US20200408757A1 - Methods and Compositions for Modulating TH-GM Cell Function - Google Patents

Methods and Compositions for Modulating TH-GM Cell Function Download PDF

Info

Publication number
US20200408757A1
US20200408757A1 US16/902,189 US202016902189A US2020408757A1 US 20200408757 A1 US20200408757 A1 US 20200408757A1 US 202016902189 A US202016902189 A US 202016902189A US 2020408757 A1 US2020408757 A1 US 2020408757A1
Authority
US
United States
Prior art keywords
stat5
cells
csf
mice
tnf
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US16/902,189
Inventor
Xin-Yuan Fu
Wanqiang Sheng
Yongliang Zhang
Fan Yang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
National University of Singapore
Original Assignee
National University of Singapore
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 National University of Singapore filed Critical National University of Singapore
Priority to US16/902,189 priority Critical patent/US20200408757A1/en
Assigned to NATIONAL UNIVERSITY OF SINGAPORE reassignment NATIONAL UNIVERSITY OF SINGAPORE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FU, XIN-YUAN, SHENG, Wanqiang, YANG, FAN, ZHANG, YONGLIANG
Publication of US20200408757A1 publication Critical patent/US20200408757A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4433Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46433Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464436Cytokines
    • A61K39/464439Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0637Immunosuppressive T lymphocytes, e.g. regulatory T cells or Treg
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6869Interleukin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2307Interleukin-7 (IL-7)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/24Interferons [IFN]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/51B7 molecules, e.g. CD80, CD86, CD28 (ligand), CD152 (ligand)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/515CD3, T-cell receptor complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/70Enzymes
    • C12N2501/72Transferases (EC 2.)
    • C12N2501/727Kinases (EC 2.7.)
    • 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
    • C12Q2600/118Prognosis of disease development
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4703Regulators; Modulating activity
    • G01N2333/4706Regulators; Modulating activity stimulating, promoting or activating activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/53Colony-stimulating factor [CSF]
    • G01N2333/535Granulocyte CSF; Granulocyte-macrophage CSF
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/54Interleukins [IL]
    • G01N2333/5403IL-3
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/52Assays involving cytokines
    • G01N2333/54Interleukins [IL]
    • G01N2333/5418IL-7
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/10Musculoskeletal or connective tissue disorders
    • G01N2800/101Diffuse connective tissue disease, e.g. Sjögren, Wegener's granulomatosis
    • G01N2800/102Arthritis; Rheumatoid arthritis, i.e. inflammation of peripheral joints
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/285Demyelinating diseases; Multipel sclerosis

Definitions

  • T H CD4 + helper T
  • TCR T-cell receptor
  • RA rheumatoid arthritis
  • TNF- ⁇ tumor necrosis factor ⁇
  • the present disclosure relates, in part, to the identification of an interleukin-7 (IL-7)/signal transducer and activator of transcription 5 (STAT5)-regulated granulocyte macrophage colony-stimulating factor (GM-CSF)/IL-3-producing T H cells, termed T H -GM, which represent a distinct helper T cell subset with unique developmental and functional characteristics.
  • T H -GM an interleukin-7
  • STAT5 granulocyte macrophage colony-stimulating factor
  • GM-CSF granulocyte macrophage colony-stimulating factor
  • T H -GM which represent a distinct helper T cell subset with unique developmental and functional characteristics.
  • T H -GM-GM an inflammatory pathway mediated by T H -GM cells
  • T H -GM-mediated inflammatory pathway which represents an independent inflammatory pathway apart from known non-T H -GM-mediated inflammatory pathways (e.g., TNF-a, IL-6, and IL-1b pathways of inflammation).
  • the present disclosure provides a method of diagnosing a T H -GM-mediated inflammatory disorder in a patient suffering from an inflammatory disorder, comprising: a) contacting a sample collected from a patient suffering from an inflammatory disorder with a detecting agent that detects a polypeptide or nucleic acid level of STAT5 (e.g., phospho-STAT5 (Tyr694)).
  • a detecting agent that detects a polypeptide or nucleic acid level of STAT5 (e.g., phospho-STAT5 (Tyr694)).
  • IL-7 IL-7, GM-CSF or IL-3, or a combination thereof; and h) quantifying the polypeptide or nucleic acid level of STAT5 (e.g., phospho-STAT5 (Tyr694)), IL-7, GM-CSF or IL-3, or a combination thereof, wherein an increased level of STAT5 (e.g., phospho-STAT5 (Tyr694)), interleukin-7 (IL-7), GM-CSF or interleukin-3 (IL-3), or a combination thereof relative to a reference level indicates that the patient suffers from a T H -GM-mediated inflammatory disorder.
  • STAT5 e.g., phospho-STAT5 (Tyr694)
  • IL-7 interleukin-7
  • IL-3 interleukin-3
  • the present disclosure provides an isolated population of GM-CSF-secreting T-helper cells (T H -GM), wherein the T H -GM cells are differentiated from cluster of differentiation 4 (CD4+) precursor cells in the presence of IL-7 and activated STAT5, and wherein the T H -GM cells express GM-CSF and IL-3.
  • T H -GM GM-CSF-secreting T-helper cells
  • the present disclosure provides a method of modulating T H -GM function, comprising contacting the T H -GM, or CD4+ precursor cells, or both, with a modulating agent that modulates T H -GM function.
  • the present disclosure provides a method of treating a T H -GM-mediated inflammatory disorder in a patient in need thereof, comprising administering to said patient an effective amount of a modulating agent that modulates T H -GM cell function.
  • the present disclosure provides a method of treating rheumatoid arthritis in a patient who exhibits limited response to anti-tumor necrosis factor alpha (TNF- ⁇ ) therapy, comprising administering to said patient an effective amount of a modulating agent that modulates T H -GM function.
  • TNF- ⁇ anti-tumor necrosis factor alpha
  • the present disclosure provides a method of treating a STAT5-mediated inflammatory disorder in a patient in need thereof, comprising administering to the patient an effective amount of an agent that modulates STAT5 function.
  • the present disclosure provides a method of screening to identify a modulator of T H -GM cell function, comprising contacting an isolated population of T H -GM cells, or an isolated population of CD4+ precursor cells, with a candidate agent, and determining a readout of T H -GM function in the presence or absence of the candidate agent, wherein a change in the readout of T H -GM function indicates that the candidate agent is a modulator of T H -GM function.
  • the present disclosure enables the identification or classification between inflammatory disorders that are either primarily T H -GM-mediated, or primarily non-T H -GM-mediated (e.g., mediated by TNF- ⁇ , IL-6, and/or IL-1 ⁇ ), or both.
  • T H -GM-mediated e.g., mediated by TNF- ⁇ , IL-6, and/or IL-1 ⁇
  • RA a patient suffering from, e.g., RA
  • This differentiation allows for a more targeted and tailored method of treating inflammatory disorders such as RA, for which current treatments are only 40% effective.
  • compositions for prognosing the progression of an inflammatory disorder so as to tailor the treatment according to the stage of the disease.
  • compositions and methods for and the treatment of inflammatory disorders particularly those that are T H -GM-mediated.
  • FIGS. 1A-1D depict Stat5-conditional mutant mice are resistant to EAE.
  • Clinical EAE scores FIG. 1A
  • incidence FIG. 1B
  • immunized twice with MOG 35-55 /LPS FIG. 1D
  • Data are representative of two independent experiments.
  • FIGS. 3A and 3B depict the resistance to EAE in Stat5-deficient mice is independent of T H 1, T H 17 or T reg cells.
  • Flow cytometric analysis of IL-17 and IFN- ⁇ expression by CNS-infiltrating CD4 + T cells at peak of disease FIG. 3A .
  • Data are representative of three independent experiments.
  • Percentage of CD25 + among CD4 + T cells in the CNS of Stat5 +/+ and Stat5 ⁇ / ⁇ EAE mice at peak of disease were analyzed by flow cytometry ( FIG. 3B ).
  • FIGS. 4A-4C depict conditional Stat5 mutant mice have no defect in CD4 + T cell generation in periphery.
  • Spleens were obtained from MOG 35-55 /CFA-immunized Stat5 +/+ and Stat5 ⁇ / ⁇ mice at day 7 ( FIG. 4A ) and day 21 ( FIG. 4B ).
  • the proportions of CD4 + and CD8 + T cells were analyzed by flow cytometry.
  • the absolute number of CD4 + T cells was calculated (right panels).
  • Data are representative of two independent experiments ( FIG. 4A ) or pooled from two independent experiments ( FIG. 4B ).
  • IL-17 and IFN- ⁇ expression by splenic CD4 + T cells of Stat5 +/+ and Stat5 ⁇ / ⁇ EAE mice was determined by intracellular cytokine staining ( FIG. 4C ). Data are representative of three independent experiments. *p ⁇ 0.5, **p ⁇ 0.005, ***p ⁇ 0.0005.
  • FIGS. 5A-5D depict Stat5-deficient CD4 + T cells can infiltrate CNS but fail to induce effective neuroinflammation.
  • Data are representative of two independent experiments with three to five mice per group ( FIG. 5A ).
  • CNS-infiltrating CD4 + T cells were analyzed at day 7, 9 and 21 after first MOG 35-55 /CFA immunization ( FIGS. 5B-5D ). Cell numbers were calculated ( FIG. 5D ). Data are representative of two independent experiments with three mice per group. *p ⁇ 0.5.
  • FIGS. 6A-6C show resistance to EAE in Stat5 ⁇ / ⁇ mice is not caused by any defect in the survival of CD4 + T cells in the absence of STAT5.
  • CD4 + T cell infiltration FIG. 6A
  • clinical scores FIG. 6B
  • Clinical scores and frequencies of CD4 + T cells in the CNS at day 21 (disease peak) of EAE induction FIG. 6C ). *p ⁇ 0.05. ***p ⁇ 0.0005.
  • FIGS. 7A-7C depict the intrinsic defect of Stat5-deficient CD4 + T cells in encephalitogenicity.
  • Clinical EAE scores FIG. 7A
  • incidence FIG. 7B
  • IL-17 and IFN- ⁇ expression by CNS-infiltrating CD4 + T cells was measured at peak of disease ( FIG. 7C ).
  • Data represent two independent experiments, *p ⁇ 0.05.
  • FIGS. 8A-8D depict the diminished induction of GM-CSF in splenic Stat5 ⁇ / ⁇ CD4 + T cells.
  • GM-CSF secretion was measured by ELISA ( FIG. 8A ).
  • Golgiplug was added in the last 4 h of MOG 35-55 (20 ⁇ g/ml) challenge and the frequencies of IL-17 + and GM-CSF + cells among CD4 + CD44 hi T cells were measured ( FIG.
  • FIGS. 8C and 8C splenocytes were obtained from MOG 35-55 /CFA-immunized Stat5 ⁇ / ⁇ , Stat3 ⁇ / ⁇ or wild-type control mice and stimulated with PMA/Ionomycin in the presence of Golgiplug for 4 h.
  • the frequencies of IL-17 + and GM-CSF + cells among splenic CD4 + CD44 hi T cells were measured by intracellular cytokine staining. *p ⁇ 0.05, ***p ⁇ 0.001.
  • FIGS. 9A-9C depict the diminished induction of GM-CSF in CNS-infiltrating Stat5 ⁇ / ⁇ CD4 + T cells.
  • FIG. 9A IL-17, IFN- ⁇ and GM-CSF expression by CNS-infiltrating CD4 + T cells of Stat5 +/+ and Stat5 ⁇ / ⁇ mice was measured at peak of disease. The percentage of GM-CSF + cells among IL-17 + or IFN- ⁇ + cells was calculated (bottom right. FIG. 9A ).
  • IL-17 IFN- ⁇ and GM-CSF expression by CNS-infiltrating CD4 + T cells of Rag2 ⁇ / ⁇ recipient mice at peak of disease in adoptive transfer EAE ( FIG. 9B ).
  • FIGS. 10A-10C show STAT5-mediated GM-CSF induction is independent of IL-23 or IL-1 ⁇ signaling.
  • FIG. 10A purified CD4 + T cells were cultured with TGF- ⁇ and IL-6 for 3 days, followed by starvation for 6 h. Then cells were treated with various cytokines for 30 min. and pSTAT3 and pSTAT5 was determined by immunoblotting. STAT3 and STAT5 were further detected after stripping.
  • splenocytes were obtained from MOG 35-55 /CFA-immunized WT mice before disease onset and challenged with MOG 35-55 (20 ⁇ g/ml) in the absence or presence of IL-2 for 48 h.
  • the frequencies of GM-CSF + and IL-17 + cells among CD4 + CD44 hi T cells were measured by flow cytometry. *p ⁇ 0.05.
  • FIGS. 11A-11C depict IL-7-induced STAT5 activation promotes GM-CSF expression in autoreactive CD4 + T cells.
  • Splenocytes were obtained from MOG 35-55 /CFA-immunized Stat5 +/+ and Stat5 ⁇ / ⁇ mice before disease onset and challenged with MOG 35-55 (20 ⁇ g/ml) in the absence or presence of IL-7 for 48 h.
  • Frequencies of GM-CSF + and IL-17 + cells among CD4 + CD44 hi T cells were measured by flow cytometry ( FIG. 11A ).
  • GM-CSF secretion was measured by ELISA ( FIG. 11B ). Data represent two independent experiments with two to three mice per group.
  • FIGS. 12A-12F depict IL-7R ⁇ neutralization attenuates GM-CSF expression and ameliorates EAE.
  • Data represent two independent experiments ( FIG. 12A ).
  • Spinal cord sections were obtained from EAE mice at day 11 after 2 nd immunization. Immune cell infiltration was assessed histologically. Images shown are representative of three individuals per group. Scale bars, 200 ⁇ m (top), 50 ⁇ m ( FIG. 12B , bottom). The percentages of CD4 + and CD8 + T cells in spleens of EAE mice. Data represent two independent experiments ( FIG.
  • FIGS. 12D and 12E illustrate the frequencies of GM-CSF + , IL-17 + and IFN- ⁇ + cells among CD4 + T cells in the CNS of EAE mice receiving different treatment.
  • the mRNA expression of IFN- ⁇ , IL-17 and GM-CSF in the CNS of EAE mice ( FIG. 12F ). *p ⁇ 0.05
  • FIGS. 13A and 13B depict the differentiation of GM-CSF-expressing T H cells is distinct from T H 17 and T H 1.
  • Na ⁇ ve CD4 + T cells were primed with plate-bound anti-CD3 and soluble anti-CD28 in the presence of a combination of various cytokines and neutralizing antibodies as indicated.
  • GM-CSF, IL-17 and IFN- ⁇ expression was analyzed by intracellular staining ( FIG. 13A ) or RT-PCR ( FIG. 13B )
  • FIGS. 14A-14D show the effect of IL-2 and IL-6 on T H -GM differentiation from na ⁇ ve T cells.
  • GM-CSF and IFN- ⁇ expression in naive CD4 + T cells activated for 72 h with anti-CD3 alone or plus anti-CD28 ( FIG. 14A ).
  • FIG. 14B sorted na ⁇ ve CD4 + T cells were stimulated with anti-CD3 and anti-CD28 in the presence of neutralizing antibodies against IL-12 and IFN- ⁇ without or with the addition of IL-6.
  • the frequencies of GM-CSF + and IL-17 + cells were measured by intracellular staining ( FIG. 14B ).
  • FIG. 14B shows the effect of IL-2 and IL-6 on T H -GM differentiation from na ⁇ ve T cells.
  • na ⁇ ve CD4 + T cells from Stat3 +/+ and Stat3 ⁇ / ⁇ mice were polarized under conditions as indicated for 72 h. The frequencies of GM-CSF + and IL-17 + cells were analyzed. In FIG. 14D , na ⁇ ve CD4 + T cells were activated with anti-CD3 and anti-CD28 in the presence of IL-2 or anti-IL-2. The frequencies of GM-CSF + , IL-17 + and IFN- ⁇ + cells were analyzed.
  • FIGS. 15A-15F depict IL-7-STAT5 signaling programs T H -GM differentiation from na ⁇ ve precursor cells.
  • Na ⁇ ve CD4 + T cells were primed with plate-hound anti-CD3 and soluble anti-CD28 in the presence of various concentration of IL-7 as indicated.
  • GM-CSF and IFN- ⁇ expression was analyzed by intracellular staining ( FIG. 15A ) or ELISA ( FIG. 15B ).
  • FIGS. 15C and 15D Stat5 +/+ and Stat5 ⁇ / ⁇ na ⁇ ve CD4 + T cells were activated with anti-CD3 and anti-CD28 in the presence IL-7 for 3 days.
  • GM-CSF, IL-17 and IFN- ⁇ expression was analyzed by intracellular cytokine staining ( FIG. 15C ).
  • GM-CSF secretion was measured by ELISA ( FIG. 15D ).
  • the ChIP assay was performed with Stat5 +/+ and Stat5 ⁇ / ⁇ CD4 + T cells using normal IgG or STAT5-specific antibody.
  • the binding of antibodies to Csf2 promoter region was detected by RT-PCR ( FIG. 15F ).
  • FIGS. 16A and 16B depict the differentiation conditions for T H -GM subset.
  • Na ⁇ ve CD4 + T cells were activated with anti-CD3 and anti-CD28 in the presence of IL-7 or/and anti-IFN- ⁇ as indicated.
  • GM-CSF. IL-17 and IFN- ⁇ expression was analyzed ( FIG. 16A ).
  • T H 1 IL-12+anti-IL- ⁇
  • T H 17 TGF- ⁇ +IL-6+anti-IFN- ⁇ +anti-IL-4)
  • T H -GM cells IL-7 anti-IFN- ⁇
  • FIGS. 17A-17E illustrate that IL-7 but not IL-2 induces STAT5 activation and GM-CSF expression in na ⁇ ve CD4 + T cells.
  • FIGS. 17A-17C show flow cytometry of CD25 and CD127 on the surface of na ⁇ ve CD4 + T cells or cells activated with anti-CD3 and anti-CD28 at various time points as indicated. Activation of STAT5 in na ⁇ ve CD4 + T cells stimulated with IL-2 or IL-7 for 30 min ( FIG. 17D ).
  • FIG. 17E shows the mRNA expression of GM-CSF in na ⁇ ve CD4 + T cells stimulated with anti-CD3 and anti-CD28 in the presence of IL-2 or IL-7.
  • the RT-PCR data were normalized to ⁇ -Actin, and expression in na ⁇ ve T cells activated for 2 h without cytokine was set to 1.
  • FIGS. 18A-18C show that both IL-2 and IL-7 can induce STAT5 activation and GM-CSF expression in activated CD4 + T cells.
  • CD4 + T cells were activated with anti-CD3 and anti-CD28 for 3 days. After resting in fresh medium, cells were stimulated with IL-2 or IL-7 at various time points. The pTyr-STAT5 and ⁇ -Actin were detected by immunoblotting ( FIG. 18A ).
  • GM-CSF mRNA expression was measured by RT-PCR ( FIG. 18B ).
  • the RT-PCR data were normalized to 13-Actin, and expression in cells without cytokine stimulation was set to 1.
  • the ChIP assay shown in FIG. 18C was performed with normal IgG or STAT5-specific antibody. The binding of antibodies to Csf2 promoter region was detected by RT-PCR.
  • FIG. 19 depicts surface molecules selectively expressed at high level or low level in T H -GM subset as characterized by microarray analysis. These surface molecules specific for each lineage serves as markers, signatures and potential targets for novel diagnosis, treatment and prevention of autoimmune inflammation including, but not limited to multiple sclerosis and rheumatoid arthritis. These cell surface molecules are listed in detail in Table 1. The order of na ⁇ ve, Th1, Th17, and Th-GM as indicated in the figure insert is the same as that appears for the bars in each graph.
  • FIGS. 20A-20D show that IL-3 is preferentially expressed in T H -GM cells.
  • na ⁇ ve CD4 + T cells were activated with anti-CD3 and anti-CD28 under T H 1—(IL-12+anti-IL-4), T H 17 ⁇ (TGF- ⁇ +IL-6+anti-IFN- ⁇ +anti-IL-4) and T H -GM—(GM-CSF + T H , IL-7+anti-IFN- ⁇ +anti-IL-4) polarizing conditions.
  • GM-CSF and IL-3 expression was analyzed by intracellular staining ( FIG. 20A ).
  • IL-3, EBI-3, PENK or RANKL cytokines was measured by RT-PCR ( FIG. 20B ).
  • Frequency of IL-3+ cells differentiated without or with IL-7 FIG. 20C .
  • GM-CSF and IL-3 expression by WT or STAT5-deficient GM-CSF-producing TH cells FIG. 20D ).
  • FIGS. 22A-27E depict inhibition of STAT5 activation suppresses T H -GM cell differentiation in vitro.
  • CD4 + T cells were pre-incubated with STAT5 inhibitor (Calbiochem) ( FIG. 22A ) or JAK3 inhibitor ( FIG. 22B ) at indicated concentrations or vehicle ( ⁇ ) for 1 h before stimulation with IL-7 for 30 min.
  • Activation (Tyr694 phosphorylation) of STAT5 was determined by immunoblotting.
  • CD4 + T cells were pre-incubated with STAT5 inhibitor at indicated concentrations or vehicle ( ⁇ ) for 1 h before stimulation with IL-6 for 30 min.
  • Activation (Tyr705 phosphorylation) of STAT3 was determined by immunoblotting ( FIG. 22C ).
  • FIG. 22C depict inhibition of STAT5 activation suppresses T H -GM cell differentiation in vitro.
  • CD4 + T cells were pre-incubated with STAT5 inhibitor at indicated concentrations or vehicle ( ⁇ ) for 1 h before stimulation with IFN- ⁇ for 30 min.
  • Activation (Tyr701 phosphorylation) of STAT1 was determined by immunoblotting.
  • na ⁇ ve CD4 + T cells were isolated and activated under neutral condition or T H -GM cell-favoring condition with the addition of different concentrations of STAT5 inhibitor for 3 days.
  • GM-CSF and IFN- ⁇ expression was analyzed by intracellular cytokine staining and flow cytometry.
  • FIGS. 23A-23D depict targeting STAT5 activation by chemical inhibitor ameliorates EAE.
  • FIG. 23B Histology of spinal cords at clay 18 of EAE mice receiving different treatments.
  • FIG. 23C Intracellular staining and flow cytometry of CNS-infiltrating CD4 + T cells at peak of disease.
  • FIG. 23D Whole CNS was harvest for RNA extraction. GM-CSF mRNA expression was analyzed by RT-PCR. Data represent two independent experiments. *p ⁇ 0.05.
  • FIGS. 24A-24E depict GM-CSF-producing T H cells are in association with human RA.
  • FIGS. 24B and 24C peripheral blood mononuclear cells (PBMCs) were collected from healthy control (HC) and rheumatoid arthritis (RA) patients, and were stimulated for 4 h with PMA/lonomycin in the presence of Golgiplug, followed by intracellular cytokine staining. Representative flow cytometry of GM-CSF, IFN- ⁇ and IL-17 in CD4 + T cells ( FIG.
  • Cytokine expression by CD4 + T cells derived from synovial fluid of RA patients was analyzed by intracellular cytokine staining and flow cytometry ( FIG. 24E ). A representative image of three cases was shown. *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001; ns, not significant.
  • FIGS. 25A-25E depict distinguishable effects of GM-CSF and TNF- ⁇ in mouse AIA.
  • FIG. 25D at day 7 after arthritis induction as in FIG. 25C .
  • FIGS. 26A-26D depicts mice with Stat5 deletion in T cells are resistant to CIA.
  • FIG. 26A Representative images of paw swelling of Stat5 +/+ and Stat5 ⁇ / ⁇ mice at day 40 after collagen II/CFA immunization in CIA model.
  • FIG. 26C Representative images of paw sections stained with H&E at clay 40.
  • FIGS. 27A-27E depicts STAT5-deficient CD4 + T cells are defective in arthritogenic potential.
  • FIGS. 27A and 27B Representative flow cytometry of CD4 + T cells in spleens ( FIG. 27A ) and inguinal lymph nodes ( FIG. 27B ) of Stat5 +/+ and Stat5 ⁇ / ⁇ mice at day 7 after AIA induction.
  • FIGS. 27C and 27D Synovial tissues were harvested from Stat5 +/+ and Stat5 ⁇ / ⁇ mice at day 7 after AIA induction, and dissociated into single cells. Cell numbers of CD45 + leukocytes were calculated ( FIG. 27C ).
  • FIG. 27D The percentages of CD4 + T cells among CD45 + fraction were analyzed by flow cytometry, and cell numbers were calculated ( FIG. 27D ).
  • FIG. 27E Histological analysis of joint sections from wild-type na ⁇ ve mice at day 7 after being transferred with in vitro-expanded antigen-reactive CD4 + T cells and followed with intraarticular injection of mBSA. Bars, 100 ⁇ m. Data represent two independent experiments. *p ⁇ 0.05; ns, not significant.
  • FIGS. 28A-28G depicts STAT5-regulated GM-CSF-producing T H cells are crucial for AIA.
  • Spleens and synovial tissues were collected from Stat5 +/+ and Stat5 ⁇ / ⁇ mice at day 7 after arthritis induction.
  • FIGS. 28B-28D Intracellular staining and flow cytometry of GM-CSF, IL-17 and IFN- ⁇ in splenic CD4 + CD44 hi effector T cells ( FIG.
  • FIG. 28E Protein expression of several proinflammatory cytokines in synovial tissues was measured by ELISA.
  • FIGS. 28F and 28G Representative images of joint sections stained with H&E ( FIG. 28F ) or Safranin-O/Fast Green ( FIG.
  • FIGS. 29A-29C depicts loss of STAT5 results in impaired GM-CSF production by antigen-specific CD4 + T cells.
  • Spleens and inguinal LNs were collected from Stat5 +/+ and Stat5 ⁇ / ⁇ mice at day 7 after arthritis induction, and dissociated into single cell suspensions. Then, cells were stimulated with mBSA (20 ⁇ g/ml) for 24 h.
  • FIG. 29A Golgiplug was added in the last 4 h of culture, followed by intracellular staining and flow cytometry. Representative plots of GM-CSF, IL-17 and IFN- ⁇ expression in CD4 + CD44 hi effector T cells was shown, representing two independent experiments.
  • FIGS. 30A-30C depicts loss of STAT5 impairs IL-6 and IL-1 ⁇ expression in synovial tissues of arthritic mice.
  • FIGS. 31A-31D depicts SAT5-induced GM-CSF expression mediates CD11b + cell accumulation in inflamed synovial tissues.
  • FIG. 31B Synovial tissues were harvested
  • FIG. 31C Representative flow cytometry of CD11b + and CD4 + cells gated on synovial CD45 + fraction over 7 days after arthritis induction.
  • FIG. 31D Flow cytometric analysis of CD4 + , CD11b + and B220 + cell infiltration in synovial tissues of Stat5 +/+ and Stat5 ⁇ / ⁇ mice at day 7 after intraarticular injection of mBSA alone to the right knee joints and mBSA supplemented with GM-CSF to the left knee joints. Representative images were shown. All data shown are representative of two independent experiments. **p ⁇ 0.01; ns, not significant.
  • FIGS. 32A-32D depicts GM-CSF mediates neutrophil accumulation in arthritic mice.
  • FIG. 32A Flow cytometric analysis of Ly6C and Ly6G expression gated on synovial CD45 + CD11b + fraction over 7 days after arthritis induction.
  • FIG. 32B Giemsa stain of sorted Ly6C hi Ly6G ⁇ and Ly6C lo Ly6G hi cells from synovial tissues of AIA mice. Scale bar, 100 ⁇ m (left) or 20 ⁇ m (right).
  • FIG. 32A Flow cytometric analysis of Ly6C and Ly6G expression gated on synovial CD45 + CD11b + fraction over 7 days after arthritis induction.
  • FIG. 32B Giemsa stain of sorted Ly6C hi Ly6G ⁇ and Ly6C lo Ly6G hi cells from synovial tissues of AIA mice. Scale bar, 100 ⁇ m (left) or 20 ⁇ m (right).
  • FIG. 32C Flow cytometric analysis of Ly6C hi Ly6G ⁇ and Ly6C lo Ly6G hi populations in synovial tissues of Stat5 +/+ and Stat5 ⁇ / ⁇ mice at day 7 after intraarticular injection of mBSA alone to the right knee joints and mBSA supplemented with GM-CSF to the left knee joints.
  • FIGS. 33A-33C depicts GM-CSF enhances neutrophil transmigration and delay apoptosis in vitro.
  • FIG. 33A Percentages of migrated neutrophils with or without GM-CSF as chemoattractant in transmigration assay at 3 h post start.
  • FIG. 33B Microscopic images of CFSE-labeled neutrophils in the bottom of the lower chamber in transmigration assay.
  • FIG. 33C Sorted neutrophils were cultured in vitro with or without GM-CSF (20 ng/ml) for 24 h. Neutrophils undergoing apoptosis were examined by Annexin V and propidium iodide (PI) co-staining. A representative flow cytometry of three repeats was shown. *p ⁇ 0.05.
  • FIGS. 34A-34I depicts GM-CSF mediates proinflammatory cytokine expression by myeloid cells and synovial fibroblasts in arthritic mice. Synovial tissues were dissected from wild-type AIA mice and dissociated into single cell suspensions.
  • FIG. 34A Flow cytometry plots depicting the fractionation into different populations based on differential expression of surface markers.
  • FIG. 34B The mRNA expression of several proinflammatory cytokines in sorted CD45 + TCR ⁇ + (TCR ⁇ + in short). CD45 + TCR ⁇ ⁇ CD11c ⁇ CD11b + (CD11b + ) and CD45 + TCR ⁇ ⁇ CD11c + (CD11c + ) populations was measured by qPCR.
  • FIG. 34C The mRNA expression of IL-6, IL-1 ⁇ and TNF- ⁇ in sorted Ly6C hi Ly6G ⁇ and Ly6C lo Ly6G hi populations (gated on CD11b + cells).
  • the qPCR data were normalized to GAPDH.
  • FIGS. 34D and 34E The mRNA expression of IL-6 and IL-1 ⁇ by BMDMs ( FIG. 34D ) and BMDCs ( FIG. 34E ) upon stimulation with 20 ng/ml GM-CSF for 1 h.
  • the qPCR data were normalized to GAPDH.
  • FIGS. 34F and 34G BMDMs ( FIG.
  • FIG. 34F BMDCs
  • FIG. 34G BMDCs
  • the secretion of IL-1 ⁇ in the culture supernatant was quantified by ELISA.
  • T H -GM granulocyte macrophage colony stimulating factor
  • IL-7/STAT5 signaling programs the differentiation of precursor CD4+ cells to T H -GM, a process which is further modulated by IL-2 and IL-23 signaling.
  • T H -GM cells are characterized by, e.g., GM-CSF and IL-3 production.
  • T H -GM cells are distinct from the known helper T cells T H 1 and T H 17, with respect to, e.g., differentiation conditions, transcriptional regulation and effector cytokine expression.
  • IL-12/IFN- ⁇ and TGF-0/IL-6 which mediate (e.g., promote the development of) T H 1 and T H 17, respectively, potently suppress the development of T H -GM from na ⁇ ve CD4 + precursor cells, establishing that T H -GM cells develop via a lineage distinct from T H 1 and T H 17.
  • the present disclosure provides a distinct network of factors, unique from factors known to mediate T H 1 or T H 17, that mediate T H -GM function (e.g., its differentiation and pathogenicity).
  • T H -GM cells preferentially induce EAE as compared with T H 1 and T H 17 cells, indicating that T H -GM cells represent the primary effectors in the pathogenesis of autoimmune neuroinflammation in humans.
  • blockade of IL-7 signaling and/or inhibition of STAT5 function e.g., abrogation of expression or inhibition of STAT5 activity attenuates autoimmune neuroinflammation associated with diminished GM-CSF production by T H -GM cells.
  • blockade of T H -GM cell-secreted GM-CSF ameliorates experimental arthritis in a TNT- ⁇ -independent manner, indicating an approach for the treatment of, e.g., rheumatoid arthritis patients who are unresponsive to TNF- ⁇ antagonistic drugs.
  • the present disclosure enables one to distinguish between an inflammatory disorder (e.g., RA) that is mediated by the T H -GM pathway (e.g., a disorder that results from T H -GM pathogenicity through the action of, e.g., GM-CSF and/or IL-3, or any factor associated with the T H -GM pathway), or an inflammatory disorder that is mediated by, e.g., TNF- ⁇ , IL-6, and/or IL-1 ⁇ pathways (i.e., non-T H -GM-mediated pathway).
  • an inflammatory disorder e.g., RA
  • T H -GM pathway e.g., a disorder that results from T H -GM pathogenicity through the action of, e.g., GM-CSF and/or IL-3, or any factor associated with the T H -GM pathway
  • an inflammatory disorder that is mediated by, e.g., TNF- ⁇ , IL-6, and/or IL-1 ⁇ pathways
  • a patient who has, e.g., RA may be afflicted with a type of RA that is primarily T H -GM-mediated, or primarily non-T H -GM-mediated (e.g., TNF- ⁇ -mediated or IL-6 mediated).
  • a type of RA that is primarily T H -GM-mediated, or primarily non-T H -GM-mediated (e.g., TNF- ⁇ -mediated or IL-6 mediated).
  • the present disclosure enables the classification between T H -GM-mediated and non-T H -GM-mediated inflammation, allowing for a more precise diagnosis, prognosis, and treatment in an individual who is afflicted with an inflammatory disorder such as RA or MS.
  • T H -GM helper T cell subset
  • T H -GM helper T cell subset
  • TNF- ⁇ therapy e.g., TNF- ⁇ inhibitor based therapy
  • compositions and methods for modulating T H -GM function to treat autoimmune and inflammatory disorders are provided herein.
  • the methods of modulating T H -GM function include, e.g., administering agents to modulate the function (e.g., signaling, expression or activity) of the network of factors (e.g., IL-2/IL-7/STAT5/GM-CSF/IL-3) that mediate T H -GM function in an effective amount to modulate the function (e.g., development and pathogenicity) of T H -GM cells.
  • the disclosure provides methods and composition for differentiating and diagnosing an inflammatory disorder, e.g., multiple sclerosis (MS), rheumatoid arthritis (RA) as primarily mediated by either T H -GM cells (i.e.
  • compositions and methods for and the treatment of inflammatory disorders particularly those that are T H -GM-mediated.
  • the present disclosure provides a method of diagnosing a T H -GM-mediated inflammatory disorder in a patient suffering from an inflammatory disorder.
  • the method comprises contacting a sample collected from a patient suffering from an inflammatory disorder with a detecting agent that detects a polypeptide or nucleic acid level of a T H -GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof; and quantifying the polypeptide or nucleic acid level of the T H -GM-mediating factor (e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof), wherein an increased level of a T H -GM-mediating factor (e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof) relative to a reference level indicates that the patient suffers from a T H -GM-mediated inflammatory disorder,
  • a detecting agent that detects
  • a “T H -GM-mediated” inflammatory disorder refers to a subtype of an inflammatory disorder (e.g., a subtype of RA or MS) that results from the physiological action of any one or more of the network of factors in the pathway that modulate T H -GM function (a “T H -GM-mediating factor”), as described herein.
  • factors include, e.g., GM-CSF, activated STAT5, IL-7, IL-2, and IL-3.
  • STAT5 is activated STAT5, wherein tyrosine at position 694 is phosphorylated.
  • the level of a T H -GM-mediating factor (e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof) that is not increased relative to a reference level indicates that the patient suffers from a non-T H -GM-mediated inflammatory disorder.
  • a T H -GM-mediating factor e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof
  • the method further comprises administering to the patient a TNF- ⁇ therapy, as described herein, if the level of a T H -GM-mediating factor (e.g. STAT5, IL-7, GM-CSF or IL-3, or a combination thereof) is not increased relative to a reference level.
  • a T H -GM-mediating factor e.g. STAT5, IL-7, GM-CSF or IL-3, or a combination thereof
  • a “non-T H -GM-mediated” inflammatory disorder refers to an inflammatory disorder (e.g., RA or MS) that is primarily caused by, e.g., TNF- ⁇ , IL-6, or IL-1 ⁇ (and/or factors in the TNF- ⁇ , IL-6, or IL-1 ⁇ pathway).
  • a “T H -GM-mediated” inflammatory disorder results primarily (or exclusively) from a pathway that is distinct from one or more of the pathways that leads to a “non-T H -GM-mediated” inflammatory disorder (e.g., the pathways associated with TNF- ⁇ , IL-6, or IL-1 ⁇ ).
  • a T H -GM-mediated inflammatory disorder does not necessarily exclude the possibility that the inflammatory disorder could also be partially non-T H -GM-mediated (e.g., mediated by TNF- ⁇ , IL-6, or IL- and/or factors in the TNF- ⁇ , IL-6, or IL-1 ⁇ pathway).
  • T H -GM-mediated is synonymous with “primarily/predominantly T H -GM-mediated”
  • non-T H -GM-mediated is synonymous with “primarily/predominantly non-T H -GM-mediated.”
  • an inflammatory disorder in its early stage may be T H -GM-mediated.
  • the inflammatory condition advances to a late stage characterized by, e.g., tissue damage, the inflammatory disorder becomes progressively non-T H -GM-mediated.
  • a T H -GM-mediated inflammatory disorder is a condition that is responsive to modulation of T H -GM function, as determined by clinical standards;
  • a non-T H -GM-mediated inflammatory disorder is a condition that is responsive to, e.g., TNF- ⁇ , IL-6, or IL-1 ⁇ therapy, as determined by clinical standards.
  • an inflammatory disorder can be responsive to modulation of T H -GM function as well as TNF- ⁇ , IL-6, and/or IL-1 ⁇ therapy.
  • the sample can be e.g., peripheral blood, cerebrospinal fluid, synovial fluid, or synovial membrane, or a combination thereof.
  • the inflammatory disorder is an autoimmune disorder.
  • the inflammatory disorder can be any inflammatory disorder mediated by T H -GM cells, and includes, but is not limited to rheumatoid arthritis, multiple sclerosis, ankylosing spondylitis, Crohn's disease, diabetes.
  • Hashimoto's thyroiditis hyperthyroidism, hypothyroidism, Irritable Bowel Syndrome (IBS), lupus erythematosus, polymyalgia rheumatic, psoriasis, psoriatic arthritis, Raynaud's syndrome/phenomenon, reactive arthritis (Reiter syndrome), sarcoidosis, scleroderma, Sjögren's syndrome, ulcerative colitis, uveitis, or vasculitis.
  • IBS Irritable Bowel Syndrome
  • lupus erythematosus polymyalgia rheumatic
  • psoriasis psoriatic arthritis
  • Raynaud's syndrome/phenomenon reactive arthritis
  • Reiter syndrome reactive arthritis
  • sarcoidosis scleroderma
  • Sjögren's syndrome ulcerative colitis
  • uveitis or vasculitis.
  • a “detecting agent” refers to, e.g., an antibody, a peptide, a small molecule, or a nucleic acid that binds to a polypeptide or nucleic acid to be detected (e.g., STAT5 (e.g., phospho-STAT5 (Tyr694)), IL-7, GM-CSF or IL-3), and enables the quantification of the polypeptide or nucleic acid to be detected.
  • the detecting agent can be detectably labeled, or quantifiable by other means known in the art.
  • the detecting agent is an antibody that binds to the polypeptide of STAT5, IL-7, GM-CSF or IL-3.
  • the antibody is one that binds to an activated STAT5 (e.g., phosphorylated STAT5), as described herein.
  • STAT5 e.g., phospho-STAT5 (Tyr694)
  • IL-7 IL-7
  • GM-CSF IL-3
  • STAT5 Ab C-17 from Santa Cruz Biotech
  • IL-7 Ab clone BVD10-40F6 from BD Pharmingen
  • IL-7R Ab: clone SB/14 from BD Pharmingen
  • GM-CSF clone MP1-22E9 from BD Pharmingen
  • IL-3 Ab: clone MP2-8F8 from BD Pharmingen.
  • the detecting agent is a nucleic acid that hinds to the nucleic acid of STAT5.
  • IL-7, GM-CSF and/or IL-3 sequence, or fragments or oligonucleotides thereof, that hybridize to a nucleic acid molecule encoding a e.g., STAT5, IL-7, GM-CSF and/or IL-3 polypeptide sequence at high stringency may be used as a probe to monitor expression of nucleic acid levels of STAT5, IL-7, GM-CSF and/or IL-3 in a sample for use in the diagnostic methods of the disclosure. Methods of quantifying nucleic acid levels are routine and available in the art.
  • the method further comprises contacting the sample with a detecting agent that detects a polypeptide or nucleic acid level of one or more genes (as well as the gene product) listed in Table 1.
  • a detecting agent that detects a polypeptide or nucleic acid level of one or more genes (as well as the gene product) listed in Table 1.
  • Table 1 lists genes that are differentially expressed in T H -GM cells as well as genes that are differentially expressed on the surface of T H -GM cells, as compared to T H 1 or T H 17 cells.
  • the method further comprises contacting the sample with a detecting agent that detects the polypeptide or nucleic acid level of basic helix-loop-helix family member e40 (BHLHe40), chemokine (C-C Motif) Receptor 4 (CCR4), and/or CCR6.
  • a detecting agent that detects the polypeptide or nucleic acid level of basic helix-loop-helix family member e40 (BHLHe40), chemokine (C-C Motif) Receptor 4 (CCR4), and/or CCR6.
  • Standard methods may be used to quantify polypeptide levels in any sample. Such methods include, e.g., ELISA, Western blotting, immunohistochemistry, fluorescence activated cells sorting (FACS) using antibodies directed to a polypeptide, and quantitative enzyme immunoassay techniques known in the art. Such methods are routine and available in the art. Similarly, methods for quantifying nucleic acid levels (e.g., mRNA) are known in the art.
  • an increased level of STAT5 e.g., activated phospho-STAT5 (Tyr694)
  • IL-7 e.g., IL-7
  • GM-CSF e.g., IL-3
  • a STAT5 e.g., activated phospho-STAT5 (Tyr694)
  • IL-7 e.g., IL-7
  • GM-CSF e.g., IL-3
  • IL-3 level that is increased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 110%, at least 120%, at least 130%, at least 140%, at least 150%, at least 160%, at least 170%, at least 180%, at least 190%, at least 200%, at least 220%, at least 240%, at least 260%, at least 280%, at least 300%, at least 350%, at least 400%, at least 450%, at least 500%, at least 550%, or at least 600% relative to a reference level indicates that the patient suffers from a T H -GM-mediated inflammatory disorder.
  • an increase of at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, or at least 150% relative to a reference level indicates that the patient suffers from a T H -GM-mediated inflammatory disorder.
  • a STAT5 e.g., activated phospho-STAT5 (Tyr694)
  • IL-7 e.g., IL-7
  • GM-CSF e.g., IL-3
  • IL-3 level that is not increased by at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, or at least 150% relative to a reference level indicates that the patient suffers from a non-T H -GM-mediated inflammatory disorder.
  • a STAT5 e.g., activated phospho-STAT5 (Tyr694)
  • IL-7 e.g., IL-7
  • GM-CSF e.g., IL-3
  • IL-3 e.g., IL-3
  • a level that is “comparable” to that of a reference level refers to a level that is unchanged, or a change relative to the reference level that is statistically insignificant according to clinical standards.
  • a comparable level can include a level that is not increased by at least 40%, at least 50%, at least 60%, or at least 70% relative to a reference level as, for example, it may not indicate a clinically significant change.
  • a level of a T H -GM-mediating factor e.g., STAT5 (e.g., activated phospho-STAT5 (Tyr694)).
  • STAT5 e.g., activated phospho-STAT5 (Tyr694)
  • IL-7, GM-CSF, and/or IL-3 that is decreased relative to a reference level can also indicate that the patient suffers from a non-T H -GM-mediated disorder.
  • the reference level is a level that is used for comparison purposes, and may be obtained from, for example, a prior sample taken from the same patient; a normal healthy subject; a sample from a subject not having an autoimmune disease or an inflammatory disorder; a subject that is diagnosed with a propensity to develop an autoimmune disease but does not yet show symptoms of the disease; a patient that has been treated for an autoimmune disease; or a sample of a purified reference polypeptide or nucleic acid molecule of the disclosure (e.g., STAT5) at a known normal concentration.
  • a purified reference polypeptide or nucleic acid molecule of the disclosure e.g., STAT5
  • reference standard or level is meant a value or number derived from a reference sample, or a value or range accepted in the art as indicative of being healthy (e.g., an individual that does not have an inflammatory disorder).
  • a normal reference standard or level can also be a value or number derived from a normal subject who does not have an autoimmune disease.
  • the reference sample, standard, or level is matched to the sample subject by at least one of the following criteria; age, weight, body mass index (BMI), disease stage, and overall health.
  • BMI body mass index
  • a standard curve of levels of purified DNA. RNA or mRNA within the normal reference range can also be used as a reference.
  • a standard curve of levels of purified protein within the normal reference range can also be used as a reference.
  • the patient afflicted with an inflammatory disorder who has been diagnosed or classified as having a T H -GM-mediated inflammatory disorder does not have a non-T H -GM-mediated inflammatory disorder (i.e., does not have a TNF- ⁇ , IL-6, or IL-1 ⁇ -mediated inflammatory disorder). That is, the patient diagnosed as suffering from a T H -GM-mediated inflammatory disorder responds to modulation of T H -GM function (e.g., inhibition of STAT5, IL-7, GM-CSF and/or IL-3), but does not respond (or exhibits a limited response) to TNF- ⁇ therapy, as determined by clinical standards.
  • T H -GM function e.g., inhibition of STAT5, IL-7, GM-CSF and/or IL-3
  • a T H -GM-mediated inflammatory disorder does not exclude the possibility that the inflammatory disorder is also partially (though not primarily) contributed by a non-T H -GM-mediated pathway (e.g., TNF- ⁇ , IL-6, IL-1 ⁇ ).
  • a non-T H -GM-mediated pathway e.g., TNF- ⁇ , IL-6, IL-1 ⁇ .
  • the methods of the present disclosure further comprise administering an effective amount of a modulating agent that modulates T H -GM cell function to the patient diagnosed or classified as having a T H -GM-mediated inflammatory disorder.
  • the modulating agent inhibits T H -GM function.
  • the methods of the present disclosure further comprise administering an effective amount of, e.g., a TNF- ⁇ therapy, an IL-6 therapy, or an IL-1 ⁇ therapy to a patient diagnosed or classified as having a non-T H -GM-mediated inflammatory disorder, as described herein.
  • an effective amount of, e.g., a TNF- ⁇ therapy, an IL-6 therapy, or an IL-1 ⁇ therapy to a patient diagnosed or classified as having a non-T H -GM-mediated inflammatory disorder, as described herein.
  • the present disclosure also provides a method of classifying a patient suffering from an inflammatory disorder as having a T H -GM-mediated inflammatory disorder or a non-T H -GM-mediated inflammatory disorder.
  • the method comprises contacting a sample collected from a patient suffering from an inflammatory disorder with a detecting agent that detects a polypeptide or nucleic acid level of a T H -GM-mediating factor, such as, e.g., STAT5 (e.g., phosphorylated STAT5, Tyr694), IL-7, GM-CSF or IL-3, or a combination thereof.
  • STAT5 e.g., phosphorylated STAT5, Tyr694
  • IL-7 e.g., IL-7
  • GM-CSF GM-CSF
  • IL-3 e.g., IL-3
  • the method further comprises quantifying the polypeptide or nucleic acid level of the T H -GM-mediating factor, such as, e.g., STAT5. IL-7, GM-CSF or IL-3, or a combination thereof, wherein an increased level of the T H -GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof relative to a reference level indicates that the patient suffers from a T H -GM-mediated inflammatory disorder; or a comparable level of the T H -GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof relative to a reference level indicates that the patient suffers from a non-T H -GM-mediated inflammatory disorder, thereby classifying the patient suffering from an inflammatory disorder as a T H -GM-mediated inflammatory disorder or a non-T H -GM-mediated inflammatory disorder.
  • the methods disclosed herein can further comprise measuring the polypeptide or nucleic acid level of a factor that mediates a non-T H -GM-mediated inflammatory disorder.
  • factors include, e.g., TNF- ⁇ , IL-6, and IL-1 ⁇ .
  • the present disclosure provides a method of determining a treatment regimen in a patient suffering from an inflammatory disorder.
  • the method comprises quantifying a polypeptide or nucleic acid level of, e.g., activated STAT5 or GM-CSF in a sample collected from a patient suffering from an inflammatory disorder, and quantifying the polypeptide or nucleic acid level of, e.g., TNF- ⁇ in a sample collected from the patient.
  • At least four scenarios can be considered.
  • the activated STAT5 or GM-CSF level is increased (e.g., by at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, or at least 150%) relative to a first reference level and the TNF- ⁇ level is comparable to a second reference level, then the patient is classified as having a T H -GM-mediated inflammatory disorder and the patient can be treated with an agent that modulates T H -GM function, as described herein.
  • the activated STAT5 or GM-CSF level is comparable to the first reference level and the TNF- ⁇ level is increased (e.g., by at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, or at least 150%) relative to the second reference level, then the patient is classified as having a non-T H -GM-mediated inflammatory disorder and the patient can be treated with, e.g., a TNF- ⁇ therapy.
  • the patient is classified as having an inflammatory disorder that is equally T H -GM-mediated and non-T H -GM mediated (e.g., TNF- ⁇ -mediated).
  • the patient can be treated with an effective amount of an agent that modulates T H -GM function and an effective amount of, e.g., a TNF- ⁇ therapy.
  • the combination of both agents can have a synergistic effect.
  • the inflammatory disorder is primarily mediated by the pathway that shows a greater increase. For example, if GM-CSF is increased by 40% relative to a reference level, and TNF- ⁇ is increased by 90% relative to a reference level, then the inflammatory disorder is primarily non-T H -GM-mediated.
  • the patient may receive a combined treatment with an agent that modulates T H -GM function as well a TNF- ⁇ therapy (e.g., anti-TNF- ⁇ therapy), since GM-CSF is increased by, e.g., at least 40% relative to a reference level.
  • a TNF- ⁇ therapy e.g., anti-TNF- ⁇ therapy
  • the first and second reference levels are obtained from the same reference sample.
  • the disclosure also provides a method of tailoring the treatment of a patient suffering from an inflammatory disorder according to the progression of a patient's inflammatory disorder.
  • the first scenario increased T H -GM-mediating factor, e.g. STAT5 or GM-CSF but TNF- ⁇ level is comparable to a reference level
  • the first scenario may indicate that the patient is in an early stage of an inflammatory disorder.
  • na ⁇ ve T cells are stimulated by antigen and programmed by IL-7/STAT5 to differentiate into GM-CSF/IL-3 producing T H -GM cells.
  • T H -GM cytokines e.g., IL-3 and GM-CSF
  • TNF- ⁇ , IL-6, IL-1 ⁇ progressively stimulate more inflammatory cells such as macrophages and neutrophils resulting in the production of, e.g., TNF- ⁇ , IL-6, IL-1 ⁇ , resulting in full-scale inflammation.
  • the second scenario activated STAT5 Or GM-CSF level is comparable to the first reference level and the TNF- ⁇ level is increased
  • the patient is in a late stage of an inflammatory disorder characterized by, e.g., tissue damage.
  • the present disclosure enables the prognosis of a patient depending on the quantifiable level of one or more T H -GM-mediating factor (e.g., STAT5 (e.g., activated phospho-STAT5 (Tyr694)), IL-7, GM-CSF, and/or IL-3) and one or more non-T H -GM-mediating factor (e.g., TNF- ⁇ , IL-6, IL-1 ⁇ ), thereby tailoring the treatment according to the progression of the disease.
  • T H -GM-mediating factor e.g., STAT5 (e.g., activated phospho-STAT5 (Tyr694)
  • IL-7 e.g., activated phospho-STAT5 (Tyr694)
  • IL-7 e.g., IL-7
  • GM-CSF e.g., CD34
  • IL-1 ⁇ e.g., TNF- ⁇ , IL-6, IL-1 ⁇
  • a patient suffering from an inflammatory disorder can be monitored for disease progression to ensure effective and tailored treatment according to the level of one or more T H -GM-mediating factor, as described herein, and one or more non-T H -GM-mediating factor (e.g., TNF- ⁇ , IL-6, IL-1 ⁇ ).
  • T H -GM-mediating factor e.g., TNF- ⁇ , IL-6, IL-1 ⁇ .
  • non-T H -GM-mediating factor e.g., TNF- ⁇ , IL-6, IL-1 ⁇ .
  • the present disclosure also provides a method of prognosing progression of an inflammatory disorder in a patient in need thereof.
  • the method comprises a) quantifying a polypeptide or nucleic acid level of a T H -GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof, in a first sample collected from a patient suffering from an inflammatory disorder, and b) quantifying a polypeptide or nucleic acid level of, e.g., TNF- ⁇ , IL-6, or IL-1 ⁇ , or a combination thereof, in a second sample collected from the patient, wherein i) an increased level of the T H -GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof relative to a first reference level and an unchanged level of TNF- ⁇ , IL-6, or IL-1 ⁇ , or a combination
  • the first sample and the second sample are the same.
  • the present disclosure also provides an isolated population of GM-CSF-secreting T-helper cells (T H -GM).
  • T H -GM cells are differentiated from a precursor cell (e.g., CD4+ cells) in the presence of signal transducer and activator of transcription 5 (STAT5) and/or IL-7, and wherein the T H -GM cells express GM-CSF and IL-3.
  • STAT5 signal transducer and activator of transcription 5
  • IL-7 IL-7
  • the T H -GM cells are differentiated from a precursor cell (e.g., CD4+ cells) in the presence of an agent that inhibits IL-12, IFN- ⁇ . TGF- ⁇ , and/or IL-6.
  • a precursor cell e.g., CD4+ cells
  • the differentiation of a precursor cell (e.g., CD4+ precursor cell) into a T H -GM cell is inhibited by IL-12, IFN- ⁇ , TFG- ⁇ , and/or IL-6.
  • the T H -GM cells are differentiated from a precursor cell in vitro, under artificial conditions, but wherein the T H -GM cells retain physiological properties as described herein.
  • the T H -GM cells are further characterized by an overexpression of one or more genes listed in Table 1.
  • the T H -GM cells are further characterized by an overexpression of, for example, basic helix-loop-helix family, member e40 (BHLHe40), preproenkephalin (PENK), IL-2, serine (or cysteine) peptidase inhibitor, Glade B member 6 h (Serpinb6b), neuritin 1 (Nrn1), stearoyl-Coenzyme A desaturase 1 (Scd1), or phosphotriesterase related C1q-like 3 (Pter), or a combination thereof.
  • the T H -GM cells are further characterized by an underexpression of one or more genes listed in Table 1.
  • the T H -GM cells are further characterized an underexpression of lymphocyte antigen 6 complex, locus A (Ly6a); CD27; or selectin, lymphocyte (Sell).
  • the identification of a distinct network of factors (unique from factors known to mediate T H 1 or T H 17) that mediate T H -GM function enables targeted modulation of T H -GM function to treat T H -GM-mediated disorders, e.g., disorders that result from aberrant T H -GM function.
  • the present disclosure provides a method of modulating T H -GM function, comprising contacting the T H -GM, or cluster of differentiation 4 (CD4+) precursor cells, or both, with a modulating agent that modulates T H -GM function.
  • the modulating agent is contacted with the T H -GM cells or CD4+ precursor cells in vitro or in VIVO.
  • T H -GM function refers to the commitment, development, maintenance, survival, proliferation, or activity, or a combination thereof, of T H -GM cells.
  • an agent that modulates (e.g., enhances or inhibits) T H -GM function is one that modulates T H -GM commitment, development, survival, proliferation, or activity, or combination thereof, of T H -GM cells.
  • T H -GM function can be modulated by modulating its: commitment from a CD4 + precursor T cell; development of a CD4 + precursor cell that has been committed to the T H -GM developmental pathway; maintenance of a T H -GM phenotype; survival or proliferation under development or effector T H -GM cells; and/or activity of effector T H -GM cells (e.g., modulating function of a secreted factor such as GM-CSF or IL-3).
  • a secreted factor such as GM-CSF or IL-3
  • a modulation in T H -GM function includes, but is not limited to, a modulation in: the number of T H -GM cells; the survival of T H -GM cells; the proliferation of T H -GM cells; and/or the activity of T H -GM cells.
  • the activity of T H -GM cells herein includes the activity induced by the cytokines, chemokines, growth factors, enzymes and other factors secreted by T H -GM cells, as described herein, and the activity induced by direct contact with T H -GM cells.
  • a T helper subset cell “T H -GM” refers to a cell that, similar to T H 1 and T H 17 cells, differentiates from precursor CD4+ precursor cells, but which commits and develops through a pathway that is mediated by a subset of factors (the T H -GM-mediating factors) that is distinct and unique from the known subset of factors that commit and develop T H 1 or T H 17 cell subtypes, as described herein.
  • a T H -GM cell produces a distinct and unique set of genes (see, e.g., Table 1) and effects pathogenicity through a different mechanism and pathway than the known factors that mediate pathogenicity of T H 1 or T H 17 cell subtypes.
  • a T H -GM cell commits and develops by IL-7/STAT5 function (its regulators), and effects pathogenicity by GM-CSF/IL-3 (its effectors).
  • the present disclosure provides a method of treating a T H -GM-mediated inflammatory disorder in a patient in need thereof, comprising administering to said patient an effective amount of a modulating agent that modulates T H -GM cell function.
  • the patient is previously diagnosed as having a T H -GM-mediated inflammatory disorder, as described herein.
  • the present disclosure also provides a method of treating rheumatoid arthritis in a patient who exhibits limited response to TNF- ⁇ therapy, comprising administering to said patient an effective amount of a modulating agent that modulates T H -GM function.
  • limited response refers to no response or insignificant response such that a patient is not treated by the therapy, as determined by clinical standards.
  • Treatment refers to therapy, prevention and prophylaxis and particularly refers to the administration of medicine or the performance of medical procedures with respect to a patient, for either prophylaxis (prevention) or to reduce the extent of or likelihood of occurrence of the condition or event in the instance where the patient is afflicted. It also refers to reduction in the severity of one or more symptoms associated with the disease or condition. In the present application, it may refer to amelioration of one or more of the following: pain, swelling, redness or inflammation associated with an inflammatory condition or an autoimmune disease.
  • treatment is an approach for obtaining beneficial or desired results, including clinical results.
  • beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms, diminishment of extent of disease, stabilized (e.g., not worsening) state of disease, delay or slowing of disease progression, and/or amelioration or palliation of the disease state. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • an “effective amount” of an agent is that amount sufficient to effect beneficial or desired results, including clinical results.
  • An “effective amount” depends upon the context in which it is being applied. In the context of administering a composition that modulates an autoimmune response, an effective amount of an agent which is a modulator of T H -GM function is an amount sufficient to achieve such a modulation as compared to the response obtained when there is no agent administered.
  • An effective amount can confer immediate, short term or long term benefits of disease modification, such as suppression and/or inhibition of T H -GM function, as defined herein.
  • An effective amount can be administered in one or more administrations.
  • An “effective amount” as used herein, is intended to mean an amount sufficient to reduce by at least 10%, at least 25%, at least 50%, at least 75%, or an amount that is sufficient to cause an improvement in one or more clinically significant symptoms in the patient.
  • the modulating agent inhibits T H -GM function to, e.g., reduce inflammation.
  • the inhibition conferred by the modulating agent does not imply a specific mechanism of biological action.
  • the term “antagonist” or “inhibitor” as used herein includes all possible pharmacological, physiological, and biochemical interactions with factors that mediate T H -GM function (e.g., IL-7, IL-7 receptor, STAT5, GM-CSF, IL-3, IL-2, IL-2 receptor, PENK, RANKL, JAK1/3, or any of the genes that are differentially expressed in T H -GM cells, e.g., genes in Tables 1 and 2), whether direct or indirect, and includes interaction with a factor (or its active fragment) that mediates T H -GM function at the protein and/or nucleic acid level, or through another mechanism.
  • a modulating agent that inhibits T H -GM function includes an antibody, a polypeptide (e.g., a soluble receptor that hinds and inhibits, for example, IL-7), a small molecule, a nucleic acid (e.g., antisense, small interfering RNA molecule, short hairpin RNA, microRNA), or a protein (e.g., cytokine), or a combination thereof that prevents the function (e.g., expression and/or activity) of a factor that mediates T H -GM function.
  • a polypeptide e.g., a soluble receptor that hinds and inhibits, for example, IL-7
  • a small molecule e.g., a nucleic acid (e.g., antisense, small interfering RNA molecule, short hairpin RNA, microRNA), or a protein (e.g., cytokine), or a combination thereof that prevents the function (e.g., expression and/or
  • binds is used interchangeably with “specifically binds,” which means a polypeptide (e.g., a soluble receptor) or antibody which recognizes and hinds a polypeptide of the present disclosure, but that does not substantially recognize and bind other molecules in a sample, for example, a biological sample, which naturally includes a polypeptide of the present disclosure.
  • an antibody specifically binds an activated STAT5 polypeptide does not hind a non-STAT5 polypeptide.
  • antibody refers to an intact antibody or antigen-binding fragment of an antibody, including an intact antibody or antigen-binding fragment that has been modified or engineered, or that is a human antibody.
  • the antibody binds to and inhibits the function of any one or more of the factors that mediate T H -GM function.
  • the antibody binds to and inhibits the function of a gene listed in Table 1.
  • the antibody hinds to and inhibits the protein or any functional fragment thereof.
  • Methods of designing, producing and using suitable antibodies are known and available to those of skill in the art. Examples of antibodies suitable for use in the present disclosure include. e.g., daclizumab, basiliximab, methosimumab, MOR103, KB003, namilumab, and MOR Ab-022.
  • protein and “polypeptide” are used interchangeably, and can include full-length polypeptide or functional fragments thereof (e.g., degradation products, alternatively spliced isoforms of the polypeptide, enzymatic cleavage products of the polypeptide), the polypeptide hound to a substrate or ligand, or free (unbound) forms of the polypeptide.
  • functional fragment refers to a portion of a full-length protein that retains some or all of the activity (e.g., biological activity, such as the ability to bind a cognate ligand or receptor) of the full-length polypeptide.
  • the modulating agent that inhibits T H -GM function can be a particular biological protein (e.g., cytokines) that inhibits, directly or indirectly, one or more of the factors that mediate T H -GM function.
  • cytokines include, e.g., IL-12, IFN- ⁇ , TGF- ⁇ , and IL-6.
  • the modulating agent that inhibits T H -GM function can be a small molecule that inhibits, directly or indirectly, one or more of the factors that mediate T H -GM function.
  • a “small molecule” is an organic compound or such a compound complexed with an inorganic compound (e.g., metal) that has biological activity and is not a polymer.
  • a small molecule generally has a molecular weight of less than about 3 kilodaltons. Examples of known small molecules include CAS 285986-31-4 (Calbiochem), pimozide, and tofacitinib.
  • the modulating agent enhances T H -GM function in disorders such as, e.g., viral, fungal and bacterial infections, cancers and/or conditions associated with therewith.
  • modulating agents that enhance T H -GM function include. e.g., CD28 activator; IL-7 and/or IL-2 on na ⁇ ve (precursor) CD4 + T cells; activator of STAT5; or effectors of T H -GM cells (e.g., GM-CSF. IL-3).
  • the present disclosure provides a method of treating a STAT5-mediated inflammatory disorder in a patient in need thereof, comprising administering to the patient an effective amount of an agent that modulates STAT5 function.
  • STAT5-mediated inflammatory disorder refers to an inflammatory disorder that is caused by aberrant STAT5 function (aberrantly enhanced or inhibited), and which is responsive to modulation of STAT5 function, as determined by clinical standards.
  • the STAT5 is activated STAT5 (e.g., phospho-STAT5, Tyr694).
  • the inflammatory disorder is an autoimmune disorder.
  • the inflammatory disorder can be any inflammatory disorder mediated by STAT5 (e.g., activated STAT5), and includes, but is not limited to rheumatoid arthritis, multiple sclerosis, ankylosing spondylitis. Crohn's disease, diabetes. Hashimoto's thyroiditis, hyperthyroidism, hypothyroidism, Irritable Bowel Syndrome (IBS), lupus erythematosus, polymyalgia rheumatic, psoriasis, psoriatic arthritis. Raynaud's syndrome/phenomenon, reactive arthritis (Reiter syndrome), sarcoidosis, scleroderma. Sjögren's syndrome, ulcerative colitis, uveitis, or vasculitis.
  • STAT5 e.g., activated STAT5
  • STAT5 e.g., activated STAT5
  • IBS Irritable
  • the term “patient” refers to a mammal, preferably human, but can also include an animal in need of veterinary treatment, e.g., companion animals (e.g., dogs, cats, and the like), farm animals (e.g., cows, sheep, pigs, horses, and the like), and laboratory animals (e.g., rats, mice, guinea pigs, and the like).
  • companion animals e.g., dogs, cats, and the like
  • farm animals e.g., cows, sheep, pigs, horses, and the like
  • laboratory animals e.g., rats, mice, guinea pigs, and the like.
  • the agent inhibits STAT5 function (e.g., expression and/or activity).
  • STAT5 function e.g., expression and/or activity.
  • agents that inhibit STAT5 e.g., activated STAT5, Tyr694 are described herein.
  • the methods of the present disclosure further comprise administering to the patient a TNF- ⁇ c therapy.
  • TNF- ⁇ therapy is administered in a patient determined to have an inflammatory condition that is non-T H -GM-mediated.
  • a TNF- ⁇ therapy is administered if a quantified TNF- ⁇ level is increased by, e.g., at least 40% relative to a reference level.
  • TNF- ⁇ therapy examples include those that are TNF- ⁇ -inhibitor based, and those that are non-TNF- ⁇ -inhibitor based.
  • TNF- ⁇ -inhibitor based therapy includes etanercept, adalimuinab, infliximab, golimumab, and certolizumab pegol.
  • non-TNF- ⁇ -inhibitor based therapy includes corticosteroid medications (e.g., prednisone), nonsteroidal anti-inflammatory drugs (e.g., methotrexate), and JAK inhibitors (e.g., tofacitinib).
  • Other examples of non-TNF- ⁇ -inhibitor based therapy include anakinra, abatacept, rituximab and tocilizumab.
  • the TNF- ⁇ therapy can be administered before, simultaneously with, or alter the administration of an effective amount of an agent that modulates T H -GM function.
  • an agent that modulates T H -GM function and the TNF- ⁇ therapy can be administered together in a single dose, or can be administered in separate doses, e.g., either simultaneously or sequentially, or both.
  • the duration of time between the administration of an agent that modulates T H -GM function and a TNF- ⁇ therapy will depend on the nature of the therapeutic agent(s).
  • an agent that modulates T H -GM function and a TNF- ⁇ therapy may or may not be administered on similar dosing schedules.
  • the agent that modulates T H -GM function and the TNF- ⁇ therapy may have different half-lives and/or act on different time-scales such that the agent that modulates T H -GM function is administered with greater frequency than the TNF- ⁇ therapy, or vice-versa.
  • the number of days in between administration of therapeutic agents can be appropriately determined by persons of ordinary skill in the art according to the safety and pharmacodynamics of each drug.
  • T H -GM cells as well as the identification of genes differentially produced by T H -GM cells relative to T H 1 or T H 17 enables the use of T H -GM cells to identify novel therapeutics for modulating T H -GM function, thereby enabling new therapeutics for treating T H -GM-mediated disorders (e.g., inflammatory disorders).
  • T H -GM-mediated disorders e.g., inflammatory disorders
  • the present disclosure provides a method of screening to identify a modulator of T H -GM cell function, comprising contacting an isolated population of T H -GM cells, or an isolated population of CD4+ precursor cells, with a candidate agent, and measuring a readout of T H -GM function in the presence or absence of the candidate agent, wherein a change in the readout of T H -GM function indicates that the candidate agent is a modulator of T H -GM function.
  • a candidate agent refers to an agent that may modulate T H -GM function by modulating the function (e.g., expression and/or activity) of a factor that mediates T H -GM function.
  • candidate agents include, e.g., an antibody, a peptide, a small molecule, a nucleic acid (e.g., antisense, small interfering RNA molecule), or a protein (e.g., cytokine), or a combination thereof.
  • a candidate agent can be designed to target any of the factors (at the protein and/or nucleic acid level) that mediate T H -GM function, as described herein, including the genes listed in Table 1 (e.g., genes preferentially upregulated in T H -GM cells, genes preferentially overexpressed/underexpressed on the surface of T H -GM cells).
  • readout refers to any change (or lack of change) in T H -GM function that can be measured or quantified.
  • a candidate agent can be assessed for its effect on, e.g., GM-CSF secretion by T H -GM cells, or its effect on the abundance of T H -GM cells (through an effect on the commitment/development/proliferation of T H -GM cells), as described herein.
  • Assays for determining such readouts are known and available in the art, and are exemplified herein.
  • the change in the presence of the candidate agent is a reduction in the measurement of the readout, indicating an inhibition of T H -GM function (e.g., decrease in GM-CSF or IL-3 production, or decrease in the abundance of T H -GM cells), thereby identifying the candidate agent as an inhibitor of T H -GM function.
  • an inhibition of T H -GM function e.g., decrease in GM-CSF or IL-3 production, or decrease in the abundance of T H -GM cells
  • the change in the presence of the candidate agent is an increase in the measurement of the readout, indicating an enhancement of T H -GM function (e.g., increase in GM-CSF or IL-3 production, or increase in the abundance of T H -GM cells), thereby identifying the candidate agent as an enhancer of T H -GM function.
  • an enhancement of T H -GM function e.g., increase in GM-CSF or IL-3 production, or increase in the abundance of T H -GM cells
  • the readout can be any one or more of the genes listed in Tables 1 and 2 which are preferentially upregulated or downregulated in T H -GM cells.
  • a candidate agent that downregulates a gene that is preferentially upregulated in a T H -GM cell is a inhibitor of T H -GM function.
  • a candidate agent that upregulates a gene that is preferentially downregulated in a T H -GM cell is an enhancer of T H -GM function.
  • the method of screening if performed with precursor CD4+ cells, is performed under T H -GM polarizing conditions, as described herein.
  • the method can be performed in the presence of IL-7/STAT5, TCR activation, CD28 co-stimulation, in combination with the blockade of IFN-gamma and IL-4.
  • the practice of the present disclosure includes use of conventional techniques of molecular biology such as recombinant DNA, microbiology, cell biology, biochemistry, nucleic acid chemistry, and immunology as described for example in: Molecular Cloning: A Laboratory Manual, second edition (Sambrook et al., 1989) and Molecular Cloning: A Laboratory Manual, third edition (Sambrook and Russel, 2001), jointly and individually referred to herein as “Sambrook”); Oligonucleotide Synthesis (M. J. Gait, ed., 1984); Animal Cell Culture (R. I. Freshney, ed., 1987); Handbook of Experimental Immunology (D. M. Weir & C. C.
  • Stat5 f/f mice were provided by L. Hennighausen (National Institute of Diabetes and Digestive and Kidney Diseases).
  • Stat3 f/f mice were generated as described 2 .
  • Cd4-Cre transgenic mice were purchased from Taconic Farms.
  • Rag2 ⁇ / ⁇ mice were obtained from Jean-Pierre Abastado (Singapore Immunology Network). All mice are on a C57BL/6 genetic background and housed under specific-pathogen-free conditions at National University of Singapore. All experiments were performed with mice 6 ⁇ 8 weeks old and approved by the Institutional Animal Care and Use Committee of NUS.
  • CD4 + T cells were obtained from spleens and lymph nodes by positive selection and magnetic separation (Miltenyi Biotech), followed by purification of na ⁇ ve CD4 + T cell population (CD4 + CD25 ⁇ CD62L hi CD44 lo ) sorted with FACS Aria.
  • Na ⁇ ve CD4 + T cells were stimulated with plate-hound anti-CD3 (3 ⁇ g/ml; BD Pharmingen) and anti-CD28 (1 ⁇ g/ml; BD Pharmingen) in presence of different combinations of neutralizing antibodies and cytokines for 3 ⁇ 4 days: for neutral conditions, no addition of any cytokine or neutralizing antibody; for T H 1 conditions, IL-12 (10 ng/ml), and anti-IL-4 (10 ⁇ g/ml, BD Pharmingen); for T H 17 conditions, hTGF- ⁇ (3 ng/ml), IL-6 (20 ng/ml), anti-IFN- ⁇ (10 ⁇ g/ml, eBioscience), and anti-IL-4 (10 ⁇ g/ml); for an alternative T H 17 conditions, IL-6 (20 ng/ml), IL-23 (10 ng/ml), IL-1 (10 ng/ml), anti-IFN- ⁇ (10 ⁇ g/ml), and anti-IL-4 (10 ⁇ g/ml
  • na ⁇ ve CD4 + T cells were stimulated with plate-bound anti-CD3 (2 ⁇ g/ml) and soluble anti-D28 (1 ⁇ g/ml) with the addition of IL-7 and/or anti-IFN- ⁇ (10 ⁇ g/ml) as indicated. All cytokines were obtained from R&D Systems. All cells were cultured in RPMI 1640 supplemented with 10% FBS, 100 units/ml penicillin, 0.1 mg/ml streptomycin, 1 mM sodium pyruvate, 0.1 mM nonessential amino acid and 5 ⁇ M beta-mercaptoethanol.
  • EAE induction procedures were modified from previous report 3 .
  • mice were immunized in two sites on the hind flanks with 300 ⁇ g MOG 35-55 in 100 ⁇ l CFA containing 5 mg/ml heat-killed M. tuberculosis strain H37Ra (Difco) at day 0 and day 7.
  • Pertussis toxin List Bio Lab
  • the similar procedure was performed at day 0 and day 1 only.
  • LPS 600 ⁇ g/ml in IFA, O111:B4 from Sigma
  • Clinical symptoms were scored as follows: 0, no clinical sign; 1, loss of tail tone; 2, wobbly gait; 3, hind limb paralysis; 4, hind and fore limb paralysis; 5, death.
  • IL-7R ⁇ neutralizing antibody (SB/14, BD Pharmingen) and isotype control was administrated intraperitoneally at 200 ⁇ g per mouse every other day.
  • SB/14 BD Pharmingen
  • isotype control was administrated intraperitoneally at 200 ⁇ g per mouse every other day.
  • CNS-infiltrating cells both spinal cord and brain were collected and minced from perfused mice, and mononuclear cells were isolated by gradient centrifuge with Percoll (GE Healthcare).
  • splenocytes and LNs were harvested 10-14 days post-immunization and passed through a 70 ⁇ m cell strainer (BD Falcon). Cells were cultured in vitro for 3 days with MOG 35-55 (20 ⁇ g/ml) in the presence of IL-23 (5 ng/ml) and IL-1 ⁇ (2 ng/ml). After harvesting, CD4 + T cells were purified by positive selection to a purity >90%. CD4 + T cells (2 million in sterile PBS) were injected intraperitoneally into Rag2 ⁇ / ⁇ mice, followed by Pertussis toxin administration on the following day.
  • T H mice were observed daily for the signs of EAE as described above.
  • EAE induction by transferring various T H subsets similar procedures was performed as described above. Different subsets skewing conditions were as follows: Non-skewed, MOG 35-55 only; T H 1: MOG 35-55 plus IL-12 (10 ng/ml) and anti-IL-4 (5 ⁇ g/ml); T H 17: MOG 35-55 plus TGF- ⁇ (3 ng/ml), IL-6 (10 ng/ml), anti-IFN- ⁇ (5 ⁇ g/ml) and anti-IL-4 (5 ⁇ g/ml); GM-CSF-expressing T H : MOG 35-55 plus IL-7 (2 ng/ml) and anti-IFN- ⁇ (5 ⁇ g/ml). 6 ⁇ 10 5 CD4 + T cells were transferred per recipient mouse.
  • AIA Antigen-Induced Arthritis
  • mice were immunized subcutaneously in two sites on the hind flanks with 100 ⁇ g methylated bovine serum albumin (mBSA, Sigma) in 100 ⁇ l complete Freund's adjuvant (CFA) containing 5 mg/ml heat-killed M. tuberculosis strain H37Ra (Difco) at day 0.
  • CFA complete Freund's adjuvant
  • Pertussis toxin List Bio Lab was administrated intraperitoneally at the dosage of 250 ng per mouse at day 1.
  • Arthritis was induced by intraarticular injection of 100 ⁇ g mBSA (in 10 saline) into the hind right knee joint at day 7 after immunization. The hind left knee joint was injected with same volume of saline as control.
  • splenocytes and inguinal LN cells were isolated from mBSA/CFA-immunized mice at day 7, and cultured in vitro with mBSA (10 ⁇ g/ml) in the presence of IL-7 (2 ng/ml) for 3 days. After harvesting, CD4 + T cells were purified by positive selection (Miltenyi Biotec) to a purity >90%. Then CD4 + T cells (1 million in sterile PBS) were transferred into WT na ⁇ ve mice, followed by intraarticular injection of mBSA on the next day.
  • CIA was induced in a similar procedure as AIA as described above, by immunizing mice with chicken collagen II/CFA emulsion (purchased from Chondrex, Inc), followed with pertussis toxin injection. Mice were monitored and scored for arthritis; 0, normal; 1, mild swelling of ankle or wrist, or apparent swelling limited to individual digits; 2, moderate swelling of entire paw; 3, severe swelling of entire paw with ankylosis. Scores for four limbs were summed for each mouse.
  • Sections (10 ⁇ m) were fixed in ice-cold acetone and stained with primary anti-CD4 (Biolegend) and anti-CD11b (eBioscience), followed by incubation with fluorescence-conjugated secondary antibodies (Invitrogen).
  • ALA experiments knee joint were fixed in 10% formalin for 5 days, followed by decalcification in 5% formic acid for 5 clays.
  • Sections (10 ⁇ m) were stained with hematoxylin and eosin (H&E) to assess immune cell infiltration and inflammation, or stained with Safranin-O/fast green to access cartilage destruction.
  • H&E hematoxylin and eosin
  • Monocytes/macrophages (Ly6C hi Ly6G ⁇ ) and neutrophils (Ly6C lo Ly6G hi ) gated on CD45 + CD11b + were sorted with FACS Aria from spleens or synovial single cell suspensions. Sorted cells were cytospun onto glass slides and subsequently stained with May Grünwald and Giemsa dye following a standard procedure.
  • Complementary DNA (cDNA) was synthesized with Superscript reverse transcriptase (Invitrogen). Gene expressions were measured by 7500 real-time PCR system (Applied Biosystems) with SYBR qPCR kit (KAPA). Actinb, Gapdh or Rn18S was used as internal control. The primer sequences are available upon request.
  • TNF- ⁇ , IL-6, IL-1 ⁇ , IFN- ⁇ , GM-CSF and IL-2 levels were assayed by Ready-SET-Go ELISA kit (eBioscience), and IL-17 level was measured by DuoSet ELISA kit (R&D Systems) according to the manufactures' instructions.
  • Cell lysates were fragmented by sonication and precleared with protein G Dynabeals, and subsequently precipitated with anti-STAT5 antibody (Santa Cruz) or normal rabbit IgG (Santa Cruz) overnight at 4° C.
  • crosslink reversal was clone by incubating at 65° C. for 8 hr.
  • the eluted DNA was purified and analyzed by RT-PCR with primers specific to C42 promoter as described previously 5 .
  • STAT5 negatively regulates T H 17 differentiation by restraining IL-17 production (Laurence et al., 2007; Yang et al., 2011). However, the function of STAT5 in T H 17-mediated pathogenesis is not well understood.
  • EAE was induced in Cd4-Cre; Stat5 f/f (Stat5 ⁇ / ⁇ ) mice, where Stat5 was specifically deleted in T cell compartment, and in littermate controls by immunizing the mice with MOG 35-55 /CFA at day 0 and day 7. Development of paralysis was assessed by daily assignment of clinical scores. Surprisingly, diminished occurrence and severity of clinical disease in Stat5 ⁇ / ⁇ mice was observed ( FIGS.
  • FIGS. 1A and 1B a result that was opposite to expectations based on an antagonistic role for STAT5 in T H 17 generation. Similar results were observed when a single MOG 35-55 /CFA immunization was performed or replaced CFA with LPS as the adjuvant to induce EAE ( FIGS. 1C and 1D ). Consistent with reduced EAE disease in Stat5 ⁇ / ⁇ mice, a remarkable reduction of immune cell infiltration in the spinal cord of Stat5 ⁇ / ⁇ mice was observed ( FIG. 2A ). Furthermore, the infiltration of various immune cell populations, including CD4 + , CD8 + . B220 + and CD11b + cells was reduced in Stat5 ⁇ / ⁇ mice ( FIGS. 2B-D and data not shown).
  • Stat5 deletion (Cd4-cre; Stat5 f/ ⁇ ) mice was reported to develop peripheral lymphopenia, with a reduction of both CD4 + and CD8 + T cells (Yao et al., 2006). However, another study showed that Stat5 deletion (Cd4-cre; Stat5 f/f ) did not affect the proportion of peripheral CD4 + T cells (Burchill et al., 2007). In the experimental setting, a change in the absolute number of peripheral CD4 + T cells was not detected by Stat5 deletion during EAE development ( FIGS. 4A and 4B ), suggesting the resistance to EAE in Stat5 ⁇ / ⁇ mice was not caused by peripheral lymphopenia.
  • mice receiving Stat5 +/+ CD4 + T cells spontaneously developed EAE disease 1 week after transfer.
  • mice receiving Stat5 ⁇ / ⁇ CD4 + T cells had significantly reduced disease severity and incidence.
  • the frequencies of IL-17 + and IFN- ⁇ + cells among CD4 + T cells in the CNS of Rag2 ⁇ / ⁇ mice were comparable between two groups ( FIG.
  • GM-CSF production was impaired by Stat5 deletion, its expression was examined in MOG 35-53 -specific Stat5 +/+ and Stat5 +/+ CD4 + T cells.
  • Splenocytes derived from MOG 35-55 /CFA-immunized Stat5 +/+ and Stat5 ⁇ / ⁇ mice were challenged with various concentrations of MOG 35-55 for 24 h, to examine the secretion of GM-CSF.
  • GM-CSF production was observed to increase in a MOG 35-55 dose-dependent manner in Stat5 +/+ cells ( FIG. 8A ). In contrast, GM-CSF production was severely diminished in Stat5 +/ ⁇ cells in all conditions.
  • splenocytes derived from mice were stimulated during the development of EAE with PMA/Ionomycin in the presence of GolgiPlug for GM-CSF and IL-17 intracellular staining.
  • IL-17 expression was enhanced in Stat5 ⁇ / ⁇ cells, a significantly reduced proportion of GM-CSF + IL-17′′ and GM-CSF + IL-17 + cells was observed among CD4 + CD44 hi cells in the absence of STAT5 ( FIG. 8B ).
  • the frequency of MOG 35,55 -specific GM-CSF + T cells was also significantly reduced in spleens of Stat5 ⁇ / ⁇ mice ( FIG. 8C ). Together, these results indicate that STAT5 is required for GM-CSF expression in autoreactive CD4 + T cells.
  • STAT5 an important transcription factor in T H 17 differentiation, was required for GM-CSF expression ( FIG. 8D ).
  • GM-CSF induction in the CNS during EAE development was examined. Although IL-17 and IFN- ⁇ production by CNS-infiltrating CD4 + T cells was not impaired by Stat5 deficiency, a diminished frequency of CD4 + GM-CSF + cells in the CNS of Stat5 ⁇ / ⁇ mice was detected compared with control mice ( FIG. 9A ). Further analysis showed a reduced GM-CSF + percentage among CD4 + IL-17; cells and among CD4 + IFN- ⁇ + cells ( FIG. 9A ).
  • FIG. 9B GM-CSF mRNA expression in the CNS of Stat5 ⁇ / ⁇ mice was markedly lower than that of Stat5 +/+ mice at day 8 after EAE induction ( FIG. 9C ), when comparable CD4 + T cell infiltration was detected in Stat5 ⁇ / ⁇ and Stat5 +/+ mice ( FIGS. 5C and 5D ). Meanwhile, no significant difference of IL-17 and IFN- ⁇ expression was detected between Stat5 ⁇ / ⁇ and Stat5 +/+ mice ( FIG. 9C ).
  • IL-7 carried out this function in a STAT5-dependent manner, as Stat5 deletion abrogated its effect on GM-CSF expression as assessed by intracellular cytokine staining and ELISA ( FIG. 11A , lower panels, and FIG. 11B ).
  • IL-7R ⁇ is expressed in both CD62L hi CD44 lo T cells and CD62 lo CD44 hi T cells, suggesting IL-7 may directly act on CD4 + T cells to regulate GM-CSF expression.
  • CD62L hi CD44 lo and CD62L lo CD44 hi T cells were sorted from Stat5 ⁇ / ⁇ mice and littermate controls during EAE development, and then activated cells in the presence or absence of IL-7.
  • CD62L lo CD44 hi T cells potently expressed GM-CSF
  • STAT5 deletion resulted in reduced basal GM-CSF production in CD62L lo CD44 hi T cells.
  • IL-7 promoted GM-CSF expression in both subsets of CD4 + T cells in a STAT5-dependent manner ( FIG. 11C ).
  • mice were treated with IL-7R ⁇ -specific antibody (clone SB/14) during EAE development.
  • the treatment resulted in a significant reduction of disease severity, which was accompanied with reduced CNS inflammation ( FIGS. 12A and 12B ).
  • this neutralizing antibody did not have T cell depleting activity ( FIG. 12C ).
  • the blocking of IL-7 signaling resulted in decreased GM-CSF expression in CNS-infiltrating CD4 + T cells ( FIGS. 120-12F ).
  • the present findings indicate that IL-7 induces STAT5-dependent GM-CSF expression in autoreactive CD4 + T cells, which contributes to the development of neuroinflammation.
  • Example 5 GM-CSF-Expressing T H Cells are Distinct from T H 17 and T H 1
  • T H 17 and T H 1 can produce GM-CSF, it was determined if the IL-7-stimulated phenotype was related to either of these subsets.
  • na ⁇ ve CD4 + T cells were stimulated with plate-bound anti-CD3 and soluble anti-CD28 under T H 1- or T H 17-polarizing conditions. It was observed that anti-CD3 together with anti-CD28 induced the expression of GM-CSF ( FIG. 14A ).
  • T H 1 differentiation conditions promoted IFN- ⁇ expression
  • T H 17 conditions promoted IL-17 expression as expected
  • both T H 1 and T H 17 differentiation conditions greatly suppressed the production of GM-CSF ( FIGS.
  • FIG. 13A and 13B Conversely, IL-12 and IFN- ⁇ neutralization promoted GM-CSF-expressing cell generation ( FIG. 13A ), consistent with a previous report (Codarri et al., 2011). IL-23 and IL-1 ⁇ did not increase GM-CSF-expressing cell differentiation from na ⁇ ve CD4 + T cells ( FIG. 13A ), which was consistent with the finding that na ⁇ ve CD4 + T cells did not express their receptors. TGF- ⁇ inhibits GM-CSF expression (El-Behi et al., 2011). IL-6, an essential cytokine for T H 17 differentiation, had a profound inhibitory effect on GM-CSF expression ( FIG.
  • the present findings disclosed herein (including .g., diminished GM-CSF expression in Stat5 ⁇ / ⁇ CD4 + T cells in vivo, IL-7/STAT5-mediated induction of GM-CSF expression in na ⁇ ve CD4 + T cells, and the distinct features of GM-CSF-expressing T H cells versus T H 1 and T H 17 cells) indicates a distinct T H cell subset that is regulated by IL-7-STAT5 signaling.
  • This finding was further explored by examining GM-CSF-expressing T H cell differentiation in vitro by activating na ⁇ ve CD4 + T cells with anti-CD3 and anti-CD28 in the presence of different concentrations of IL-7. As shown in FIGS.
  • IL-7 strongly promoted the generation of GM-CSF-expressing cells and GM-CSF secretion. Moreover, the generation of GM-CSF-expressing T H by IL-7 was mediated by STAT5. Without STAT5, IL-7 was unable to promote the generation of GM-CSF-expressing cells ( FIGS. 15C and 15D ). Further investigation showed that IL-7-induced STAT5 activation directly hound promoter regions of the Csf2 gene ( FIGS. 15E and 15F ).
  • T H -GM novel GM-CSF-expressing helper T cell subset
  • IL-2 signaling could influence T H -GM differentiation from na ⁇ ve CD4 + T cells.
  • the addition of IL-2 or antibody against IL-2 only had modest effect on the frequency of GM-CSF + cells ( FIG. 14D ), indicating a minimal effect of IL-2 on T H -GM differentiation.
  • IL-2 high-affinity receptor IL-2Ra was not expressed in na ⁇ ve CD4 + T cells, but its expression was gradually induced by TCR activation ( FIGS. 17A-17C ).
  • the minimal effect of IL-2 at least in part is due to the unresponsiveness of na ⁇ ve CD4 + T cells to IL-2 stimulation.
  • IL-7 induced STAT5 activation and upregulated GM-CSF mRNA expression in na ⁇ ve CD4 + T cells ( FIGS. 17D and 17E ).
  • activated CD4 + T cells were stimulated with IL-2 or IL-7, which showed that both cytokines induced STAT5 activation, Csf2 promoter binding and GM-CSF mRNA upregulation ( FIGS. 18A-18C ).
  • IL-2 induced a prolonged STAT5 activation compared with IL-7 ( FIG. 18A ).
  • T H -GM as distinct from known T cell subsets (e.g., T H 1 and T H 17)
  • a whole transcriptome analysis was performed by microarray to validate its specificity compared with known T cell subsets, in particular T H 17 cells.
  • Na ⁇ ve CD4 + T cells were differentiated into T H 1, T H 17 and T H -GM.
  • Microarray analysis was performed to examine their gene expression profiles.
  • Whole transcriptome clustering indicates T H -GM cells as representing a novel subset distinct from T H 1 or T H 17 cells. T cell lineage-specific gene expression is shown in Table 1.
  • T H 1 cells A list of 202 genes preferentially expressed in T H 1 cells were identified, compared with na ⁇ ve, T H 17 or T H -GM cells (fold change >1.7), among which IFN- ⁇ and T-bet are on the top of the list (Table 1).
  • T H 17-feature genes such as IL-17, IL-17F, ROR ⁇ t and ROR ⁇ , were identified in the list including 411 genes specific to T H 17 cells (Table 1).
  • the T H -GM cell-specific gene list (“Genes preferentially upregulated in T H -GM” the T H -GM signature genes) contains 210 genes including the gene encoding GM-CSF as the top gene in the list (Table 1).
  • T H -GM signature genes can be used for further characterization by surface markers to identify the T H -GM subset of T cells.
  • genes of interest were also identified, including genes encoding cytokines and transcriptional factors, in particular IL-3.
  • helper T cells were differentiated in vitro and confirmed that T H -GM cells are potent IL-3 producers as compared with T H 1 and T H 17 cells ( FIGS. 20A, 20C and 20D ).
  • cytokines including EBI-3, PENL and RANKL were found preferentially expressed in T H -GM cells ( FIG. 20B ), indicating diverse biological functions of T H -GM cells.
  • T H -GM Cells are the Primary Pathogenic Population
  • T H subset (T H -GM) was the primary encephalitogenic effector cells
  • adoptive transfer of different subsets of MOG 35-55 -specific CD4 + T cells was performed into Rag2 ⁇ / ⁇ mice for EAE induction.
  • GM-CSF-expressing T H cells were preferentially able to induce EAE compared with T H 17 and T H 1 subsets.
  • Example 9 The Suppression of STAT5 Activity by Chemical Inhibitor Attenuates GM-CSF Expression by T H -GM and Ameliorates EAE
  • the effect of disrupting STAT5 activation by chemical inhibitor was examined to explore possible methods of treating autoimmune neuroinflammation.
  • the phosphorylation on the key tyrosine residue in SH2 domain is crucial for STAT5 activation and function.
  • a commercial STAT5 inhibitor (CAS 285986-31-4, Calbiochem) has been reported to selectively disrupt tyrosine phosphorylation and DNA binding of STAT5 (Muller et al., 2008).
  • the inhibitory effect of this inhibitor on STAT5 activation upon IL-7 stimulation in CD4+ T cells was tested. At a concentration of 50 ⁇ M, the inhibitor had about 50% inhibitory effect, which was further enhanced with the increase of concentration ( FIG. 22A ).
  • STAT5 inhibitor had low affinity and thus required a high concentration to fully block STAT5 activation, whereas JAK3 inhibitor showed potent inhibitory effect even at low concentration ( FIG. 22B ).
  • the specificity of STAT5 inhibitor was next tested by examining its effect on the activation of STAT3 and STAT1. As shown in FIGS. 22C and 22D , this STAT5 inhibitor at relatively lower concentration (50 or 100 ⁇ M) showed minimal inhibitory effect on both STAT3 and STAT1 activation.
  • STAT5 inhibition The effect of STAT5 inhibition on T H -GM differentiation was examined. As shown, STAT5 inhibitor suppressed T H -GM differentiation in a dosage-dependent manner ( FIG. 22E ). Reduced T H 1 differentiation upon STAT5 inhibitor treatment was observed (data not shown), but T H 17 differentiation was not suppressed by STAT5 inhibitor (data not shown).
  • STAT5 inhibition ameliorated EAE severity, associated with reduced immune cell infiltration in the CNS ( FIGS. 23A and 23B ).
  • JAK3 inhibitor can potently block STAT5 activation ( FIG. 22B ), it showed detrimental effect on EAE ( FIG. 23B ).
  • STAT5 inhibitor resulted in reduced GM-CSF production in CNS-infiltrating CD4 + T cells ( FIGS. 23C and 23D ). This study indicates that targeting STAT5 by chemical inhibitor is useful in therapeutic intervention in MS.
  • Plasma concentrations of GM-CSF and TNF- ⁇ in peripheral blood of RA patients were examined in comparison with gender/age-matched healthy control (HC), and found that both cytokines were elevated in RA ( FIG. 24A ).
  • Ex vivo frequencies of IFN- ⁇ -, IL-17- or GM-CSF-producing T H cells were quantified in RA and HC. High frequencies of IFN- ⁇ - and/or GM-CSF-producing T H cells were detected in all samples, but observed low frequency ( ⁇ 1%) of IL-17-producing T H cells ( FIG. 24B ).
  • GM-CSF-single-producing (GM-CSF + IFN- ⁇ ⁇ ) T H cells represented a substantial population in both RA and HC ( FIG. 24B ).
  • GM-CSF-producing T H cells were isolated from synovial fluid of RA patients and analyzed the abundance of these cells.
  • a marked elevation of GM-CSF-producing T H cell frequency was observed in synovial fluid compared with peripheral blood, but most of these cells co-expressed IFN- ⁇ ( FIG. 24E ).
  • both T H 1 and T H 17 frequencies were also increased in synovial fluid, with T H 17 remaining to be a minor population compared with T H 1 ( FIG. 24E ).
  • GM-CSF and TNF- ⁇ level in plasma of RA in comparison to HC may suggest a therapeutic approach by targeting these two cytokines.
  • the efficacy of blocking both GM-CSF and TNF- ⁇ was tested in treating arthritic mice in antigen-induced arthritis (AIA) model, which is a T-cell driven RA model and is easily inducible in C57BL/6 strain with a rapid and synchronized disease onset, facilitating the exploration of RA pathogenesis.
  • AIA antigen-induced arthritis
  • Either GM-CSF or TNF- ⁇ individual blockade attenuated MA development ( FIG. 25A ).
  • the combination of GM-CSF- and TNF- ⁇ -specific neutralizing antibodies showed better efficacy in controlling arthritis development than individual treatments ( FIG.
  • GM-CSF may have beneficial efficacy in treating arthritis in a way independent of TNF- ⁇ activity.
  • a mouse strain Cd4-Cre; Stat5 f/f , or Stat5 ⁇ / ⁇ in short
  • conditional Stat5 deletion was used in T cells for AIA induction.
  • These conditional Stat5-knockout mice resisted arthritis development, as exemplified by milder joint selling, fewer immune cell infiltration in synovia, and reduced joint destruction ( FIGS. 25B-25D ), even though they had markedly increased level of serum TNF- ⁇ as well as IFN- ⁇ ( FIG. 25E ).
  • Splenocytes were collected from wild-type AIA mice and separated cells into three fractions: splenocytes, splenocytes depleted of CD4 + T cells and CD4 + T cells; and stimulated each fraction at same cell numbers under various conditions. Splenocytes produced low but detectable level of GM-CSF without stimulation, which was markedly increased by PMA/Ionomycin or mBSA antigen stimulation ( FIG. 28A ). Under all conditions, splenocytes depleted of CD4 + T cells almost completely abrogated GM-CSF production ( FIG.
  • CD4 + T cells produced dramatically elevated GM-CSF in comparison to splenocytes under all conditions ( FIG. 28A ).
  • CD4 + T cells are predominant producers of GM-CSF at least in spleens of arthritic mice, which is somehow consistent the observed correlation of plasma GM-CSF concentration with GM-CSF-single-producing T H cell frequency in RA ( FIG. 24D ).
  • the functional significance of T H -cell-secreted GM-CSF was examined in arthritis development.
  • T H 1 is a predominant population among synovial infiltrating CD4 + T cells in human RA (Berner et al., 2000; Yamada et al., 2008), defective IFN- ⁇ signaling results in increased disease susceptibility in animal models of arthritis (Guedez et al., 2001; Irmler et al., 2007; Manoury-Schwartz et al., 1997; Vermeire et al., 1997).
  • T H 17 cells are proven crucial in animal models of arthritis (Pernis, 2009), but predominance of T H 17 cells is limited in both peripheral blood and synovial compartment of human RA (Yamada et al., 2008) and ( FIGS. 1B and 1E ). As demonstrated herein, STAT5-regulated GM-CSF-producing T H cells potentiate arthritis pathogenesis.
  • splenocytes derived from AIA mice were stimulated with PMA/Ionomycin plus Golgiplug ex viva, followed by intracellular cytokine staining and flow cytometry.
  • the frequency of GM-CSF-single-producing cells among CD4 + CD44 hi population was significantly decreased in Stat5 ⁇ / ⁇ mice ( FIG. 28 34 B).
  • no significant differences were observed in frequencies of IL-17-single-producing (T H 17) or IFN- ⁇ -single-producing (T H 1) cells between two groups ( FIG. 28B ).
  • both T H 1 and T H 17 cells exhibited similar percentages between two groups ( FIG. 28C ).
  • inflamed synovial tissues were harvested from AIA mice for RNA and protein extraction to examine cytokine level by qPCR and ELISA.
  • lower synovial GM-CSF but not IFN- ⁇ or IL-17 was detected in Stat5 ⁇ / ⁇ mice than Stat5 +/+ mice at day 5 or 7 after arthritis induction ( FIGS. 28E and 30A-30C ).
  • FIGS. 28E and 30A-30C two important proinflammatory cytokines IL-6 and IL-1 ⁇ were also found persistently and significantly reduced in STAT5-deficient mice ( FIGS. 28E and 30A-30C ), indicating the attenuated synovial inflammation. Notably. TNF- ⁇ production was reduced at day 7 but not at day 5 in STAT5-deficient mice ( FIGS. 28E and 30A -C). Together, these results indicate that STAT5-regulated GM-CSF expression by arthritogenic T H cells is crucial for evoking synovial inflammation.
  • GM-CSF was administered via intra-articular injection in mixture with mBSA to the left knee joints of mBSA/CFA-immunized mice, whereas mBSA was injected alone to the right knee joints. Injection with mBSA alone was sufficient to induce abundant immune cell infiltration in the synovial compartments of Stat5 +/+ mice but failed to do so in Stat5 ⁇ / ⁇ mice ( FIG. 28F ). Administration of GM-CSF together with mBSA efficiently restored synovial inflammation in Stat5 ⁇ / ⁇ mice ( FIG. 34F ).
  • GM-CSF-producing Th cells evoke synovial inflammation and drive arthritis development was examined.
  • Myeloid lineage-derived cells including neutrophils, DCs and macrophages, express GM-CSF receptor and are common targets of GM-CSF (Hamilton, 2008).
  • those cells invade synovial compartments in RA patients and mouse arthritis models, and contribute to synovitis (McInnes and Schett, 2011).
  • the infiltration of myeloid lineage-derived cells in synovial compartments of ALA mice was examined.
  • CD11b + myeloid cells represented a predominant population ( ⁇ 70%) among synovial infiltrating leukocytes ( FIG. 31B ).
  • CD4 + T H cell infiltration was not altered by STAT5 deletion, synovial CD11b + cell infiltration was significantly reduced in Stat5 ⁇ / ⁇ mice compared with Stat5 +/+ mice when examined at day 7 after arthritis induction ( FIG. 31B ). This reduction is unlikely due to defective hematopoiesis, as similar frequencies of CD11b + cells were detected in spleens of two group ( FIG. 31A ). Further, CD11b + cells continuously increased in synovial tissues of wild-type mice, but not STAT5-deficient mice, over a 7-day time course ( FIG. 31C ).
  • MoDCs Monocyte-derived dendritic cells
  • mBSA/IL-1 ⁇ arthritis model Campbell et al., 2011
  • MoDCs were identified at low abundance in spleens and synovial tissues (data not shown).
  • comparable frequencies of MoDCs were detected in both peripheral lymphoid tissues and synovial tissues between Stat5 +/+ and Stat5 ⁇ / ⁇ mice (data not shown).
  • Neutrophils have great cytotoxic potential and contribute to the RA initiation and progression in multiple ways (Wright et al., 2014). It has been suggested that RA disease activity and joint destruction directly correlates with neutrophil influx to joints (Wright et al., 2014). Based on the differential expression of Ly6C and Ly6G, CD11b + myeloid cells can be classified into Ly6C lo Ly6G hi population (neutrophils) and Ly6C hi Ly6G ⁇ population (monocytes/macrophages).
  • Ly6C lo Ly6G hi population continued to accumulate in synovial tissues over a 7-clay time course, and represented a predominant population among synovial CD11b + cells in wild-type mice at day 7 after AIA induction, whereas this population was persistently and dramatically diminished in STAT5-deficient mice ( FIG. 32A ).
  • Giemsa stain it was validated that synovial-infiltrating Ly6C lo Ly6G hi population were neutrophils, which displayed typical polymorphonuclear characteristics with ring-shaped nuclei ( FIG. 32B ).
  • synovial-infiltrating Ly6C hi Ly6G ⁇ population had mononuclear morphology and were likely monocytes/macrophages ( FIG.
  • Neutrophils are recruited during inflammation, in which complex interactions between neutrophils and vascular endothelial cells direct neutrophil adhesion and transmigration from circulation to inflamed tissues (Kolaczkowska and Kubes, 2013).
  • neutrophil adhesion and migration across monolayers of endothelial cells was significantly enhanced by GM-CSF as chemoattractant ( FIGS. 33A and 33B ), suggesting GM-CSF may mediate neutrophil recruitment to inflamed joints in AIA.
  • Effective neutrophil apoptosis is crucial for the resolution of inflammation.
  • neutrophil apoptosis is delayed with a result of extended survival and persistent inflammation (Wright et al., 2014).
  • GM-CSF may mediate neutrophil recruitment and sustain neutrophil survival in synovial compartments and contribute to persistent synovitis.
  • ⁇ a neutralizing antibody (1A8) specific for Ly6G was used to deplete neutrophils in vivo.
  • the administration of neutralizing antibody resulted in significant improvement of joint swelling in AIA ( FIG. 32D ).
  • neutrophils accumulation mediated by T H -cell-derived GM-CSF are important for AIA development.
  • Cytokines are important mediators in the cross-talk between innate and adaptive immunity. As shown herein, several proinflammatory cytokines (IL-6, IL-1 ⁇ and TNF- ⁇ ), which are in association with RA pathogenesis (Choy and Panayi, 2001), were significantly reduced in synovial tissues of STAT5-deficient AIA mice ( FIGS. 28E and 30A-30C ). To gain insights into the mechanism underlying the observed cytokine reduction, the cellular sources of these proinflammatory cytokines were examined by fractionating synovial cells into different populations based the differential expression of surface markers ( FIG. 34A ).
  • Cytokine mRNA expression level in CD45 + TCR ⁇ + population (T cells), CD45 + TCR ⁇ ⁇ CD11c ⁇ CD11b + population (mostly monocytes/macrophages and neutrophils) and CD45 + TCR ⁇ ⁇ CD11c + population (dendritic cells) was assessed by RT-PCR.
  • GM-CSF as similar to IL-17 (as a control), was predominantly produced by synovial T cells ( FIG. 34B ), further reinforcing the importance of GM-CSF-producing T H cells.
  • IL-6 and IL-1 ⁇ were mainly produced by myeloid cells, e.g. CD11b + population and CD11c + population ( FIG. 34B ).
  • TNF- ⁇ was expressed by all three populations, with relatively lower abundance in T cells ( FIG. 34B ).
  • Ly6C lo Ly6G hi population (neutrophils) and Ly6C hi Ly6G ⁇ population (monocytes/macrophages) were further analyzed, which showed that monocytes/macrophages were likely the major IL-6 producers whereas neutrophils seemed to be better producers of IL-1 ⁇ and TNF- ⁇ ( FIG. 34C ).
  • BMDMs bone marrow-derived macrophages
  • BMDCs bone marrow-derived dendritic cells
  • BMDMs were primed with LPS for 6 h during which different concentrations of GM-CSF was added, followed by ATP stimulation.
  • the addition of GM-CSF significantly enhanced the secretion of IL-1 ⁇ into culture supernatant as measured by ELISA ( FIG. 34H ).
  • Synovial fibroblasts the active players in synovial inflammation (Muller-Ladner et al., 2007), also showed increased IL-1 ⁇ mRNA expression upon GM-CSF stimulation ( FIG. 34I ).
  • An inducible effect of GM-CSF on TNF- ⁇ expression was not observed in BMDMs, BMDCs or synovial fibroblasts (data not shown).
  • T H -cell-secreted GM-CSF may mediate synovial inflammation also via eliciting the expression of IL- and IL-1 ⁇ from myeloid cells and synovial fibroblasts.
  • elegans 10526941 RIKEN cDNA 10394816 predicted gene D830046C22 gene 9282 10567448 dynein, axonemal, 10587503 SH3 domain heavy chain 3 binding glutamic acid-rich protein like 2 10437885 myosin, heavy 10411359 proteolipid polypeptide 11, protein 2 smooth muscle 10600122 X-linked 10579939 ubiquitin specific lymphocyte- peptidase 38 /// regulated 3B /// X- predicted gene linked lymphocyte- 9725 regulated 3C /// X- linked lymphocyte- regulated 3A 10587665 RIKEN cDNA 10370242 poly(rC) binding 4930579C12 gene protein 3 10350753 glutamate- ammonia ligase (glutamine synthetase) 10456296 mucosa associated lymphoid tissue lymphoma translocation gene 1 10380571 guanine nucleotide binding protein (G protein), gamm
  • RNA exonuclease 2 homolog 10560791 predicted gene, EG381936 /// predicted gene 6176 10407159 ankyrin repeat domain 55 10603659 mediator complex subunit 14 10576854 cortexin 1 10353775 BEN domain containing 6 10573865 predicted gene 3579 10356886 solute carrier organic anion transporter family, member 4C1 10507273 phosphatidylinositol 3 kinase, regulatory subunit, polypeptide 3 (p55) 10424252 WDYHV motif containing 1 10518735 splA/ryanodine receptor domain and SOCS box containing 1 10562576 pleckstrin homology domain containing, family F (with FYVE domain) member 1 10375667 ring finger protein 130 10528268 protein tyrosine phosphatase, non- receptor type 12 10593205 REX2, RNA exonuclease 2 homolog ( S.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Hematology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Mycology (AREA)
  • Urology & Nephrology (AREA)
  • General Engineering & Computer Science (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Rheumatology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Rehabilitation Therapy (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)

Abstract

Disclosed herein is a T-helper cell (“TH-GM” cell) that is regulated by IL-7/STAT5 and which secrete GM-CSF/IL-3. Also disclosed are methods and compositions for modulating TH-GM function for the treatment of, e.g., inflammatory disorders. Diagnostic and prognostic methods for specifically identifying TH-GM-mediated inflammatory disorders (e.g., rheumatoid arthritis), as distinct from and/or in addition to non-TH-GM-mediated (e.g., TNF-α-mediated) inflammatory disorders, are also provided.

Description

    RELATED APPLICATION
  • This application claims the benefit of Singapore Patent Application No. 10201406130P, filed Sep. 26, 2014. The entire teachings of the above application are incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • A significant body of research has led to the current model of immunity and inflammation, as well as the dysregulation in immune and inflammatory disorders. It is currently understood that CD4+ helper T (TH) cells play a crucial role in host defense against various pathogens by orchestrating adaptive and innate immune responses. Upon T-cell receptor (TCR) activation by cognate antigen, naïve CD4+ T cells are committed to differentiate into at least live major subsets: T H1, T H2, T H17, iTreg and TFH, which are modulated by cytokine milieus. T H1 and T H17 cells are known to be the primary effectors of inflammation. However, the pathogenic roles of either T H1 or T H17 in various inflammatory disorders remain unclear. For example, recent studies conflict with previously understood paradigm of T H17 in multiple sclerosis (MS) pathogenicity (Haak et al., 2009), making it more challenging to identify potential drug targets for MS therapy. Similarly, while rheumatoid arthritis (RA) is traditionally understood to be a disorder mediated by tumor necrosis factor α (TNF-α), up to 40% of RA patients fail to respond to anti-TNF-α treatment.
  • Accordingly, there remains a significant unmet need for effective treatment methods for autoimmune and inflammatory disorders such as, e.g., MS and RA.
  • SUMMARY OF THE INVENTION
  • The present disclosure relates, in part, to the identification of an interleukin-7 (IL-7)/signal transducer and activator of transcription 5 (STAT5)-regulated granulocyte macrophage colony-stimulating factor (GM-CSF)/IL-3-producing TH cells, termed TH-GM, which represent a distinct helper T cell subset with unique developmental and functional characteristics. Identified herein is an inflammatory pathway mediated by TH-GM cells (TH-GM-mediated inflammatory pathway), which represents an independent inflammatory pathway apart from known non-TH-GM-mediated inflammatory pathways (e.g., TNF-a, IL-6, and IL-1b pathways of inflammation). The present disclosure provides methods and compositions for diagnosing inflammatory disorders that are TH-GM-mediated, and modulating TH-GM cell function for the treatment of inflammatory disorders mediated by the TH-GM pathway.
  • Accordingly, in one aspect, the present disclosure provides a method of diagnosing a TH-GM-mediated inflammatory disorder in a patient suffering from an inflammatory disorder, comprising: a) contacting a sample collected from a patient suffering from an inflammatory disorder with a detecting agent that detects a polypeptide or nucleic acid level of STAT5 (e.g., phospho-STAT5 (Tyr694)). IL-7, GM-CSF or IL-3, or a combination thereof; and h) quantifying the polypeptide or nucleic acid level of STAT5 (e.g., phospho-STAT5 (Tyr694)), IL-7, GM-CSF or IL-3, or a combination thereof, wherein an increased level of STAT5 (e.g., phospho-STAT5 (Tyr694)), interleukin-7 (IL-7), GM-CSF or interleukin-3 (IL-3), or a combination thereof relative to a reference level indicates that the patient suffers from a TH-GM-mediated inflammatory disorder.
  • In another aspect, the present disclosure provides an isolated population of GM-CSF-secreting T-helper cells (TH-GM), wherein the TH-GM cells are differentiated from cluster of differentiation 4 (CD4+) precursor cells in the presence of IL-7 and activated STAT5, and wherein the TH-GM cells express GM-CSF and IL-3.
  • In another aspect, the present disclosure provides a method of modulating TH-GM function, comprising contacting the TH-GM, or CD4+ precursor cells, or both, with a modulating agent that modulates TH-GM function.
  • In some aspects, the present disclosure provides a method of treating a TH-GM-mediated inflammatory disorder in a patient in need thereof, comprising administering to said patient an effective amount of a modulating agent that modulates TH-GM cell function.
  • In other aspects, the present disclosure provides a method of treating rheumatoid arthritis in a patient who exhibits limited response to anti-tumor necrosis factor alpha (TNF-α) therapy, comprising administering to said patient an effective amount of a modulating agent that modulates TH-GM function.
  • In another aspect, the present disclosure provides a method of treating a STAT5-mediated inflammatory disorder in a patient in need thereof, comprising administering to the patient an effective amount of an agent that modulates STAT5 function.
  • In further aspects, the present disclosure provides a method of screening to identify a modulator of TH-GM cell function, comprising contacting an isolated population of TH-GM cells, or an isolated population of CD4+ precursor cells, with a candidate agent, and determining a readout of TH-GM function in the presence or absence of the candidate agent, wherein a change in the readout of TH-GM function indicates that the candidate agent is a modulator of TH-GM function.
  • The present disclosure enables the identification or classification between inflammatory disorders that are either primarily TH-GM-mediated, or primarily non-TH-GM-mediated (e.g., mediated by TNF-α, IL-6, and/or IL-1β), or both. Thus, using the methods described herein, it is possible to determine whether a patient suffering from, e.g., RA, suffers from an RA that is primarily TH-GM-mediated, or primarily non-TH-GM-mediated, or both. This differentiation allows for a more targeted and tailored method of treating inflammatory disorders such as RA, for which current treatments are only 40% effective. Further, the present disclosure provides methods and compositions for prognosing the progression of an inflammatory disorder so as to tailor the treatment according to the stage of the disease. Also provided herein are compositions and methods for and the treatment of inflammatory disorders, particularly those that are TH-GM-mediated.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The foregoing will be apparent from the following more particular description of example embodiments of the invention.
  • FIGS. 1A-1D depict Stat5-conditional mutant mice are resistant to EAE. Clinical EAE scores (FIG. 1A) and incidence (FIG. 1B) of Stat5+/+ and Stat5−/− mice immunized twice with MOG35-55/CFA. Data are representative of three independent experiments (FIG. 1A) or pooled from three experiments (FIG. 1B, n=18 per group). Clinical scores of EAE mice immunized once with MOG35-55/CFA (FIG. 1C, n=5 per group) or immunized twice with MOG35-55/LPS (FIG. 1D). Data are representative of two independent experiments.
  • FIGS. 2A-2D depict reduced neuroinflammation in Stat5 conditional mutant mice. Histology of spinal cord sections obtained from EAE mice at day 9 after 2nd immunization (FIG. 1A). Images shown are representative of two independent experiments with three mice per group. Scale bars, 200 μm (top), 50 μm (bottom). CD4+ and CD11b+ cells in spinal cord sections were stained by immunofluorescence (FIG. 1B). Images shown are representative of two independent experiments with three mice per group. Scale bars, 200 μm. CNS mononuclear cells were analyzed by flow cytometry at peak of disease (FIGS. 2C and 2D). Right panels are cell proportions (FIG. 2C, right) or cell numbers (FIG. 2D, right) pooled from two experiments (n=9).
  • FIGS. 3A and 3B depict the resistance to EAE in Stat5-deficient mice is independent of T H1, T H17 or Treg cells. Flow cytometric analysis of IL-17 and IFN-γ expression by CNS-infiltrating CD4+ T cells at peak of disease (FIG. 3A). Data are representative of three independent experiments. Percentage of CD25+ among CD4+ T cells in the CNS of Stat5+/+ and Stat5−/− EAE mice at peak of disease were analyzed by flow cytometry (FIG. 3B).
  • FIGS. 4A-4C depict conditional Stat5 mutant mice have no defect in CD4+ T cell generation in periphery. Spleens were obtained from MOG35-55/CFA-immunized Stat5+/+ and Stat5−/− mice at day 7 (FIG. 4A) and day 21 (FIG. 4B). The proportions of CD4+ and CD8+ T cells were analyzed by flow cytometry. The absolute number of CD4+ T cells was calculated (right panels). Data are representative of two independent experiments (FIG. 4A) or pooled from two independent experiments (FIG. 4B). IL-17 and IFN-γ expression by splenic CD4+ T cells of Stat5+/+ and Stat5−/− EAE mice was determined by intracellular cytokine staining (FIG. 4C). Data are representative of three independent experiments. *p<0.5, **p<0.005, ***p<0.0005.
  • FIGS. 5A-5D depict Stat5-deficient CD4+ T cells can infiltrate CNS but fail to induce effective neuroinflammation. CCR6, CXCR3 and CD69 expression by splenic CD4+ T cells of Stat5+/+ and Stat5−/− EAE mice was measured. Data are representative of two independent experiments with three to five mice per group (FIG. 5A). CNS-infiltrating CD4+ T cells were analyzed at day 7, 9 and 21 after first MOG35-55/CFA immunization (FIGS. 5B-5D). Cell numbers were calculated (FIG. 5D). Data are representative of two independent experiments with three mice per group. *p<0.5.
  • FIGS. 6A-6C show resistance to EAE in Stat5−/− mice is not caused by any defect in the survival of CD4+ T cells in the absence of STAT5. CD4+ T cell infiltration (FIG. 6A) and clinical scores (FIG. 6B) of Rag2−/− recipient mice transferred with different numbers of Stat5+/+ and Stat5−/− CD4+ T cells. Clinical scores and frequencies of CD4+ T cells in the CNS at day 21 (disease peak) of EAE induction (FIG. 6C). *p<0.05. ***p<0.0005.
  • FIGS. 7A-7C depict the intrinsic defect of Stat5-deficient CD4+ T cells in encephalitogenicity. Clinical EAE scores (FIG. 7A) and incidence (FIG. 7B) of Rag2−/− mice (n=5 per group) after adoptive transfer of 2 million MOG35-55-specific Stat5+/+ or Stat5−/− CD4+ T cells respectively. IL-17 and IFN-γ expression by CNS-infiltrating CD4+ T cells was measured at peak of disease (FIG. 7C). Data represent two independent experiments, *p<0.05.
  • FIGS. 8A-8D depict the diminished induction of GM-CSF in splenic Stat5−/− CD4+ T cells. In FIGS. 8A-8D, splenocytes were obtained from MOG35-55/CFA-immunized Stat5+/+ and Stat5−/− mice (n=3 per group) before disease onset and challenged with MOG35-55 at various concentrations for 24 h. GM-CSF secretion was measured by ELISA (FIG. 8A). Golgiplug was added in the last 4 h of MOG35-55 (20 μg/ml) challenge and the frequencies of IL-17+ and GM-CSF+ cells among CD4+CD44hi T cells were measured (FIG. 8B). In FIGS. 8C and 8C, splenocytes were obtained from MOG35-55/CFA-immunized Stat5−/−, Stat3−/− or wild-type control mice and stimulated with PMA/Ionomycin in the presence of Golgiplug for 4 h. The frequencies of IL-17+ and GM-CSF+ cells among splenic CD4+CD44hi T cells were measured by intracellular cytokine staining. *p<0.05, ***p<0.001.
  • FIGS. 9A-9C depict the diminished induction of GM-CSF in CNS-infiltrating Stat5−/− CD4+ T cells. In FIG. 9A, IL-17, IFN-γ and GM-CSF expression by CNS-infiltrating CD4+ T cells of Stat5+/+ and Stat5−/− mice was measured at peak of disease. The percentage of GM-CSF+ cells among IL-17+ or IFN-γ+ cells was calculated (bottom right. FIG. 9A). IL-17. IFN-γ and GM-CSF expression by CNS-infiltrating CD4+ T cells of Rag2−/− recipient mice at peak of disease in adoptive transfer EAE (FIG. 9B). Time-course analysis of cytokine mRNA expression in the CNS of naïve and MOG35-55/CFA-immunized Stat5+/+ and Stat5−/− mice (n=3 per group at each time point). The RT-PCR data were normalized to Rn18S, and expression in naïve mice was set to 1 (FIG. 9C). Data represent two independent experiments. *p<0.05.
  • FIGS. 10A-10C show STAT5-mediated GM-CSF induction is independent of IL-23 or IL-1β signaling. In FIG. 10A, purified CD4+ T cells were cultured with TGF-β and IL-6 for 3 days, followed by starvation for 6 h. Then cells were treated with various cytokines for 30 min. and pSTAT3 and pSTAT5 was determined by immunoblotting. STAT3 and STAT5 were further detected after stripping. FIG. 10B shows the mRNA expression of IL-23R and IL-1R1 in splenic CD4+ T cells of Stat5+/+ and Stat5−/− EAE mice (n=3 per group). The RT-PCR data were normalized to β-Actin. In FIG. 10C, splenocytes were obtained from MOG35-55/CFA-immunized WT mice before disease onset and challenged with MOG35-55 (20 μg/ml) in the absence or presence of IL-2 for 48 h. The frequencies of GM-CSF+ and IL-17+ cells among CD4+CD44hi T cells were measured by flow cytometry. *p<0.05.
  • FIGS. 11A-11C depict IL-7-induced STAT5 activation promotes GM-CSF expression in autoreactive CD4+ T cells. Splenocytes were obtained from MOG35-55/CFA-immunized Stat5+/+ and Stat5−/− mice before disease onset and challenged with MOG35-55 (20 μg/ml) in the absence or presence of IL-7 for 48 h. Frequencies of GM-CSF+ and IL-17+ cells among CD4+CD44hi T cells were measured by flow cytometry (FIG. 11A). GM-CSF secretion was measured by ELISA (FIG. 11B). Data represent two independent experiments with two to three mice per group. Splenic CD62LhiCD44lo and CD62LloCD44hi T cells from MOG35-55/CFA-immunized mice were sorted out. Cells were stimulated with anti-CD3 and anti-CD28 in the absence or presence of IL-7 for 4 h and then harvested for the analysis of GM-CSF expression by RT-PCR (FIG. 11C). *p<0.05
  • FIGS. 12A-12F depict IL-7Rα neutralization attenuates GM-CSF expression and ameliorates EAE. Clinical scores of EAE mice (n=5) treated with anti-IL-7Rα or normal IgG given every other day from day 5 after 2nd immunization, as indicated by arrows. Data represent two independent experiments (FIG. 12A). Spinal cord sections were obtained from EAE mice at day 11 after 2nd immunization. Immune cell infiltration was assessed histologically. Images shown are representative of three individuals per group. Scale bars, 200 μm (top), 50 μm (FIG. 12B, bottom). The percentages of CD4+ and CD8+ T cells in spleens of EAE mice. Data represent two independent experiments (FIG. 12C). FIGS. 12D and 12E illustrate the frequencies of GM-CSF+, IL-17+ and IFN-γ+ cells among CD4+ T cells in the CNS of EAE mice receiving different treatment. The mRNA expression of IFN-γ, IL-17 and GM-CSF in the CNS of EAE mice (FIG. 12F). *p<0.05
  • FIGS. 13A and 13B depict the differentiation of GM-CSF-expressing TH cells is distinct from T H17 and T H1. Naïve CD4+ T cells were primed with plate-bound anti-CD3 and soluble anti-CD28 in the presence of a combination of various cytokines and neutralizing antibodies as indicated. GM-CSF, IL-17 and IFN-γ expression was analyzed by intracellular staining (FIG. 13A) or RT-PCR (FIG. 13B)
  • FIGS. 14A-14D show the effect of IL-2 and IL-6 on TH-GM differentiation from naïve T cells. GM-CSF and IFN-γ expression in naive CD4+ T cells activated for 72 h with anti-CD3 alone or plus anti-CD28 (FIG. 14A). In FIG. 14B, sorted naïve CD4+ T cells were stimulated with anti-CD3 and anti-CD28 in the presence of neutralizing antibodies against IL-12 and IFN-γ without or with the addition of IL-6. The frequencies of GM-CSF+ and IL-17+ cells were measured by intracellular staining (FIG. 14B). In FIG. 14C, naïve CD4+ T cells from Stat3+/+ and Stat3−/− mice were polarized under conditions as indicated for 72 h. The frequencies of GM-CSF+ and IL-17+ cells were analyzed. In FIG. 14D, naïve CD4+ T cells were activated with anti-CD3 and anti-CD28 in the presence of IL-2 or anti-IL-2. The frequencies of GM-CSF+, IL-17+ and IFN-γ+ cells were analyzed.
  • FIGS. 15A-15F depict IL-7-STAT5 signaling programs TH-GM differentiation from naïve precursor cells. Naïve CD4+ T cells were primed with plate-hound anti-CD3 and soluble anti-CD28 in the presence of various concentration of IL-7 as indicated. GM-CSF and IFN-γ expression was analyzed by intracellular staining (FIG. 15A) or ELISA (FIG. 15B). In FIGS. 15C and 15D, Stat5+/+ and Stat5−/− naïve CD4+ T cells were activated with anti-CD3 and anti-CD28 in the presence IL-7 for 3 days. GM-CSF, IL-17 and IFN-γ expression was analyzed by intracellular cytokine staining (FIG. 15C). GM-CSF secretion was measured by ELISA (FIG. 15D). Immunoblotting of pSTAT5 and STAT5 in IL-7-stimulated CD4+ T cells isolated from Stat5−/− or control mice (FIG. 15E). The ChIP assay was performed with Stat5+/+ and Stat5−/− CD4+ T cells using normal IgG or STAT5-specific antibody. The binding of antibodies to Csf2 promoter region was detected by RT-PCR (FIG. 15F).
  • FIGS. 16A and 16B depict the differentiation conditions for TH-GM subset. Naïve CD4+ T cells were activated with anti-CD3 and anti-CD28 in the presence of IL-7 or/and anti-IFN-γ as indicated. GM-CSF. IL-17 and IFN-γ expression was analyzed (FIG. 16A). The mRNA expression of T-bet and RORγt in naïve. TH1 (IL-12+anti-IL-γ), TH17 (TGF-β+IL-6+anti-IFN-γ+anti-IL-4) and TH-GM cells (IL-7 anti-IFN-γ) (FIG. 16B). The RT-PCR data were normalized to Gapdh, and expression in naïve T cells was set to 1.
  • FIGS. 17A-17E illustrate that IL-7 but not IL-2 induces STAT5 activation and GM-CSF expression in naïve CD4+ T cells. FIGS. 17A-17C show flow cytometry of CD25 and CD127 on the surface of naïve CD4+ T cells or cells activated with anti-CD3 and anti-CD28 at various time points as indicated. Activation of STAT5 in naïve CD4+ T cells stimulated with IL-2 or IL-7 for 30 min (FIG. 17D). FIG. 17E shows the mRNA expression of GM-CSF in naïve CD4+ T cells stimulated with anti-CD3 and anti-CD28 in the presence of IL-2 or IL-7. The RT-PCR data were normalized to β-Actin, and expression in naïve T cells activated for 2 h without cytokine was set to 1.
  • FIGS. 18A-18C show that both IL-2 and IL-7 can induce STAT5 activation and GM-CSF expression in activated CD4+ T cells. As shown in FIGS. 1.8A and 18B, CD4+ T cells were activated with anti-CD3 and anti-CD28 for 3 days. After resting in fresh medium, cells were stimulated with IL-2 or IL-7 at various time points. The pTyr-STAT5 and β-Actin were detected by immunoblotting (FIG. 18A). GM-CSF mRNA expression was measured by RT-PCR (FIG. 18B). The RT-PCR data were normalized to 13-Actin, and expression in cells without cytokine stimulation was set to 1. The ChIP assay shown in FIG. 18C was performed with normal IgG or STAT5-specific antibody. The binding of antibodies to Csf2 promoter region was detected by RT-PCR.
  • FIG. 19 depicts surface molecules selectively expressed at high level or low level in TH-GM subset as characterized by microarray analysis. These surface molecules specific for each lineage serves as markers, signatures and potential targets for novel diagnosis, treatment and prevention of autoimmune inflammation including, but not limited to multiple sclerosis and rheumatoid arthritis. These cell surface molecules are listed in detail in Table 1. The order of naïve, Th1, Th17, and Th-GM as indicated in the figure insert is the same as that appears for the bars in each graph.
  • FIGS. 20A-20D show that IL-3 is preferentially expressed in TH-GM cells. In FIGS. 20A and 20B, naïve CD4+ T cells were activated with anti-CD3 and anti-CD28 under T H1—(IL-12+anti-IL-4), T H17−(TGF-β+IL-6+anti-IFN-γ+anti-IL-4) and TH-GM—(GM-CSF+ TH, IL-7+anti-IFN-γ+anti-IL-4) polarizing conditions. GM-CSF and IL-3 expression was analyzed by intracellular staining (FIG. 20A). The mRNA expression of IL-3, EBI-3, PENK or RANKL cytokines was measured by RT-PCR (FIG. 20B). Frequency of IL-3+ cells differentiated without or with IL-7 (FIG. 20C). GM-CSF and IL-3 expression by WT or STAT5-deficient GM-CSF-producing TH cells (FIG. 20D).
  • FIG. 21 depicts clinical EAE scores of Rag2−/− mice (n=3˜6 mice per group) after adoptive transfer of 6×105 various MOG35-55-specific TH subsets.
  • FIGS. 22A-27E depict inhibition of STAT5 activation suppresses TH-GM cell differentiation in vitro. CD4+ T cells were pre-incubated with STAT5 inhibitor (Calbiochem) (FIG. 22A) or JAK3 inhibitor (FIG. 22B) at indicated concentrations or vehicle (−) for 1 h before stimulation with IL-7 for 30 min. Activation (Tyr694 phosphorylation) of STAT5 was determined by immunoblotting. CD4+ T cells were pre-incubated with STAT5 inhibitor at indicated concentrations or vehicle (−) for 1 h before stimulation with IL-6 for 30 min. Activation (Tyr705 phosphorylation) of STAT3 was determined by immunoblotting (FIG. 22C). In FIG. 221), CD4+ T cells were pre-incubated with STAT5 inhibitor at indicated concentrations or vehicle (−) for 1 h before stimulation with IFN-γ for 30 min. Activation (Tyr701 phosphorylation) of STAT1 was determined by immunoblotting. In FIG. 22E, naïve CD4+ T cells were isolated and activated under neutral condition or TH-GM cell-favoring condition with the addition of different concentrations of STAT5 inhibitor for 3 days. GM-CSF and IFN-γ expression was analyzed by intracellular cytokine staining and flow cytometry.
  • FIGS. 23A-23D depict targeting STAT5 activation by chemical inhibitor ameliorates EAE. (FIG. 23A) Clinical EAE scores of wild-type control mice (n=5) or administrated with STAT5 inhibitor (Calbiochem). Arrow indicates the treatment points. (FIG. 23B) Histology of spinal cords at clay 18 of EAE mice receiving different treatments. (FIG. 23C) Intracellular staining and flow cytometry of CNS-infiltrating CD4+ T cells at peak of disease. (FIG. 23D) Whole CNS was harvest for RNA extraction. GM-CSF mRNA expression was analyzed by RT-PCR. Data represent two independent experiments. *p<0.05.
  • FIGS. 24A-24E depict GM-CSF-producing TH cells are in association with human RA. Plasma concentrations of GM-CSF and TNF-α in healthy control HC (n=32) and RA (n=47) were quantified by ELISA (FIG. 24A). In FIGS. 24B and 24C, peripheral blood mononuclear cells (PBMCs) were collected from healthy control (HC) and rheumatoid arthritis (RA) patients, and were stimulated for 4 h with PMA/lonomycin in the presence of Golgiplug, followed by intracellular cytokine staining. Representative flow cytometry of GM-CSF, IFN-γ and IL-17 in CD4+ T cells (FIG. 24B) and statistics of n>=9 per group (FIG. 24C) are shown. FIG. 241D shows the correlation between the frequency of GM-CSF+IFN-γ TH cells and the level of plasma GM-CSF in peripheral blood of RA patients (n=18). Cytokine expression by CD4+ T cells derived from synovial fluid of RA patients was analyzed by intracellular cytokine staining and flow cytometry (FIG. 24E). A representative image of three cases was shown. *p<0.05, **p<0.01, ***p<0.001; ns, not significant.
  • FIGS. 25A-25E depict distinguishable effects of GM-CSF and TNF-α in mouse AIA. FIG. 25A shows knee joint swelling of wild-type mice over 7 days after intraarticular injection of 100 μg mBSA in AIA model, receiving treatment with control IgG, GM-CSF-specific and TNF-α-specific neutralizing antibodies separately or in combination (n=5 per group) at indicated times (arrows). FIG. 25B shows knee joint swelling of Stat5+/+ and Stat5−/− mice (n=6 per group) over 7 days after arthritis induction. Data are representative of more than three independent experiments. Representative images of joint sections stained with H&E (FIG. 25C) or Safranin-O/Fast Green (FIG. 25D) at day 7 after arthritis induction as in FIG. 25C. Bars, 500 μm (FIG. 25C upper panels and FIG. 25D) or 100 μm (FIG. 25C lower panels). Arrow in upper panels (FIG. 25C) indicated hone destruction. In FIG. 25E, serum concentrations of GM-CSF, IFN-γ and TNF-α in Stat5+/+ and Stat5−/− AIA mice were quantified by ELISA. Statistics of n>=8 per group were shown. *p<0.05, **p<0.01, ***p<0.001.
  • FIGS. 26A-26D depicts mice with Stat5 deletion in T cells are resistant to CIA. (FIG. 26A) Representative images of paw swelling of Stat5+/+ and Stat5−/− mice at day 40 after collagen II/CFA immunization in CIA model. (FIG. 26B) Clinical score of Stat5+/+ and Stat5−/− mice (n=5 per group) over 40 days after collagen II/CFA immunization. Data are representative of two independent experiments. (FIG. 26C) Representative images of paw sections stained with H&E at clay 40. (FIG. 26D) Serum concentrations of TNF-α (n=8 per group) were quantified by ELISA. *p<0.05, **p<0.01, ***p<0.001.
  • FIGS. 27A-27E depicts STAT5-deficient CD4+ T cells are defective in arthritogenic potential. (FIGS. 27A and 27B) Representative flow cytometry of CD4+ T cells in spleens (FIG. 27A) and inguinal lymph nodes (FIG. 27B) of Stat5+/+ and Stat5−/− mice at day 7 after AIA induction. (FIGS. 27C and 27D) Synovial tissues were harvested from Stat5+/+ and Stat5−/− mice at day 7 after AIA induction, and dissociated into single cells. Cell numbers of CD45+ leukocytes were calculated (FIG. 27C). The percentages of CD4+ T cells among CD45+ fraction were analyzed by flow cytometry, and cell numbers were calculated (FIG. 27D). (FIG. 27E) Histological analysis of joint sections from wild-type naïve mice at day 7 after being transferred with in vitro-expanded antigen-reactive CD4+ T cells and followed with intraarticular injection of mBSA. Bars, 100 μm. Data represent two independent experiments. *p<0.05; ns, not significant.
  • FIGS. 28A-28G depicts STAT5-regulated GM-CSF-producing TH cells are crucial for AIA. Spleens and synovial tissues were collected from Stat5+/+ and Stat5−/− mice at day 7 after arthritis induction. (FIG. 28A) Splenic fractions of wild-type AIA mice (n=3) were stimulated under indicated conditions for 18 h. GM-CSF levels in supernatant were quantified by ELISA. (FIGS. 28B-28D) Intracellular staining and flow cytometry of GM-CSF, IL-17 and IFN-γ in splenic CD4+CD44hi effector T cells (FIG. 28B) or in synovial infiltrating CD4+ T cells (FIGS. 28C and 28D) after restimulation for 4 h with PMA/Ionomycin in the presence of Golgiplug. Representative images and statistics of n=5 (FIG. 28B, right panels) or n=3 (FIG. 28D, right panels) per group were shown. Data represent two independent experiments. (FIG. 28E) Protein expression of several proinflammatory cytokines in synovial tissues was measured by ELISA. (FIGS. 28F and 28G) Representative images of joint sections stained with H&E (FIG. 28F) or Safranin-O/Fast Green (FIG. 28G) at day 7 after intraarticular injection of mBSA alone to the right knee joints and mBSA supplemented with GM-CSF to the left knee joints. Bars, 500, 50 or 200 μm as indicated. Data represent two independent experiments.*p<0.05, **p<0.01. ***p<0.001; ns, not significant. “Splenocytes” represent the left-most bars in each group, “splenocytes depleted of CD4+ T cells” represent the middle bars in each group, and “CD4+ T cells” represent the right-most bars in each group.
  • FIGS. 29A-29C depicts loss of STAT5 results in impaired GM-CSF production by antigen-specific CD4+ T cells. Spleens and inguinal LNs were collected from Stat5+/+ and Stat5−/− mice at day 7 after arthritis induction, and dissociated into single cell suspensions. Then, cells were stimulated with mBSA (20 μg/ml) for 24 h. (FIG. 29A) Golgiplug was added in the last 4 h of culture, followed by intracellular staining and flow cytometry. Representative plots of GM-CSF, IL-17 and IFN-γ expression in CD4+CD44hi effector T cells was shown, representing two independent experiments. (FIGS. 29B and 29C) Secreted cytokines in the supernatant (n=3 per group) were quantified by ELISA. Data represent two independent experiments. *p<0.05; ns, not significant.
  • FIGS. 30A-30C depicts loss of STAT5 impairs IL-6 and IL-1β expression in synovial tissues of arthritic mice. (FIGS. 30A-30C) The mRNA (FIGS. 30A and 30 C) and protein (FIG. 30B) expression of several proinflammatory cytokines in synovial tissues of Stat5+/+ and Stat5−/− mice (n>=3 per group) at day 5 or 7 after arthritis induction was measured by qPCR and ELISA. The qPCR data were normalized to Rn18S.
  • FIGS. 31A-31D depicts SAT5-induced GM-CSF expression mediates CD11b+ cell accumulation in inflamed synovial tissues. (FIG. 31A) The frequencies of CD11b+ cells in spleens of Stat5+/+ and Stat5−/− AIA mice were analyzed by flow cytometry. Statistics of n=3 per group (right panel) were shown. (FIG. 31B) Synovial tissues were harvested from Stat5+/+ and Stat5−/− mice at day 7 after arthritis induction, and dissociated into single cell suspensions. The percentage of CD11b+ myeloid cells among CD45+ fraction was analyzed by flow cytometry. Statistics of n=5 per group were shown in right panel. (FIG. 31C) Representative flow cytometry of CD11b+ and CD4+ cells gated on synovial CD45+ fraction over 7 days after arthritis induction. (FIG. 31D) Flow cytometric analysis of CD4+, CD11b+ and B220+ cell infiltration in synovial tissues of Stat5+/+ and Stat5−/− mice at day 7 after intraarticular injection of mBSA alone to the right knee joints and mBSA supplemented with GM-CSF to the left knee joints. Representative images were shown. All data shown are representative of two independent experiments. **p<0.01; ns, not significant.
  • FIGS. 32A-32D depicts GM-CSF mediates neutrophil accumulation in arthritic mice. (FIG. 32A) Flow cytometric analysis of Ly6C and Ly6G expression gated on synovial CD45+CD11b+ fraction over 7 days after arthritis induction. (FIG. 32B) Giemsa stain of sorted Ly6ChiLy6G and Ly6CloLy6Ghi cells from synovial tissues of AIA mice. Scale bar, 100 μm (left) or 20 μm (right). (FIG. 32C) Flow cytometric analysis of Ly6ChiLy6G and Ly6CloLy6Ghi populations in synovial tissues of Stat5+/+ and Stat5−/− mice at day 7 after intraarticular injection of mBSA alone to the right knee joints and mBSA supplemented with GM-CSF to the left knee joints. (FIG. 32D) Knee joint swelling of wild-type mice treated with Ly6G-specific neutralizing antibody (1A8) or IgG control (n=5 per group) over 3 days after intraarticular injection of mBSA in AIA model. Arrows indicate time points of antibody administration. *p<0.05.
  • FIGS. 33A-33C depicts GM-CSF enhances neutrophil transmigration and delay apoptosis in vitro. (FIG. 33A) Percentages of migrated neutrophils with or without GM-CSF as chemoattractant in transmigration assay at 3 h post start. (FIG. 33B) Microscopic images of CFSE-labeled neutrophils in the bottom of the lower chamber in transmigration assay. (FIG. 33C) Sorted neutrophils were cultured in vitro with or without GM-CSF (20 ng/ml) for 24 h. Neutrophils undergoing apoptosis were examined by Annexin V and propidium iodide (PI) co-staining. A representative flow cytometry of three repeats was shown. *p<0.05.
  • FIGS. 34A-34I depicts GM-CSF mediates proinflammatory cytokine expression by myeloid cells and synovial fibroblasts in arthritic mice. Synovial tissues were dissected from wild-type AIA mice and dissociated into single cell suspensions. (FIG. 34A) Flow cytometry plots depicting the fractionation into different populations based on differential expression of surface markers. (FIG. 34B) The mRNA expression of several proinflammatory cytokines in sorted CD45+TCRβ+ (TCRβ+ in short). CD45+TCRβCD11cCD11b+ (CD11b+) and CD45+TCRβCD11c+ (CD11c+) populations was measured by qPCR. The qPCR data were normalized to GAPDH. (FIG. 34C) The mRNA expression of IL-6, IL-1β and TNF-α in sorted Ly6ChiLy6G and Ly6CloLy6Ghi populations (gated on CD11b+ cells). The qPCR data were normalized to GAPDH. (FIGS. 34D and 34E) The mRNA expression of IL-6 and IL-1β by BMDMs (FIG. 34D) and BMDCs (FIG. 34E) upon stimulation with 20 ng/ml GM-CSF for 1 h. The qPCR data were normalized to GAPDH. (FIGS. 34F and 34G) BMDMs (FIG. 34F) and BMDCs (FIG. 34G) were stimulated with GM-CSF at indicated concentrations (n=3 per group) for 18 h. The secretion of IL-6 in the culture supernatant was quantified by ELISA. (FIG. 34H) BMDMs were primed with LPS (100 μg/ml) in the presence of GM-CSF at indicated concentrations (n=3 per group) for 6 h, followed by stimulation with ATP (5 mM) for 30 min. The secretion of IL-1β in the culture supernatant was quantified by ELISA. (FIG. 34I) Cells were cultured in DMEM medium supplemented with 10% PBS for over 20 days with more than 5 passages to obtain synovial fibroblasts. Synovial fibroblasts were stimulated with GM-CSF (20 ng/ml) for 1 h and harvested for RNA extraction. The mRNA expression of IL-1β was measured by qPCR. The qPCR data were normalized to GAPDH. All data shown represent two independent experiments.*p<0.05, **p<0.01.
  • DETAILED DESCRIPTION OF THE INVENTION
  • A description of example embodiments of the invention follows.
  • The present disclosure relates, in part, to the identification of a granulocyte macrophage colony stimulating factor (GM-CSF)-secreting T helper cell, termed “TH-GM”. As detailed herein. IL-7/STAT5 signaling programs the differentiation of precursor CD4+ cells to TH-GM, a process which is further modulated by IL-2 and IL-23 signaling. TH-GM cells are characterized by, e.g., GM-CSF and IL-3 production. TH-GM cells are distinct from the known helper T cells T H1 and T H17, with respect to, e.g., differentiation conditions, transcriptional regulation and effector cytokine expression. For example, IL-12/IFN-γ and TGF-0/IL-6, which mediate (e.g., promote the development of) T H1 and T H17, respectively, potently suppress the development of TH-GM from naïve CD4+ precursor cells, establishing that TH-GM cells develop via a lineage distinct from T H1 and T H17. Thus, the present disclosure provides a distinct network of factors, unique from factors known to mediate T H1 or T H17, that mediate TH-GM function (e.g., its differentiation and pathogenicity).
  • As shown herein, TH-GM cells preferentially induce EAE as compared with T H1 and T H17 cells, indicating that TH-GM cells represent the primary effectors in the pathogenesis of autoimmune neuroinflammation in humans. Moreover, blockade of IL-7 signaling and/or inhibition of STAT5 function (e.g., abrogation of expression or inhibition of STAT5 activity) attenuates autoimmune neuroinflammation associated with diminished GM-CSF production by TH-GM cells. Further, blockade of TH-GM cell-secreted GM-CSF ameliorates experimental arthritis in a TNT-α-independent manner, indicating an approach for the treatment of, e.g., rheumatoid arthritis patients who are unresponsive to TNF-α antagonistic drugs. Thus, the present disclosure enables one to distinguish between an inflammatory disorder (e.g., RA) that is mediated by the TH-GM pathway (e.g., a disorder that results from TH-GM pathogenicity through the action of, e.g., GM-CSF and/or IL-3, or any factor associated with the TH-GM pathway), or an inflammatory disorder that is mediated by, e.g., TNF-α, IL-6, and/or IL-1β pathways (i.e., non-TH-GM-mediated pathway). For example, a patient who has, e.g., RA may be afflicted with a type of RA that is primarily TH-GM-mediated, or primarily non-TH-GM-mediated (e.g., TNF-α-mediated or IL-6 mediated). The present disclosure enables the classification between TH-GM-mediated and non-TH-GM-mediated inflammation, allowing for a more precise diagnosis, prognosis, and treatment in an individual who is afflicted with an inflammatory disorder such as RA or MS.
  • As demonstrated herein, the present disclosure identifies a helper T cell subset (TH-GM), provides the molecular basis for the commitment and development of this subset from naïve precursor cells in vitro and in vivo, and demonstrates TH-GM cells as the primary pathogenic cells in autoimmune diseases and inflammatory disorders, for example, MS and RA. Thus, provided herein are compositions and methods for diagnosing inflammatory conditions primarily mediated by TH-GM cells, thereby enabling the identification of, e.g., RA patients who are non-responsive to TNF-α therapy (e.g., TNF-α inhibitor based therapy), as well as compositions and methods for modulating TH-GM function to treat autoimmune and inflammatory disorders. The methods of modulating TH-GM function include, e.g., administering agents to modulate the function (e.g., signaling, expression or activity) of the network of factors (e.g., IL-2/IL-7/STAT5/GM-CSF/IL-3) that mediate TH-GM function in an effective amount to modulate the function (e.g., development and pathogenicity) of TH-GM cells. In particular, the disclosure provides methods and composition for differentiating and diagnosing an inflammatory disorder, e.g., multiple sclerosis (MS), rheumatoid arthritis (RA) as primarily mediated by either TH-GM cells (i.e. TH-GM pathway mediated) or by non-TH-GM mechanism (e.g., TNF-α, IL-6, and/or IL-1β pathways), or both. Also provided herein are compositions and methods for and the treatment of inflammatory disorders, particularly those that are TH-GM-mediated.
  • Accordingly, in one aspect, the present disclosure provides a method of diagnosing a TH-GM-mediated inflammatory disorder in a patient suffering from an inflammatory disorder. In some embodiments, the method comprises contacting a sample collected from a patient suffering from an inflammatory disorder with a detecting agent that detects a polypeptide or nucleic acid level of a TH-GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof; and quantifying the polypeptide or nucleic acid level of the TH-GM-mediating factor (e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof), wherein an increased level of a TH-GM-mediating factor (e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof) relative to a reference level indicates that the patient suffers from a TH-GM-mediated inflammatory disorder, thereby diagnosing a TH-GM-mediated inflammatory disorder in a patient suffering from an inflammatory disorder.
  • As used herein, a “TH-GM-mediated” inflammatory disorder refers to a subtype of an inflammatory disorder (e.g., a subtype of RA or MS) that results from the physiological action of any one or more of the network of factors in the pathway that modulate TH-GM function (a “TH-GM-mediating factor”), as described herein. Such factors include, e.g., GM-CSF, activated STAT5, IL-7, IL-2, and IL-3. In a particular embodiment, STAT5 is activated STAT5, wherein tyrosine at position 694 is phosphorylated.
  • In some embodiments, the level of a TH-GM-mediating factor (e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof) that is not increased relative to a reference level indicates that the patient suffers from a non-TH-GM-mediated inflammatory disorder.
  • In certain embodiments, the method further comprises administering to the patient a TNF-α therapy, as described herein, if the level of a TH-GM-mediating factor (e.g. STAT5, IL-7, GM-CSF or IL-3, or a combination thereof) is not increased relative to a reference level.
  • As used herein, a “non-TH-GM-mediated” inflammatory disorder refers to an inflammatory disorder (e.g., RA or MS) that is primarily caused by, e.g., TNF-α, IL-6, or IL-1β (and/or factors in the TNF-α, IL-6, or IL-1β pathway). As such, a “TH-GM-mediated” inflammatory disorder results primarily (or exclusively) from a pathway that is distinct from one or more of the pathways that leads to a “non-TH-GM-mediated” inflammatory disorder (e.g., the pathways associated with TNF-α, IL-6, or IL-1β).
  • However, as those of skill in the art would appreciate, a TH-GM-mediated inflammatory disorder does not necessarily exclude the possibility that the inflammatory disorder could also be partially non-TH-GM-mediated (e.g., mediated by TNF-α, IL-6, or IL- and/or factors in the TNF-α, IL-6, or IL-1β pathway). Thus, a classification or diagnosis as “TH-GM-mediated” is synonymous with “primarily/predominantly TH-GM-mediated”, and a classification as “non-TH-GM-mediated” is synonymous with “primarily/predominantly non-TH-GM-mediated.” For example, without wishing to be bound by any particular theory, an inflammatory disorder in its early stage may be TH-GM-mediated. As the inflammatory condition advances to a late stage characterized by, e.g., tissue damage, the inflammatory disorder becomes progressively non-TH-GM-mediated. In some embodiments, a TH-GM-mediated inflammatory disorder is a condition that is responsive to modulation of TH-GM function, as determined by clinical standards; a non-TH-GM-mediated inflammatory disorder is a condition that is responsive to, e.g., TNF-α, IL-6, or IL-1β therapy, as determined by clinical standards. In certain embodiments, an inflammatory disorder can be responsive to modulation of TH-GM function as well as TNF-α, IL-6, and/or IL-1β therapy.
  • In some embodiments, the sample can be e.g., peripheral blood, cerebrospinal fluid, synovial fluid, or synovial membrane, or a combination thereof.
  • In some embodiments, the inflammatory disorder is an autoimmune disorder. In certain embodiments, the inflammatory disorder can be any inflammatory disorder mediated by TH-GM cells, and includes, but is not limited to rheumatoid arthritis, multiple sclerosis, ankylosing spondylitis, Crohn's disease, diabetes. Hashimoto's thyroiditis, hyperthyroidism, hypothyroidism, Irritable Bowel Syndrome (IBS), lupus erythematosus, polymyalgia rheumatic, psoriasis, psoriatic arthritis, Raynaud's syndrome/phenomenon, reactive arthritis (Reiter syndrome), sarcoidosis, scleroderma, Sjögren's syndrome, ulcerative colitis, uveitis, or vasculitis.
  • As used herein, a “detecting agent” refers to, e.g., an antibody, a peptide, a small molecule, or a nucleic acid that binds to a polypeptide or nucleic acid to be detected (e.g., STAT5 (e.g., phospho-STAT5 (Tyr694)), IL-7, GM-CSF or IL-3), and enables the quantification of the polypeptide or nucleic acid to be detected. The detecting agent can be detectably labeled, or quantifiable by other means known in the art.
  • In some embodiments, the detecting agent is an antibody that binds to the polypeptide of STAT5, IL-7, GM-CSF or IL-3. In one embodiment, the antibody is one that binds to an activated STAT5 (e.g., phosphorylated STAT5), as described herein. Antibodies to STAT5 (e.g., phospho-STAT5 (Tyr694)), IL-7, GM-CSF or IL-3 suitable for use in the present method are known and commercially available in the art (e.g., STAT5 Ab: C-17 from Santa Cruz Biotech; Phospho-STAT5 (Tyr694) Ab: #9351 or #9359 from Cell Signaling; IL-7 Ab: clone BVD10-40F6 from BD Pharmingen; IL-7R Ab: clone SB/14 from BD Pharmingen; GM-CSF Ab: clone MP1-22E9 from BD Pharmingen; IL-3 Ab: clone MP2-8F8 from BD Pharmingen.
  • In other embodiments, the detecting agent is a nucleic acid that hinds to the nucleic acid of STAT5. IL-7, GM-CSF and/or IL-3. Nucleic acid molecules encoding a, e.g., STAT5. IL-7, GM-CSF and/or IL-3 sequence, or fragments or oligonucleotides thereof, that hybridize to a nucleic acid molecule encoding a e.g., STAT5, IL-7, GM-CSF and/or IL-3 polypeptide sequence at high stringency may be used as a probe to monitor expression of nucleic acid levels of STAT5, IL-7, GM-CSF and/or IL-3 in a sample for use in the diagnostic methods of the disclosure. Methods of quantifying nucleic acid levels are routine and available in the art.
  • In some embodiments, the method further comprises contacting the sample with a detecting agent that detects a polypeptide or nucleic acid level of one or more genes (as well as the gene product) listed in Table 1. As described herein, Table 1 lists genes that are differentially expressed in TH-GM cells as well as genes that are differentially expressed on the surface of TH-GM cells, as compared to T H1 or T H17 cells.
  • In a particular embodiment, the method further comprises contacting the sample with a detecting agent that detects the polypeptide or nucleic acid level of basic helix-loop-helix family member e40 (BHLHe40), chemokine (C-C Motif) Receptor 4 (CCR4), and/or CCR6.
  • Standard methods may be used to quantify polypeptide levels in any sample. Such methods include, e.g., ELISA, Western blotting, immunohistochemistry, fluorescence activated cells sorting (FACS) using antibodies directed to a polypeptide, and quantitative enzyme immunoassay techniques known in the art. Such methods are routine and available in the art. Similarly, methods for quantifying nucleic acid levels (e.g., mRNA) are known in the art.
  • In the diagnostic method of the present disclosure, an increased level of STAT5 (e.g., activated phospho-STAT5 (Tyr694)), IL-7, GM-CSF and/or IL-3 relative to a reference level indicates that the patient suffers from a TH-GM-mediated inflammatory disorder.
  • In some embodiments, a STAT5 (e.g., activated phospho-STAT5 (Tyr694)), IL-7, GM-CSF and/or IL-3 level that is increased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 110%, at least 120%, at least 130%, at least 140%, at least 150%, at least 160%, at least 170%, at least 180%, at least 190%, at least 200%, at least 220%, at least 240%, at least 260%, at least 280%, at least 300%, at least 350%, at least 400%, at least 450%, at least 500%, at least 550%, or at least 600% relative to a reference level indicates that the patient suffers from a TH-GM-mediated inflammatory disorder. In a particular embodiment, an increase of at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, or at least 150% relative to a reference level indicates that the patient suffers from a TH-GM-mediated inflammatory disorder.
  • In some embodiments, a STAT5 (e.g., activated phospho-STAT5 (Tyr694)), IL-7, GM-CSF and/or IL-3 level that is not increased by at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, or at least 150% relative to a reference level indicates that the patient suffers from a non-TH-GM-mediated inflammatory disorder.
  • In certain embodiments, a STAT5 (e.g., activated phospho-STAT5 (Tyr694)), IL-7, GM-CSF and/or IL-3 level that is comparable (or unchanged) relative to a reference level indicates that the patient suffers from a non-TH-GM-mediated disorder. As used herein, a level that is “comparable” to that of a reference level refers to a level that is unchanged, or a change relative to the reference level that is statistically insignificant according to clinical standards. In certain embodiments, a comparable level (or unchanged level) can include a level that is not increased by at least 40%, at least 50%, at least 60%, or at least 70% relative to a reference level as, for example, it may not indicate a clinically significant change. In some embodiments, a level of a TH-GM-mediating factor (e.g., STAT5 (e.g., activated phospho-STAT5 (Tyr694)). IL-7, GM-CSF, and/or IL-3) that is decreased relative to a reference level can also indicate that the patient suffers from a non-TH-GM-mediated disorder.
  • In some embodiments, the reference level is a level that is used for comparison purposes, and may be obtained from, for example, a prior sample taken from the same patient; a normal healthy subject; a sample from a subject not having an autoimmune disease or an inflammatory disorder; a subject that is diagnosed with a propensity to develop an autoimmune disease but does not yet show symptoms of the disease; a patient that has been treated for an autoimmune disease; or a sample of a purified reference polypeptide or nucleic acid molecule of the disclosure (e.g., STAT5) at a known normal concentration. By “reference standard or level” is meant a value or number derived from a reference sample, or a value or range accepted in the art as indicative of being healthy (e.g., an individual that does not have an inflammatory disorder). A normal reference standard or level can also be a value or number derived from a normal subject who does not have an autoimmune disease. In one embodiment, the reference sample, standard, or level is matched to the sample subject by at least one of the following criteria; age, weight, body mass index (BMI), disease stage, and overall health. A standard curve of levels of purified DNA. RNA or mRNA within the normal reference range can also be used as a reference. A standard curve of levels of purified protein within the normal reference range can also be used as a reference.
  • In some embodiments, the patient afflicted with an inflammatory disorder who has been diagnosed or classified as having a TH-GM-mediated inflammatory disorder does not have a non-TH-GM-mediated inflammatory disorder (i.e., does not have a TNF-α, IL-6, or IL-1β-mediated inflammatory disorder). That is, the patient diagnosed as suffering from a TH-GM-mediated inflammatory disorder responds to modulation of TH-GM function (e.g., inhibition of STAT5, IL-7, GM-CSF and/or IL-3), but does not respond (or exhibits a limited response) to TNF-α therapy, as determined by clinical standards. However, as described herein, a TH-GM-mediated inflammatory disorder does not exclude the possibility that the inflammatory disorder is also partially (though not primarily) contributed by a non-TH-GM-mediated pathway (e.g., TNF-α, IL-6, IL-1β).
  • In some embodiments, the methods of the present disclosure further comprise administering an effective amount of a modulating agent that modulates TH-GM cell function to the patient diagnosed or classified as having a TH-GM-mediated inflammatory disorder. As described herein, in some embodiments, the modulating agent inhibits TH-GM function.
  • In some embodiments, the methods of the present disclosure further comprise administering an effective amount of, e.g., a TNF-α therapy, an IL-6 therapy, or an IL-1β therapy to a patient diagnosed or classified as having a non-TH-GM-mediated inflammatory disorder, as described herein.
  • In some aspects, the present disclosure also provides a method of classifying a patient suffering from an inflammatory disorder as having a TH-GM-mediated inflammatory disorder or a non-TH-GM-mediated inflammatory disorder. In some embodiments, the method comprises contacting a sample collected from a patient suffering from an inflammatory disorder with a detecting agent that detects a polypeptide or nucleic acid level of a TH-GM-mediating factor, such as, e.g., STAT5 (e.g., phosphorylated STAT5, Tyr694), IL-7, GM-CSF or IL-3, or a combination thereof. In certain aspects, the method further comprises quantifying the polypeptide or nucleic acid level of the TH-GM-mediating factor, such as, e.g., STAT5. IL-7, GM-CSF or IL-3, or a combination thereof, wherein an increased level of the TH-GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof relative to a reference level indicates that the patient suffers from a TH-GM-mediated inflammatory disorder; or a comparable level of the TH-GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof relative to a reference level indicates that the patient suffers from a non-TH-GM-mediated inflammatory disorder, thereby classifying the patient suffering from an inflammatory disorder as a TH-GM-mediated inflammatory disorder or a non-TH-GM-mediated inflammatory disorder.
  • In other aspects of the present disclosure, the methods disclosed herein can further comprise measuring the polypeptide or nucleic acid level of a factor that mediates a non-TH-GM-mediated inflammatory disorder. Such factors include, e.g., TNF-α, IL-6, and IL-1β.
  • For example, in some aspects, the present disclosure provides a method of determining a treatment regimen in a patient suffering from an inflammatory disorder. To illustrate, the method comprises quantifying a polypeptide or nucleic acid level of, e.g., activated STAT5 or GM-CSF in a sample collected from a patient suffering from an inflammatory disorder, and quantifying the polypeptide or nucleic acid level of, e.g., TNF-α in a sample collected from the patient. At least four scenarios can be considered.
  • In the first scenario, if the activated STAT5 or GM-CSF level is increased (e.g., by at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, or at least 150%) relative to a first reference level and the TNF-α level is comparable to a second reference level, then the patient is classified as having a TH-GM-mediated inflammatory disorder and the patient can be treated with an agent that modulates TH-GM function, as described herein.
  • In a second scenario, if the activated STAT5 or GM-CSF level is comparable to the first reference level and the TNF-α level is increased (e.g., by at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, or at least 150%) relative to the second reference level, then the patient is classified as having a non-TH-GM-mediated inflammatory disorder and the patient can be treated with, e.g., a TNF-α therapy.
  • In a third scenario, if the activated STAT5 or GM-CSF level is increased relative to the first reference level and the TNF-α level is also increased relative to the second reference level, and the increase is equivalent within clinical and/or statistical standards (e.g., both GM-CSF and TNF-α are at least 50% increased relative to the respective reference levels), then the patient is classified as having an inflammatory disorder that is equally TH-GM-mediated and non-TH-GM mediated (e.g., TNF-α-mediated). In such a case, the patient can be treated with an effective amount of an agent that modulates TH-GM function and an effective amount of, e.g., a TNF-α therapy. As demonstrated herein, the combination of both agents can have a synergistic effect.
  • In a fourth scenario, if the activated STAT5 or GM-CSF level is increased relative to the first reference level and the TNF-α level is also increased relative to the second reference level, but one is increased more than the other, then the inflammatory disorder is primarily mediated by the pathway that shows a greater increase. For example, if GM-CSF is increased by 40% relative to a reference level, and TNF-α is increased by 90% relative to a reference level, then the inflammatory disorder is primarily non-TH-GM-mediated. However, in this scenario, the patient may receive a combined treatment with an agent that modulates TH-GM function as well a TNF-α therapy (e.g., anti-TNF-α therapy), since GM-CSF is increased by, e.g., at least 40% relative to a reference level.
  • In some embodiments, the first and second reference levels are obtained from the same reference sample.
  • In a related aspect, the disclosure also provides a method of tailoring the treatment of a patient suffering from an inflammatory disorder according to the progression of a patient's inflammatory disorder. In the above illustrative example, the first scenario (increased TH-GM-mediating factor, e.g. STAT5 or GM-CSF but TNF-α level is comparable to a reference level) may indicate that the patient is in an early stage of an inflammatory disorder. Without wishing to be bound by any particular theory, during, for example, the early stages of an inflammatory disorder, naïve T cells are stimulated by antigen and programmed by IL-7/STAT5 to differentiate into GM-CSF/IL-3 producing TH-GM cells. During, for example, the late stages of an inflammatory disorder, TH-GM cytokines (e.g., IL-3 and GM-CSF) progressively stimulate more inflammatory cells such as macrophages and neutrophils resulting in the production of, e.g., TNF-α, IL-6, IL-1β, resulting in full-scale inflammation. Thus, in the above illustrative example, the second scenario (activated STAT5 Or GM-CSF level is comparable to the first reference level and the TNF-α level is increased) may indicate that the patient is in a late stage of an inflammatory disorder characterized by, e.g., tissue damage. Accordingly, the present disclosure enables the prognosis of a patient depending on the quantifiable level of one or more TH-GM-mediating factor (e.g., STAT5 (e.g., activated phospho-STAT5 (Tyr694)), IL-7, GM-CSF, and/or IL-3) and one or more non-TH-GM-mediating factor (e.g., TNF-α, IL-6, IL-1β), thereby tailoring the treatment according to the progression of the disease. Accordingly, as would be appreciated by those of skill in the art, a patient suffering from an inflammatory disorder can be monitored for disease progression to ensure effective and tailored treatment according to the level of one or more TH-GM-mediating factor, as described herein, and one or more non-TH-GM-mediating factor (e.g., TNF-α, IL-6, IL-1β).
  • In related aspects, the present disclosure also provides a method of prognosing progression of an inflammatory disorder in a patient in need thereof. In some embodiments, the method comprises a) quantifying a polypeptide or nucleic acid level of a TH-GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof, in a first sample collected from a patient suffering from an inflammatory disorder, and b) quantifying a polypeptide or nucleic acid level of, e.g., TNF-α, IL-6, or IL-1β, or a combination thereof, in a second sample collected from the patient, wherein i) an increased level of the TH-GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof relative to a first reference level and an unchanged level of TNF-α, IL-6, or IL-1β, or a combination thereof relative to a second reference level indicates that the patient is in an early stage of the inflammatory disorder, as described herein; or ii) an unchanged level of the TH-GM-mediating factor, such as, e.g., STAT5, IL-7, GM-CSF or IL-3, or a combination thereof relative to the first reference level and an increased level of TNF-α, IL-6, or IL-43, or a combination thereof relative to the second reference level indicates that the patient is in a late stage of the inflammatory disorder, as described herein. In some embodiments, the method further comprises administering an effective amount of an agent that modulates TH-GM function and/or, e.g., a TNF-α therapy, as described herein.
  • In some embodiments, the first sample and the second sample are the same.
  • In various aspects, the present disclosure also provides an isolated population of GM-CSF-secreting T-helper cells (TH-GM). In one embodiment, the TH-GM cells are differentiated from a precursor cell (e.g., CD4+ cells) in the presence of signal transducer and activator of transcription 5 (STAT5) and/or IL-7, and wherein the TH-GM cells express GM-CSF and IL-3.
  • In some embodiments, the TH-GM cells are differentiated from a precursor cell (e.g., CD4+ cells) in the presence of an agent that inhibits IL-12, IFN-γ. TGF-β, and/or IL-6. Similarly, the differentiation of a precursor cell (e.g., CD4+ precursor cell) into a TH-GM cell is inhibited by IL-12, IFN-γ, TFG-β, and/or IL-6.
  • In some embodiments, the TH-GM cells are differentiated from a precursor cell in vitro, under artificial conditions, but wherein the TH-GM cells retain physiological properties as described herein.
  • In some embodiments, the TH-GM cells are further characterized by an overexpression of one or more genes listed in Table 1. For example, the TH-GM cells are further characterized by an overexpression of, for example, basic helix-loop-helix family, member e40 (BHLHe40), preproenkephalin (PENK), IL-2, serine (or cysteine) peptidase inhibitor, Glade B member 6 h (Serpinb6b), neuritin 1 (Nrn1), stearoyl-Coenzyme A desaturase 1 (Scd1), or phosphotriesterase related C1q-like 3 (Pter), or a combination thereof.
  • In some embodiments, the TH-GM cells are further characterized by an underexpression of one or more genes listed in Table 1. For example, the TH-GM cells are further characterized an underexpression of lymphocyte antigen 6 complex, locus A (Ly6a); CD27; or selectin, lymphocyte (Sell).
  • As described herein, the identification of a distinct network of factors (unique from factors known to mediate T H1 or TH17) that mediate TH-GM function (e.g., its differentiation and pathogenicity) enables targeted modulation of TH-GM function to treat TH-GM-mediated disorders, e.g., disorders that result from aberrant TH-GM function. Thus, in some aspects, the present disclosure provides a method of modulating TH-GM function, comprising contacting the TH-GM, or cluster of differentiation 4 (CD4+) precursor cells, or both, with a modulating agent that modulates TH-GM function. In one embodiment, the modulating agent is contacted with the TH-GM cells or CD4+ precursor cells in vitro or in VIVO.
  • As used herein, “TH-GM function” refers to the commitment, development, maintenance, survival, proliferation, or activity, or a combination thereof, of TH-GM cells. Thus, an agent that modulates (e.g., enhances or inhibits) TH-GM function is one that modulates TH-GM commitment, development, survival, proliferation, or activity, or combination thereof, of TH-GM cells. For example, TH-GM function can be modulated by modulating its: commitment from a CD4+ precursor T cell; development of a CD4+ precursor cell that has been committed to the TH-GM developmental pathway; maintenance of a TH-GM phenotype; survival or proliferation under development or effector TH-GM cells; and/or activity of effector TH-GM cells (e.g., modulating function of a secreted factor such as GM-CSF or IL-3). For example, a modulation in TH-GM function includes, but is not limited to, a modulation in: the number of TH-GM cells; the survival of TH-GM cells; the proliferation of TH-GM cells; and/or the activity of TH-GM cells. The activity of TH-GM cells herein includes the activity induced by the cytokines, chemokines, growth factors, enzymes and other factors secreted by TH-GM cells, as described herein, and the activity induced by direct contact with TH-GM cells.
  • As used herein, a T helper subset cell “TH-GM” refers to a cell that, similar to T H1 and T H17 cells, differentiates from precursor CD4+ precursor cells, but which commits and develops through a pathway that is mediated by a subset of factors (the TH-GM-mediating factors) that is distinct and unique from the known subset of factors that commit and develop T H1 or T H17 cell subtypes, as described herein. In some embodiments, a TH-GM cell produces a distinct and unique set of genes (see, e.g., Table 1) and effects pathogenicity through a different mechanism and pathway than the known factors that mediate pathogenicity of T H1 or T H17 cell subtypes. For example, a TH-GM cell commits and develops by IL-7/STAT5 function (its regulators), and effects pathogenicity by GM-CSF/IL-3 (its effectors).
  • In some aspects, the present disclosure provides a method of treating a TH-GM-mediated inflammatory disorder in a patient in need thereof, comprising administering to said patient an effective amount of a modulating agent that modulates TH-GM cell function. In certain embodiments, the patient is previously diagnosed as having a TH-GM-mediated inflammatory disorder, as described herein.
  • In some aspects, the present disclosure also provides a method of treating rheumatoid arthritis in a patient who exhibits limited response to TNF-α therapy, comprising administering to said patient an effective amount of a modulating agent that modulates TH-GM function.
  • As used herein, “limited response” refers to no response or insignificant response such that a patient is not treated by the therapy, as determined by clinical standards.
  • “Treatment” or “treating” refers to therapy, prevention and prophylaxis and particularly refers to the administration of medicine or the performance of medical procedures with respect to a patient, for either prophylaxis (prevention) or to reduce the extent of or likelihood of occurrence of the condition or event in the instance where the patient is afflicted. It also refers to reduction in the severity of one or more symptoms associated with the disease or condition. In the present application, it may refer to amelioration of one or more of the following: pain, swelling, redness or inflammation associated with an inflammatory condition or an autoimmune disease. As used herein, and as well-understood in the art, “treatment” is an approach for obtaining beneficial or desired results, including clinical results. For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, alleviation or amelioration of one or more symptoms, diminishment of extent of disease, stabilized (e.g., not worsening) state of disease, delay or slowing of disease progression, and/or amelioration or palliation of the disease state. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • An “effective amount” of an agent is that amount sufficient to effect beneficial or desired results, including clinical results. An “effective amount” depends upon the context in which it is being applied. In the context of administering a composition that modulates an autoimmune response, an effective amount of an agent which is a modulator of TH-GM function is an amount sufficient to achieve such a modulation as compared to the response obtained when there is no agent administered. An effective amount can confer immediate, short term or long term benefits of disease modification, such as suppression and/or inhibition of TH-GM function, as defined herein. An effective amount can be administered in one or more administrations. An “effective amount” as used herein, is intended to mean an amount sufficient to reduce by at least 10%, at least 25%, at least 50%, at least 75%, or an amount that is sufficient to cause an improvement in one or more clinically significant symptoms in the patient.
  • In some embodiments, the modulating agent inhibits TH-GM function to, e.g., reduce inflammation. The inhibition conferred by the modulating agent (the inhibitor) does not imply a specific mechanism of biological action. Indeed, the term “antagonist” or “inhibitor” as used herein includes all possible pharmacological, physiological, and biochemical interactions with factors that mediate TH-GM function (e.g., IL-7, IL-7 receptor, STAT5, GM-CSF, IL-3, IL-2, IL-2 receptor, PENK, RANKL, JAK1/3, or any of the genes that are differentially expressed in TH-GM cells, e.g., genes in Tables 1 and 2), whether direct or indirect, and includes interaction with a factor (or its active fragment) that mediates TH-GM function at the protein and/or nucleic acid level, or through another mechanism.
  • In certain embodiments, a modulating agent that inhibits TH-GM function includes an antibody, a polypeptide (e.g., a soluble receptor that hinds and inhibits, for example, IL-7), a small molecule, a nucleic acid (e.g., antisense, small interfering RNA molecule, short hairpin RNA, microRNA), or a protein (e.g., cytokine), or a combination thereof that prevents the function (e.g., expression and/or activity) of a factor that mediates TH-GM function. Methods of designing, producing, and using such inhibitors are known and available in the art.
  • As used herein, “binds” is used interchangeably with “specifically binds,” which means a polypeptide (e.g., a soluble receptor) or antibody which recognizes and hinds a polypeptide of the present disclosure, but that does not substantially recognize and bind other molecules in a sample, for example, a biological sample, which naturally includes a polypeptide of the present disclosure. In one example, an antibody specifically binds an activated STAT5 polypeptide does not hind a non-STAT5 polypeptide.
  • As used herein, “antibody” refers to an intact antibody or antigen-binding fragment of an antibody, including an intact antibody or antigen-binding fragment that has been modified or engineered, or that is a human antibody.
  • In a particular embodiment, the antibody binds to and inhibits the function of any one or more of the factors that mediate TH-GM function. For example, the antibody hinds to and inhibits the function of IL-7, IL-7 receptor (IL-7R), IL-2, IL-2 receptor (IL-2R), STAT5 or janus kinase 1/3 (JAK1/3), or a combination thereof. In other examples, the antibody hinds to and inhibits the function of GM-CSF (or its receptor), IL-3, PENK, or RANKL, or a combination thereof. In some embodiments, the antibody binds to and inhibits the function of a gene listed in Table 1. In some embodiments, the antibody hinds to and inhibits the protein or any functional fragment thereof. Methods of designing, producing and using suitable antibodies are known and available to those of skill in the art. Examples of antibodies suitable for use in the present disclosure include. e.g., daclizumab, basiliximab, mavrilimumab, MOR103, KB003, namilumab, and MOR Ab-022.
  • The terms “protein” and “polypeptide” are used interchangeably, and can include full-length polypeptide or functional fragments thereof (e.g., degradation products, alternatively spliced isoforms of the polypeptide, enzymatic cleavage products of the polypeptide), the polypeptide hound to a substrate or ligand, or free (unbound) forms of the polypeptide. The term “functional fragment”, refers to a portion of a full-length protein that retains some or all of the activity (e.g., biological activity, such as the ability to bind a cognate ligand or receptor) of the full-length polypeptide.
  • In some embodiments, the modulating agent that inhibits TH-GM function can be a particular biological protein (e.g., cytokines) that inhibits, directly or indirectly, one or more of the factors that mediate TH-GM function. Such cytokines include, e.g., IL-12, IFN-γ, TGF-β, and IL-6.
  • In some embodiments, the modulating agent that inhibits TH-GM function can be a small molecule that inhibits, directly or indirectly, one or more of the factors that mediate TH-GM function. As used herein a “small molecule” is an organic compound or such a compound complexed with an inorganic compound (e.g., metal) that has biological activity and is not a polymer. A small molecule generally has a molecular weight of less than about 3 kilodaltons. Examples of known small molecules include CAS 285986-31-4 (Calbiochem), pimozide, and tofacitinib.
  • In other embodiments, the modulating agent enhances TH-GM function in disorders such as, e.g., viral, fungal and bacterial infections, cancers and/or conditions associated with therewith. In one embodiment, modulating agents that enhance TH-GM function include. e.g., CD28 activator; IL-7 and/or IL-2 on naïve (precursor) CD4+ T cells; activator of STAT5; or effectors of TH-GM cells (e.g., GM-CSF. IL-3).
  • In another aspect, the present disclosure provides a method of treating a STAT5-mediated inflammatory disorder in a patient in need thereof, comprising administering to the patient an effective amount of an agent that modulates STAT5 function.
  • As used herein, “STAT5-mediated” inflammatory disorder refers to an inflammatory disorder that is caused by aberrant STAT5 function (aberrantly enhanced or inhibited), and which is responsive to modulation of STAT5 function, as determined by clinical standards. In some embodiments, the STAT5 is activated STAT5 (e.g., phospho-STAT5, Tyr694).
  • In some embodiments, the inflammatory disorder is an autoimmune disorder. In certain embodiments, the inflammatory disorder can be any inflammatory disorder mediated by STAT5 (e.g., activated STAT5), and includes, but is not limited to rheumatoid arthritis, multiple sclerosis, ankylosing spondylitis. Crohn's disease, diabetes. Hashimoto's thyroiditis, hyperthyroidism, hypothyroidism, Irritable Bowel Syndrome (IBS), lupus erythematosus, polymyalgia rheumatic, psoriasis, psoriatic arthritis. Raynaud's syndrome/phenomenon, reactive arthritis (Reiter syndrome), sarcoidosis, scleroderma. Sjögren's syndrome, ulcerative colitis, uveitis, or vasculitis.
  • In some embodiments, the term “patient” refers to a mammal, preferably human, but can also include an animal in need of veterinary treatment, e.g., companion animals (e.g., dogs, cats, and the like), farm animals (e.g., cows, sheep, pigs, horses, and the like), and laboratory animals (e.g., rats, mice, guinea pigs, and the like).
  • In some embodiments, the agent inhibits STAT5 function (e.g., expression and/or activity). Examples of agents that inhibit STAT5 (e.g., activated STAT5, Tyr694) are described herein.
  • In certain embodiments, the methods of the present disclosure further comprise administering to the patient a TNF-αc therapy. In certain embodiments, TNF-α therapy is administered in a patient determined to have an inflammatory condition that is non-TH-GM-mediated. As described herein, in certain embodiments, a TNF-α therapy is administered if a quantified TNF-α level is increased by, e.g., at least 40% relative to a reference level.
  • Examples of TNF-α therapy include those that are TNF-α-inhibitor based, and those that are non-TNF-α-inhibitor based. In particular, TNF-α-inhibitor based therapy includes etanercept, adalimuinab, infliximab, golimumab, and certolizumab pegol. Examples of non-TNF-α-inhibitor based therapy includes corticosteroid medications (e.g., prednisone), nonsteroidal anti-inflammatory drugs (e.g., methotrexate), and JAK inhibitors (e.g., tofacitinib). Other examples of non-TNF-α-inhibitor based therapy include anakinra, abatacept, rituximab and tocilizumab.
  • The TNF-α therapy can be administered before, simultaneously with, or alter the administration of an effective amount of an agent that modulates TH-GM function. Accordingly, an agent that modulates TH-GM function and the TNF-α therapy can be administered together in a single dose, or can be administered in separate doses, e.g., either simultaneously or sequentially, or both. The duration of time between the administration of an agent that modulates TH-GM function and a TNF-α therapy will depend on the nature of the therapeutic agent(s). In addition, an agent that modulates TH-GM function and a TNF-α therapy may or may not be administered on similar dosing schedules. For example, the agent that modulates TH-GM function and the TNF-α therapy may have different half-lives and/or act on different time-scales such that the agent that modulates TH-GM function is administered with greater frequency than the TNF-α therapy, or vice-versa. The number of days in between administration of therapeutic agents can be appropriately determined by persons of ordinary skill in the art according to the safety and pharmacodynamics of each drug.
  • The identification of the TH-GM cells as well as the identification of genes differentially produced by TH-GM cells relative to T H1 or T H17 enables the use of TH-GM cells to identify novel therapeutics for modulating TH-GM function, thereby enabling new therapeutics for treating TH-GM-mediated disorders (e.g., inflammatory disorders). Thus, in further aspects, the present disclosure provides a method of screening to identify a modulator of TH-GM cell function, comprising contacting an isolated population of TH-GM cells, or an isolated population of CD4+ precursor cells, with a candidate agent, and measuring a readout of TH-GM function in the presence or absence of the candidate agent, wherein a change in the readout of TH-GM function indicates that the candidate agent is a modulator of TH-GM function.
  • As used herein, a candidate agent refers to an agent that may modulate TH-GM function by modulating the function (e.g., expression and/or activity) of a factor that mediates TH-GM function. Such candidate agents include, e.g., an antibody, a peptide, a small molecule, a nucleic acid (e.g., antisense, small interfering RNA molecule), or a protein (e.g., cytokine), or a combination thereof. A candidate agent can be designed to target any of the factors (at the protein and/or nucleic acid level) that mediate TH-GM function, as described herein, including the genes listed in Table 1 (e.g., genes preferentially upregulated in TH-GM cells, genes preferentially overexpressed/underexpressed on the surface of TH-GM cells).
  • As used herein, “readout” refers to any change (or lack of change) in TH-GM function that can be measured or quantified. For example, a candidate agent can be assessed for its effect on, e.g., GM-CSF secretion by TH-GM cells, or its effect on the abundance of TH-GM cells (through an effect on the commitment/development/proliferation of TH-GM cells), as described herein. Assays for determining such readouts are known and available in the art, and are exemplified herein.
  • In some embodiments, the change in the presence of the candidate agent is a reduction in the measurement of the readout, indicating an inhibition of TH-GM function (e.g., decrease in GM-CSF or IL-3 production, or decrease in the abundance of TH-GM cells), thereby identifying the candidate agent as an inhibitor of TH-GM function.
  • In certain embodiments, the change in the presence of the candidate agent is an increase in the measurement of the readout, indicating an enhancement of TH-GM function (e.g., increase in GM-CSF or IL-3 production, or increase in the abundance of TH-GM cells), thereby identifying the candidate agent as an enhancer of TH-GM function.
  • In some embodiments, the readout can be any one or more of the genes listed in Tables 1 and 2 which are preferentially upregulated or downregulated in TH-GM cells. Thus, a candidate agent that downregulates a gene that is preferentially upregulated in a TH-GM cell is a inhibitor of TH-GM function. Similarly, a candidate agent that upregulates a gene that is preferentially downregulated in a TH-GM cell is an enhancer of TH-GM function.
  • In certain aspects, the method of screening, if performed with precursor CD4+ cells, is performed under TH-GM polarizing conditions, as described herein. For example, the method can be performed in the presence of IL-7/STAT5, TCR activation, CD28 co-stimulation, in combination with the blockade of IFN-gamma and IL-4.
  • Unless indicated otherwise, the definitions of terms described herein apply to all aspects and embodiments of the present disclosure
  • The practice of the present disclosure includes use of conventional techniques of molecular biology such as recombinant DNA, microbiology, cell biology, biochemistry, nucleic acid chemistry, and immunology as described for example in: Molecular Cloning: A Laboratory Manual, second edition (Sambrook et al., 1989) and Molecular Cloning: A Laboratory Manual, third edition (Sambrook and Russel, 2001), jointly and individually referred to herein as “Sambrook”); Oligonucleotide Synthesis (M. J. Gait, ed., 1984); Animal Cell Culture (R. I. Freshney, ed., 1987); Handbook of Experimental Immunology (D. M. Weir & C. C. Blackwell, eds.); Gene Transfer Vectors for Mammalian Cells (J. M. Miller & M. P. Calos, eds., 1987); Current Protocols in Molecular Biology (F. M. Ausubel et al., eds., 1987, including supplements through 2001); PCR: The Polymerase Chain Reaction, (Mullis et al., eds., 1994); Current Protocols in Immunology (J. E. Coligan et al., eds., 1991); The Immunoassay Handbook (D. Wild. ed., Stockton Press NY, 1994); Bioconjugate Techniques (Greg T. Hermanson, ed., Academic Press, 1996); Methods of Immunological Analysis (R. Masseyeff, W. H. Albert, and N. A. Staines, eds., Weinheim: VCH Verlags gesellschaft mbH, 1993), Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York. and Harlow and Lane (1999) Using Antibodies: A Laboratory Manual Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. jointly and individually referred to herein as “Harlow and Lane”), Beaucage et al. eds., Current Protocols in Nucleic Acid Chemistry (John Wiley & Sons, Inc., New York, 2000); and Aerawal, ed., Protocols for Oligonucleotides and Analogs, Synthesis and Properties (Humana Press Inc., New Jersey, 1993).
  • EXEMPLIFICATION
  • Methods
  • Mice
  • Stat5f/f mice were provided by L. Hennighausen (National Institute of Diabetes and Digestive and Kidney Diseases). Stat3f/f mice were generated as described2. Cd4-Cre transgenic mice were purchased from Taconic Farms. Rag2−/− mice were obtained from Jean-Pierre Abastado (Singapore Immunology Network). All mice are on a C57BL/6 genetic background and housed under specific-pathogen-free conditions at National University of Singapore. All experiments were performed with mice 6˜8 weeks old and approved by the Institutional Animal Care and Use Committee of NUS.
  • Patients and Controls
  • Blood samples (n=47) and synovial fluid samples (n=3) were collected from RA patients admitted to the Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School. All patients fulfilled the American College of Rheumatology criteria for the classification of RA. Age and gender matched healthy controls (n=32) were obtained from Medical Examination Center of the Affiliated Drum Tower Hospital. The study protocol was approved by the Ethics Committee of the Affiliate Drum Tower Hospital of Nanjing University Medical School.
  • In Vitro T Cell Differentiation
  • CD4+ T cells were obtained from spleens and lymph nodes by positive selection and magnetic separation (Miltenyi Biotech), followed by purification of naïve CD4+ T cell population (CD4+CD25CD62LhiCD44lo) sorted with FACS Aria. Naïve CD4+ T cells were stimulated with plate-hound anti-CD3 (3 μg/ml; BD Pharmingen) and anti-CD28 (1 μg/ml; BD Pharmingen) in presence of different combinations of neutralizing antibodies and cytokines for 3˜4 days: for neutral conditions, no addition of any cytokine or neutralizing antibody; for T H 1 conditions, IL-12 (10 ng/ml), and anti-IL-4 (10 μg/ml, BD Pharmingen); for T H17 conditions, hTGF-β (3 ng/ml), IL-6 (20 ng/ml), anti-IFN-γ (10 μg/ml, eBioscience), and anti-IL-4 (10 μg/ml); for an alternative T H17 conditions, IL-6 (20 ng/ml), IL-23 (10 ng/ml), IL-1 (10 ng/ml), anti-IFN-γ (10 μg/ml), and anti-IL-4 (10 μg/ml). For GM-CSF-expressing cell differentiation, naïve CD4+ T cells were stimulated with plate-bound anti-CD3 (2 μg/ml) and soluble anti-D28 (1 μg/ml) with the addition of IL-7 and/or anti-IFN-γ (10 μg/ml) as indicated. All cytokines were obtained from R&D Systems. All cells were cultured in RPMI 1640 supplemented with 10% FBS, 100 units/ml penicillin, 0.1 mg/ml streptomycin, 1 mM sodium pyruvate, 0.1 mM nonessential amino acid and 5 μM beta-mercaptoethanol. After 3˜4 days polarization, cells were washed and restimulated with phorbol 12-myristate 13-acetate (PMA) and ionomycin in presence of Golgiplug for 4-5 h, followed by fixation and intracellular staining with a Cytofix/Cytoperm kit from BD Pharmingen. Foxp3 staining was done with a kit from eBioscience. Cells were acquired on the LSR II (BD Biosciences) and analyzed with FlowJo software (Tree Star).
  • EAE Induction
  • EAE induction procedures were modified from previous report3. For active EAE induction, mice were immunized in two sites on the hind flanks with 300 μg MOG35-55 in 100 μl CFA containing 5 mg/ml heat-killed M. tuberculosis strain H37Ra (Difco) at day 0 and day 7. Pertussis toxin (List Bio Lab) was administrated intraperitoneally at the dosage of 500 ng per mouse at day 1 and day 8. For single MOG35-55/CFA immunization, the similar procedure was performed at day 0 and day 1 only. In an alternative active EAE induction, LPS (600 μg/ml in IFA, O111:B4 from Sigma) was used as adjuvant. For active EAE induction in Rag2−/− mice, CD4+ T cells derived from Stat5f/f or Cd4-Cre; Stat5f/f mice were transferred, followed by MOG35-55/CFA immunization as described above. Clinical symptoms were scored as follows: 0, no clinical sign; 1, loss of tail tone; 2, wobbly gait; 3, hind limb paralysis; 4, hind and fore limb paralysis; 5, death. IL-7Rα neutralizing antibody (SB/14, BD Pharmingen) and isotype control was administrated intraperitoneally at 200 μg per mouse every other day. For analysis of CNS-infiltrating cells, both spinal cord and brain were collected and minced from perfused mice, and mononuclear cells were isolated by gradient centrifuge with Percoll (GE Healthcare).
  • For passive EAE induction with Stat5+/+ or Stat5−/− T cells, splenocytes and LNs were harvested 10-14 days post-immunization and passed through a 70 μm cell strainer (BD Falcon). Cells were cultured in vitro for 3 days with MOG35-55 (20 μg/ml) in the presence of IL-23 (5 ng/ml) and IL-1β (2 ng/ml). After harvesting, CD4+ T cells were purified by positive selection to a purity >90%. CD4+ T cells (2 million in sterile PBS) were injected intraperitoneally into Rag2−/− mice, followed by Pertussis toxin administration on the following day. Mice were observed daily for the signs of EAE as described above. For EAE induction by transferring various TH subsets, similar procedures was performed as described above. Different subsets skewing conditions were as follows: Non-skewed, MOG35-55 only; TH1: MOG35-55 plus IL-12 (10 ng/ml) and anti-IL-4 (5 μg/ml); TH17: MOG35-55 plus TGF-β (3 ng/ml), IL-6 (10 ng/ml), anti-IFN-γ (5 μg/ml) and anti-IL-4 (5 μg/ml); GM-CSF-expressing TH: MOG35-55 plus IL-7 (2 ng/ml) and anti-IFN-γ (5 μg/ml). 6×105 CD4+ T cells were transferred per recipient mouse.
  • Antigen-Induced Arthritis (AIA)
  • Briefly, mice were immunized subcutaneously in two sites on the hind flanks with 100 μg methylated bovine serum albumin (mBSA, Sigma) in 100 μl complete Freund's adjuvant (CFA) containing 5 mg/ml heat-killed M. tuberculosis strain H37Ra (Difco) at day 0. Pertussis toxin (List Bio Lab) was administrated intraperitoneally at the dosage of 250 ng per mouse at day 1. Arthritis was induced by intraarticular injection of 100 μg mBSA (in 10 saline) into the hind right knee joint at day 7 after immunization. The hind left knee joint was injected with same volume of saline as control. Joint swelling was recorded by measuring the difference between right and left knee joint diameters with a caliper over 7 days after arthritis induction. To assess the effect of GM-CSF administration, MA was induced by intraarticular injection of mBSA alone to the right knee joint or mBSA supplemented with 100 ng GM-CSF (ImmunoTools) to the left knee joint. To assess the effect of GM-CSF and/or TNF-α blockade, mice were administrated intraperitoneally with neutralizing antibodies (100 μg for each antibody per mouse) specific for GM-CSF (MP1-22E9, BD Pharmingen) and/or TNF-α (MP6-XT3. BD Pharmingen) at indicated times.
  • For AIA induction by adoptive transfer, splenocytes and inguinal LN cells were isolated from mBSA/CFA-immunized mice at day 7, and cultured in vitro with mBSA (10 μg/ml) in the presence of IL-7 (2 ng/ml) for 3 days. After harvesting, CD4+ T cells were purified by positive selection (Miltenyi Biotec) to a purity >90%. Then CD4+ T cells (1 million in sterile PBS) were transferred into WT naïve mice, followed by intraarticular injection of mBSA on the next day.
  • Collagen-Induced Arthritis (CIA)
  • CIA was induced in a similar procedure as AIA as described above, by immunizing mice with chicken collagen II/CFA emulsion (purchased from Chondrex, Inc), followed with pertussis toxin injection. Mice were monitored and scored for arthritis; 0, normal; 1, mild swelling of ankle or wrist, or apparent swelling limited to individual digits; 2, moderate swelling of entire paw; 3, severe swelling of entire paw with ankylosis. Scores for four limbs were summed for each mouse.
  • Histological Analysis
  • For paraffin-embedded tissues, spinal cords were fixed in 4% PFA. Knee joints or paws were removed, fixed in 10% formalin and decalcified in 5% formic acid before dehydration and embedding. Sections (5 μm) were stained with hematoxylin and eosin (H&E) to assess immune cell infiltration and inflammation, or with Safranin-O/Fast Green to assess cartilage depletion. For frozen tissues, spinal cords were embedded in OCT (Tissue-Tek) and snap frozen on dry ice. Sections (10 μm) were fixed in ice-cold acetone and stained with primary anti-CD4 (Biolegend) and anti-CD11b (eBioscience), followed by incubation with fluorescence-conjugated secondary antibodies (Invitrogen). For ALA experiments, knee joint were fixed in 10% formalin for 5 days, followed by decalcification in 5% formic acid for 5 clays. Sections (10 μm) were stained with hematoxylin and eosin (H&E) to assess immune cell infiltration and inflammation, or stained with Safranin-O/fast green to access cartilage destruction.
  • Cell Sorting and May Grünwald-Giemsa Staining
  • Monocytes/macrophages (Ly6ChiLy6G) and neutrophils (Ly6CloLy6Ghi) gated on CD45+CD11b+ were sorted with FACS Aria from spleens or synovial single cell suspensions. Sorted cells were cytospun onto glass slides and subsequently stained with May Grünwald and Giemsa dye following a standard procedure.
  • Real-Time PCR
  • Total RNA was extracted from cells with RNeasy kit (Qiagen) according to the manufacturer's instruction. Complementary DNA (cDNA) was synthesized with Superscript reverse transcriptase (Invitrogen). Gene expressions were measured by 7500 real-time PCR system (Applied Biosystems) with SYBR qPCR kit (KAPA). Actinb, Gapdh or Rn18S was used as internal control. The primer sequences are available upon request.
  • ELISA
  • TNF-α, IL-6, IL-1β, IFN-γ, GM-CSF and IL-2 levels were assayed by Ready-SET-Go ELISA kit (eBioscience), and IL-17 level was measured by DuoSet ELISA kit (R&D Systems) according to the manufactures' instructions.
  • Chromatin Immunoprecipitation Assays
  • CD4+ T cells isolated from Stat5f/f or Cd4-Cre; Stat5f/f mice were activated with plate-bound anti-CD3 and anti-CD28 for 3 days. Cells were stimulated with IL-7 (20 ng/ml) or IL-2 (25 ng/ml) for 45 min. Crosslink was performed by addition of formaldehyde at final concentration of 1% for 10 min followed by quenching with Glycine. Cell lysates were fragmented by sonication and precleared with protein G Dynabeals, and subsequently precipitated with anti-STAT5 antibody (Santa Cruz) or normal rabbit IgG (Santa Cruz) overnight at 4° C. After washing and elution, crosslink reversal was clone by incubating at 65° C. for 8 hr. The eluted DNA was purified and analyzed by RT-PCR with primers specific to C42 promoter as described previously5.
  • Statistics
  • Statistical significance was determined by Student's t test using GraphPad Prism 6.01. The p value <0.05 was considered significant. The p values of clinical scores were determined by two-way multiple-range analysis of variance (ANOVA) for multiple comparisons. Unless otherwise specified, data were presented as mean and the standard error of the mean (mean±SEM).
  • Example 1. Stat5 Conditional Knockout Mice are Resistant to EAE
  • STAT5 negatively regulates T H17 differentiation by restraining IL-17 production (Laurence et al., 2007; Yang et al., 2011). However, the function of STAT5 in TH17-mediated pathogenesis is not well understood. To explore this question, EAE was induced in Cd4-Cre; Stat5f/f (Stat5−/−) mice, where Stat5 was specifically deleted in T cell compartment, and in littermate controls by immunizing the mice with MOG35-55/CFA at day 0 and day 7. Development of paralysis was assessed by daily assignment of clinical scores. Surprisingly, diminished occurrence and severity of clinical disease in Stat5−/− mice was observed (FIGS. 1A and 1B), a result that was opposite to expectations based on an antagonistic role for STAT5 in T H17 generation. Similar results were observed when a single MOG35-55/CFA immunization was performed or replaced CFA with LPS as the adjuvant to induce EAE (FIGS. 1C and 1D). Consistent with reduced EAE disease in Stat5−/− mice, a remarkable reduction of immune cell infiltration in the spinal cord of Stat5−/− mice was observed (FIG. 2A). Furthermore, the infiltration of various immune cell populations, including CD4+, CD8+. B220+ and CD11b+ cells was reduced in Stat5−/− mice (FIGS. 2B-D and data not shown). However, the frequencies of IL-17+ and IFN-γ+ cells among CD4+ T cells in the CNS were comparable between Stat5+/+ and Stat5−/− mice (FIG. 3A), suggesting the resistance to EAE in Stat5−/− mice is independent of T H1 and T H17 cell development. Nevertheless, decreased CD4+CD25+ Treg population in the CNS of Stat5−/− mice was detected (FIG. 3B), indicating the resistance to EAE in Stat5−/− mice was unlikely due to altered Treg, cells.
  • Example 2. Resistance to EAE in Stat5-Mutant Mice is Due to an Intrinsic Defect of Antigen Specific CD4+ T Cells Independent of T H1 and T H17 Generation
  • Stat5 deletion (Cd4-cre; Stat5f/−) mice was reported to develop peripheral lymphopenia, with a reduction of both CD4+ and CD8+ T cells (Yao et al., 2006). However, another study showed that Stat5 deletion (Cd4-cre; Stat5f/f) did not affect the proportion of peripheral CD4+ T cells (Burchill et al., 2007). In the experimental setting, a change in the absolute number of peripheral CD4+ T cells was not detected by Stat5 deletion during EAE development (FIGS. 4A and 4B), suggesting the resistance to EAE in Stat5−/− mice was not caused by peripheral lymphopenia. Furthermore, increased frequencies of IL-17+ and IFN-γ+ cells were detected among CD4+ T cells in spleens of Stat5−/− mice (FIG. 4C), which further support the idea that the resistance to EAE in Stat5−/− mice is likely independent of T H1 and T H17 generation. To validate the function of STAT5 in T H1 and T H17 generation, the in vitro differentiation was performed by activating naïve CD4+ T cells under TH1- and TH17-polarizing conditions. In agreement with previous reports, that STAT5 mediated the suppressive effect of IL-2 on T H17 differentiation (data not shown). Interestingly, IL-7, which also signals through STAT5, was not observed to have a demonstrable effect on T H17 differentiation (data not shown). Nevertheless, a slight decrease of IFN-γ+ cells under TH1-polarizing condition was observed when STAT5 was deleted (data not shown).
  • To confirm if the resistance of EAE in Stat5+/+ mice is mediated by CD4+ T cells, Rag2−/− mice were reconstituted with Stat5+/+ or Stat5−/− CD4+ T cells followed by EAE induction. We found that Rag1−/− mice that received Stat5−/− CD4+ T cells were resistant to the disease compared with mice receiving wild-type cells (data not shown), demonstrating that Stat5−/− CD4+ T cells were impaired in their ability to promote EAE development.
  • Next, whether the lack of encephalitogenicity was caused by defects in migration of Stat5−/− CD4+ T cells to the CNS was examined. It has been shown that the chemokine receptor CCR6 is essential for T H17 cell entry into the CNS through the choroid plexus (Reboldi et al., 2009). Thus. CCR6 expression in both Stat5−/− and Stat5+/+ CD4+ T cells was examined. Increased CD4+CCR6+ cells in spleens of Stat5−/− mice compared with Stat5+/+ controls (FIG. 5A) was observed. CXCR3 and CD69 expression was also examined, which showed increased expression of both molecules in CD4+ T cells in the absence of STAT5 (FIG. 5A). These results indicate that Stat5−/− CD4+ T cells can infiltrate CNS. Furthermore, comparable number of CD4+ T cells present in the CNS of Stat5+/+ and Stat5−/− mice during EAE induction was observed (at day 7 and day 9) (FIG. 5B). However, CD4+ T cells in CNS of Stat5−/− mice dropped dramatically during disease onset (Day 21) (FIGS. 5C and 5D). Together, these results demonstrate that Stat5−/− CD4+ T cells can infiltrate CNS, but fail to induce effective inflammation in the CNS in EAE.
  • To further exclude the possibility that the resistance of Stat5-deficient mice to EAE was caused by any potential defect in the survival of autoreactive CD4+ T in the CNS, increased numbers of Stat5−/− CD4+ T cells than wild-type cells were transferred into Rag1−/− mice respectively to make sure comparable numbers of autoreactive CD4+ T cells were present in the CNS during EAE development. As shown in FIGS. 6A and 6B, despite similar numbers of CD4+ T cells in the CNS between two groups of mice, reduced disease severity was nevertheless observed in mice receiving Stat5-deficient CD4+ T cells. Additionally, certain numbers of Stat5-deficient mice containing similar numbers of CD4+ T cells in the CNS as wild-type mice at peak of EAE disease were observed, yet, they were relatively resistant to EAE compared with those wild-type mice (FIG. 6C), further suggesting that the resistance to EAE disease in Stat5-deficient mice was unlikely due to impaired CD4+ T cell survival in the CNS.
  • To further develop a causal link between these observations and the intrinsic impairment of Stat5−/− CD4+ T cells, MOG35-55-specific Stat5+/+ and Stat5−/− CD4+ T cells were transferred into Rag2−/− mice separately without further immunization to test if these cells were able to mediate EAE development. As shown in FIGS. 7A and 7B, mice receiving Stat5+/+ CD4+ T cells spontaneously developed EAE disease 1 week after transfer. In contrast, mice receiving Stat5−/− CD4+ T cells had significantly reduced disease severity and incidence. The frequencies of IL-17+ and IFN-γ+ cells among CD4+ T cells in the CNS of Rag2−/− mice were comparable between two groups (FIG. 7C), further suggesting that the intrinsic defect in encephalitogenicity of Stat5−/− CD4+ T cells is independent of T H1 and T H17. To exclude the possible role of CD8+ T cells in the resistance to EAE observed in Stat5 mice, Rag2−/− mice were reconstituted with MOG35-55-specific Stat5+4 or Stat5−/− CD4+ T cells together with equal numbers of Stat5+/+ CD8+ T cells. The transfer of Stat5−/− CD4+ together with Stat5+/+ CD8+ T cells still failed to induce EAE (data not shown). Together, these data demonstrate that Stat5+/+ CD4+ T cells have intrinsic defect in encephalitogenicity. The relevant teachings of all patents, published applications and references cited herein are incorporated by reference in their entirety.
  • Example 3. Diminished Expression of GM-CSF in Stat5−/− CD4+ T Cells
  • To test whether GM-CSF production was impaired by Stat5 deletion, its expression was examined in MOG35-53-specific Stat5+/+ and Stat5+/+ CD4+ T cells. Splenocytes derived from MOG35-55/CFA-immunized Stat5+/+ and Stat5−/− mice were challenged with various concentrations of MOG35-55 for 24 h, to examine the secretion of GM-CSF. GM-CSF production was observed to increase in a MOG35-55 dose-dependent manner in Stat5+/+ cells (FIG. 8A). In contrast, GM-CSF production was severely diminished in Stat5+/− cells in all conditions. To further validate this, splenocytes derived from mice were stimulated during the development of EAE with PMA/Ionomycin in the presence of GolgiPlug for GM-CSF and IL-17 intracellular staining. Although IL-17 expression was enhanced in Stat5−/− cells, a significantly reduced proportion of GM-CSF+IL-17″ and GM-CSF+IL-17+ cells was observed among CD4+CD44hi cells in the absence of STAT5 (FIG. 8B). Moreover, the frequency of MOG35,55-specific GM-CSF+ T cells was also significantly reduced in spleens of Stat5−/− mice (FIG. 8C). Together, these results indicate that STAT5 is required for GM-CSF expression in autoreactive CD4+ T cells. However, STAT5, an important transcription factor in T H17 differentiation, was required for GM-CSF expression (FIG. 8D).
  • Next. GM-CSF induction in the CNS during EAE development was examined. Although IL-17 and IFN-γ production by CNS-infiltrating CD4+ T cells was not impaired by Stat5 deficiency, a diminished frequency of CD4+GM-CSF+ cells in the CNS of Stat5−/− mice was detected compared with control mice (FIG. 9A). Further analysis showed a reduced GM-CSF+ percentage among CD4+IL-17; cells and among CD4+IFN-γ+ cells (FIG. 9A). Similarly, Rag2−/− mice transferred with MOG35-55-specific Stat5−/− CD4+ T cells also showed a reduced frequency of CD4+GM-CSF+ T cells in the CNS compared with control mice (FIG. 9B). GM-CSF mRNA expression in the CNS of Stat5−/− mice was markedly lower than that of Stat5+/+ mice at day 8 after EAE induction (FIG. 9C), when comparable CD4+ T cell infiltration was detected in Stat5−/− and Stat5+/+ mice (FIGS. 5C and 5D). Meanwhile, no significant difference of IL-17 and IFN-γ expression was detected between Stat5−/− and Stat5+/+ mice (FIG. 9C). The impaired cytokine induction (IL-17 and IFN-γ) in the CNS of Stat5−/− mice at later stage (day 14, FIG. 9C) could be explained by the inability of Stat5−/− CD4+ T to sustain neuroinflammation (FIGS. 5C and 5D). Interestingly, GM-CSF induction in the CNS preceded IL-23 induction (FIG. 9C), suggesting IL-23 might not be required for GM-CSF expression in the induction phase of EAE. In summary, the results suggest that GM-CSF expression in autoreactive CD4+ T cells is regulated by STAT5 and the impaired GM-CSF production in the absence of STAT5 caused the resistance of the mice to EAE.
  • Example 4. IL-7-STAT5 Signaling Induces GM-CSF Expression in Autoreactive CD4+ T Cells and Contributes to Neuroinflammation
  • Next, the mechanism by which STAT5 regulates GM-CSF expression was investigated. As the present disclosure indicates, neither IL-23 nor IL-1β seemed to be potent STAT5 stimulators (FIG. 10A). Furthermore, IL-1R1 expression was not changed, whereas IL-23Rα expression was increased in Stat5−/− CD4+ T cells (FIG. 10B). These data suggest that the ability of STAT5 to induce GM-CSF expression is likely independent of IL-23 and IL-1β signaling. In contrast, both IL-2 and IL-7 potently activated STAT5 by inducing tyrosine phosphorylation (FIG. 10A). Therefore, the effect of these two cytokines on GM-CSF induction in autoreactive T cells was further examined. Splenocytes derived from MOG35-55-immunized wild-type mice were challenged with MOG35-55 alone or plus IL-2. GM-CSF and IL-17 production by CD4+ T cells were analyzed by intracellular cytokine staining. As shown in FIG. 10C, IL-2 showed modest effects on the frequency of GM-CSF+ T cells. In contrast, IL-7 significantly promoted GM-CSF expression in both IL-1T and IL-17+ CD4+CD44hi T cells (FIG. 11A). Furthermore, IL-7 carried out this function in a STAT5-dependent manner, as Stat5 deletion abrogated its effect on GM-CSF expression as assessed by intracellular cytokine staining and ELISA (FIG. 11A, lower panels, and FIG. 11B).
  • IL-7Rα is expressed in both CD62LhiCD44loT cells and CD62loCD44hi T cells, suggesting IL-7 may directly act on CD4+ T cells to regulate GM-CSF expression. Thus, CD62LhiCD44lo and CD62LloCD44hi T cells were sorted from Stat5−/− mice and littermate controls during EAE development, and then activated cells in the presence or absence of IL-7. As shown in FIG. 11C, CD62LloCD44hi T cells potently expressed GM-CSF, while CD62LhiCD44lo) T cells expressed 30-fold lower GM-CSF amounts. STAT5 deletion resulted in reduced basal GM-CSF production in CD62LloCD44hi T cells. As expected, IL-7 promoted GM-CSF expression in both subsets of CD4+ T cells in a STAT5-dependent manner (FIG. 11C).
  • To examine the contribution of IL-7-induced GM-CSF expression in autoreactive CD4+ T cells to EAE development, mice were treated with IL-7Rα-specific antibody (clone SB/14) during EAE development. The treatment resulted in a significant reduction of disease severity, which was accompanied with reduced CNS inflammation (FIGS. 12A and 12B). In agreement with previous report (Lee et al., 2012), this neutralizing antibody did not have T cell depleting activity (FIG. 12C). Notably, the blocking of IL-7 signaling resulted in decreased GM-CSF expression in CNS-infiltrating CD4+ T cells (FIGS. 120-12F). In summary, the present findings indicate that IL-7 induces STAT5-dependent GM-CSF expression in autoreactive CD4+ T cells, which contributes to the development of neuroinflammation.
  • Example 5. GM-CSF-Expressing TH Cells are Distinct from T H17 and T H1
  • Since both T H17 and T H1 can produce GM-CSF, it was determined if the IL-7-stimulated phenotype was related to either of these subsets. To further understand the characteristics of GM-CSF-expressing CD4+ cells, naïve CD4+ T cells were stimulated with plate-bound anti-CD3 and soluble anti-CD28 under TH1- or TH17-polarizing conditions. It was observed that anti-CD3 together with anti-CD28 induced the expression of GM-CSF (FIG. 14A). However, while T H1 differentiation conditions promoted IFN-γ expression and T H17 conditions promoted IL-17 expression as expected, both T H1 and T H17 differentiation conditions greatly suppressed the production of GM-CSF (FIGS. 13A and 13B). Conversely, IL-12 and IFN-γ neutralization promoted GM-CSF-expressing cell generation (FIG. 13A), consistent with a previous report (Codarri et al., 2011). IL-23 and IL-1β did not increase GM-CSF-expressing cell differentiation from naïve CD4+ T cells (FIG. 13A), which was consistent with the finding that naïve CD4+ T cells did not express their receptors. TGF-β inhibits GM-CSF expression (El-Behi et al., 2011). IL-6, an essential cytokine for T H17 differentiation, had a profound inhibitory effect on GM-CSF expression (FIG. 14B), indicating STAT3 could be a negative regulator. To address this, naïve CD4+ T cells were purified from Stat3−/− mice for cell differentiation. Strikingly, in the absence of STAT3, cells polarized under T H17 condition expressed GM-CSF (FIG. 14C). Interestingly, even without exogenous IL-6, STAT3 still had a moderate suppressive effect on GM-CSF-expressing cell differentiation (FIG. 14C). In addition, RORγt and T-bet have been reported unnecessary for GM-CSF expression in CD4+ T cells (El-Behi et al., 2011). Thus, the present datasupport a model wherein GM-CSF-expressing CD4+ T cells develop via a lineage distinct from T H17 and T H1.
  • Example 6. IL-7-STAT5 Programs GM-CSF-Expressing TH Cell Differentiation
  • The present findings disclosed herein (including .g., diminished GM-CSF expression in Stat5−/− CD4+ T cells in vivo, IL-7/STAT5-mediated induction of GM-CSF expression in naïve CD4+ T cells, and the distinct features of GM-CSF-expressing TH cells versus T H1 and T H17 cells) indicates a distinct TH cell subset that is regulated by IL-7-STAT5 signaling. This finding was further explored by examining GM-CSF-expressing TH cell differentiation in vitro by activating naïve CD4+ T cells with anti-CD3 and anti-CD28 in the presence of different concentrations of IL-7. As shown in FIGS. 15A and 15B, IL-7 strongly promoted the generation of GM-CSF-expressing cells and GM-CSF secretion. Moreover, the generation of GM-CSF-expressing TH by IL-7 was mediated by STAT5. Without STAT5, IL-7 was unable to promote the generation of GM-CSF-expressing cells (FIGS. 15C and 15D). Further investigation showed that IL-7-induced STAT5 activation directly hound promoter regions of the Csf2 gene (FIGS. 15E and 15F).
  • Small proportions of IFN-γ-expressing cells were generated during GM-CSF-expressing TH differentiation (FIG. 1.5A). Thus, the effect of blocking IFN-γ on GM-CSF-expressing cell generation was tested, which showed that the combination of IL-7 and IFN-γ neutralization induced the highest frequency of GM-CSF+ cells, where few IL-17+ or IFN-γ+ cells were detected (FIG. 16A). Moreover, the expression of subset defining transcriptional factors in GM-CSF-expressing TH was examined and observed that the expression of RORγt or T-bet in GM-CSF-expressing TH was significantly lower than those in TH17 or T H1 cells, respectively (FIG. 16B), confirming that the GM-CSF-expressing TH cells are distinct from T H1 and T H17 cells. Together, these data suggest that IL-7-STAT5 signaling direct the differentiation of a novel GM-CSF-expressing helper T cell subset, termed TH-GM.
  • Next, it was determined whether IL-2 signaling could influence TH-GM differentiation from naïve CD4+ T cells. The addition of IL-2 or antibody against IL-2 only had modest effect on the frequency of GM-CSF+ cells (FIG. 14D), indicating a minimal effect of IL-2 on TH-GM differentiation. Unlike IL-7Rα, IL-2 high-affinity receptor IL-2Ra was not expressed in naïve CD4+ T cells, but its expression was gradually induced by TCR activation (FIGS. 17A-17C). Thus, the minimal effect of IL-2 at least in part is due to the unresponsiveness of naïve CD4+ T cells to IL-2 stimulation. In support of this view, IL-7, but not IL-2, induced STAT5 activation and upregulated GM-CSF mRNA expression in naïve CD4+ T cells (FIGS. 17D and 17E). To further confirm this idea, activated CD4+ T cells were stimulated with IL-2 or IL-7, which showed that both cytokines induced STAT5 activation, Csf2 promoter binding and GM-CSF mRNA upregulation (FIGS. 18A-18C). Notably, IL-2 induced a prolonged STAT5 activation compared with IL-7 (FIG. 18A).
  • Example 7. Distinct Gene Expression Profile of TH-GM
  • To demonstrate TH-GM as distinct from known T cell subsets (e.g., T H1 and TH17), a whole transcriptome analysis was performed by microarray to validate its specificity compared with known T cell subsets, in particular T H17 cells. Naïve CD4+ T cells were differentiated into T H1, T H17 and TH-GM. Microarray analysis was performed to examine their gene expression profiles. Whole transcriptome clustering indicates TH-GM cells as representing a novel subset distinct from T H1 or T H17 cells. T cell lineage-specific gene expression is shown in Table 1. A list of 202 genes preferentially expressed in T H1 cells were identified, compared with naïve, T H17 or TH-GM cells (fold change >1.7), among which IFN-