) * 2008-03-26 2009-09-30 Sanofi-Aventis Use of cathepsin C
EP2320236A1 (en) * 2009-11-06 2011-05-11 Sanofi-aventis Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia

Non-Patent Citations (59)

* Cited by examiner, † Cited by third party
Title
"Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY
"Current Protocols in Human Genetics", WILEY & SONS, INC
"Current Protocols in Molecular Biology", 1991, JOHN WILEY & SONS, pages: 6.3.1 - 6.3.6
"Current Protocols in Molecular Biology", vol. 2000, WILEY & SONS, INC
"Current Protocols in Protein Science", WILEY & SONS, INC
"Gene targeting: A Practical Approach", 2000, IRL PRESS AT OXFORD UNIVERSITY PRESS
"Helvetica Chimica Acta", 1995, article "A multilingual glossary of biotechnological terms: (IUPAC Recommendations"
"Remington's Pharmaceutical Sciences", 1985, pages: 1418
"Short Protocols in Molecular Biology", October 2002, JOHN WILEY & SONS, INC.
"The Merck Manual", 1992, pages: 1518
"Transgenic Animal Technology A Laboratory Handboook", 1994, ACADEMIC PRESS INC.
ALBERTS, B.; BRAY, D.; LEWIS, J.; RAFF, M.; ROBERTS, K.; WATSON, J.D.: "Molecular Biology of the Cell", 1994, GARLAND PUBLISHING, INC.
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
ANONYMOUS: "AF099973", 23 September 2008 (2008-09-23), XP055011141, Retrieved from the Internet <URL:http://srs.ebi.ac.uk/srsbin/cgi-bin/wgetz?[EMBL:AF099973]+-e> [retrieved on 20111103] *
ANONYMOUS: "GeneChip mouse genome array", 1 January 2004 (2004-01-01), http://media.affymetrix.com/support/technical/datasheets/mogarrays_datasheet.pdf, XP055010984, Retrieved from the Internet <URL:http://media.affymetrix.com/support/technical/datasheets/mogarrays_datasheet.pdf> [retrieved on 20111102] *
BAUGH, L.R; HILL, A.A.; BROWN, E.L.; HUNTER, C.P., NUCLEIC ACIDS RES., vol. 29, 2001, pages E29
BEIGELMAN ET AL., NUCLEIC ACIDS RES., vol. 23, no. 1465, pages 3989 - 94
BERGER ET AL., NATURE IMMUNOLOGY, vol. 11, no. 4, 2010, pages 335 - 343
BERGER, M. ET AL.: "A SIfn2 mutation causes lymphoid and myeloid immunodeficiency due to loss of immune cell quiescence", NATURE IMMUNOLOGY, vol. 11, no. 4, 2010, pages 335 - 343
BOURQUIN AF ET AL.: "Assessment and analysis of mechanical allodynia-like behavior induced by spared nerve injury (SNI) in the mouse", PAIN, vol. 122, 2006, pages 1 - 14, XP025024663, DOI: doi:10.1016/j.pain.2005.10.036
BRUDNO M., BIOINFORMATICS, vol. 19, no. 1, 2003, pages 154 - 162
CYSTATIN C, PAIN, vol. 102, 2003, pages 251 - 256
D R GERECKE ET AL: "Gene expression profiles of vesicant-induced skin injury", 1 January 2007 (2007-01-01), pages 5.1 - 5.12, XP055011160, Retrieved from the Internet <URL:http://ftp.rta.nato.int/public//PubFullText/RTO/MP/RTO-MP-HFM-149///MP-HFM-149-05.pdf> [retrieved on 20111103] *
DELEO ET AL., PAIN, vol. 56, 1994, pages 9 - 16
E. KATSOULIDIS ET AL: "Role of Schlafen 2 (SLFN2) in the Generation of Interferon -induced Growth Inhibitory Responses", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 284, no. 37, 11 September 2009 (2009-09-11), pages 25051 - 25064, XP055011144, ISSN: 0021-9258, DOI: 10.1074/jbc.M109.030445 *
HEY, T. ET AL.: "Artificial, non-antibody binding proteins for pharmaceutical and industrial applications", TRENDS IN BIOTECHNOLOGY, vol. 23, no. 10, 2005, XP025290753, DOI: doi:10.1016/j.tibtech.2005.07.007
HORTON, M.R.; POWELL, J.D.: "Quieting T cells with Slfn2", NATURE IMMUNOLOGY, vol. 11, no. 4, 2010, pages 281 - 282
HORTON; POWELL, NATURE IMMUNOLOGY, vol. 11, no. 4, 2010, pages 281 - 282
JAYASENA, CLIN. CHEM., vol. 45, pages 1628 - 50
JOURNAL OF THE AMERICAN OSTEOPATHIC ASSOCIATION, vol. 107, 2007, pages 387 - 400
KARLIN; ALTSCHUL, PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 5877
KATSOULIDIS ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 284, no. 37, 2009
KATSOULIDIS, E. ET AL.: "Role of Schlafen 2 (SLFN2) in the Generation of Interferon alpha-induced Growth Inhibitory Responses", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 284, no. 37, 2009, pages 25051 - 25064, XP055011144, DOI: doi:10.1074/jbc.M109.030445
KIM SH; CHUNG JM, PAIN, vol. 50, no. 3, September 1992 (1992-09-01), pages 355 - 63
KIM; CHUNG, PAIN, vol. 50, 1992, pages 355 - 363
KLUG; FAMULOK, M. MOL. BIOL. REP., vol. 20, pages 97 - 107
KLUSSMANN ET AL., NAT. BIOTECHNOL., vol. 14, no. 1466, pages 1112 - 5
MAIN, C.J.; SPANSWICK, C.C.: "Pain management: an interdisciplinary approach", 2001, ELSEVIER, pages: 93
NA ET AL., NEUROSCI. LETT., vol. 177, 1994, pages 50 - 52
NAGY, A; GERTSENSTEIN, M.; VINTERSTEN, K.; BEHRINGER, R.: "Manipulating the Mouse Embryo: A Laboratory Manual", 2003, COLD SPRING HARBOR PRESS
NOLTE ET AL., NAT. BIOTECHNOL., vol. 14, no. 1466, pages 1116 - 9
PERSSON ANNA-KARIN ET AL: "Correlational analysis for identifying genes whose regulation contributes to chronic neuropathic pain", MOLECULAR PAIN, BIOMED CENTRAL, LONDON, GB, vol. 5, no. 1, 19 February 2009 (2009-02-19), pages 7, XP021051777, ISSN: 1744-8069, DOI: 10.1186/1744-8069-5-7 *
PERSSON ET AL., MOLECULAR PAIN, vol. 5, 2009, pages 7
SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 1989, COLD SPRING HARBOR LABORATORY PRESS, pages: 545
SCHWARZ, D.A.; KATAYAMA, C.D.; HEDRICK, S.M.: "Schlafen, a New Family of Growth Regulatory Genes that Affect Thymocyte Development", IMMUNITY, vol. 9, 1998, pages 657 - 668
SCHWARZ; KATAYAMA; HEDRICK, IMMUNITY, vol. 9, 1998, pages 657 - 668
SELTZER ET AL., PAIN, vol. 43, 1990, pages 205 - 218
SOHN ET AL: "Novel transcriptional regulation of the schlafen-2 gene in macrophages in response to TLR-triggered stimulation", MOLECULAR IMMUNOLOGY, PERGAMON, GB, vol. 44, no. 13, 6 May 2007 (2007-05-06), pages 3273 - 3282, XP022062315, ISSN: 0161-5890, DOI: 10.1016/J.MOLIMM.2007.03.001 *
STOREY JD: "A direct approach to false discovery rates", JOURNAL OF THE ROYAL STATISTICAL SOCIETY, vol. 64, 2002, pages 479 - 498, XP055061495, DOI: doi:10.1111/1467-9868.00346
STOREY, J.D., J. R. STATIST. SOC., vol. 64, 2002, pages 479 - 498
THIENHAUS, O.; COLE, B.E.: "Pain management: A practical guide for clinicians", 2002, AMERICAN ACADEMY OF PAIN MANAGEMENT, article "Classification of pain"
THOMPSON, J. D.; HIGGINS, D. G.; GIBSON, T. J., NUCLEIC ACIDS RES., vol. 22, 1994, pages 4673 - 80
TURK, D.C.; OKIFUJI, A. ET AL.: "Bonica's management of pain", 2001, LIPPINCOTT WILLIAMS & WILKINS, article "Pain terms and taxonomies", pages: 18 - 25
UHLMANN, E.; PEYMAN, A., CHEMICAL REVIEWS, vol. 90, no. 1460, pages 543 - 584
WALKER ET AL., MOLECULAR MEDICINE TODAY, vol. 5, 1999, pages 319 - 321
WERN-JOO ET AL., MOLECULAR IMMUNOLOGY, vol. 44, 2007, pages 3273 - 3282
WERN-JOO SOHN ET AL.: "Novel transcriptional regulation of the schlafen-2 gene in macrophages in response to TLR- triggered stimulation", MOLECULAR IMMUNOLOGY, vol. 44, 2007, pages 3273 - 3282, XP022062315, DOI: doi:10.1016/j.molimm.2007.03.001
XIE, PAIN, vol. 33, 1988, pages 87 - 107

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103784922A (en) * 2014-01-15 2014-05-14 冉兆勇 Traditional Chinese medicine egg boiling powder for treating cervical vertebra, lumbar vertebra and osteoproliferation

Similar Documents

Publication Publication Date Title
CA3047466A1 (en) Compositions and methods for diagnosing autism spectrum disorders
KR20170120124A (en) Biomarker panel for cancer detection
JP2010535825A (en) IVIG modulation of chemokines to treat multiple sclerosis, Alzheimer&#39;s disease and Parkinson&#39;s disease
WO2012168453A1 (en) Methods and uses relating to the diagnosis or prognosis of pain-related tissue states or pain-related diseases such as pain
EP2547789B1 (en) Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia
EP2532755A1 (en) Methods and uses based on Slfn2 expression and relating to the identification and profiling of compounds for use in the treatment or prevention of pain
US20130274120A1 (en) Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia
EP2576817B1 (en) Methods of diagnosing algesia
US20120270749A1 (en) Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia
EP2348129A1 (en) Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia
US8673823B2 (en) Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia
EP2348128A1 (en) Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia
EP2348130A1 (en) Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia
US20130079238A1 (en) Methods and uses relating to the identification of compound involved in pain as well as methods of diagnosing algesia

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12729428

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE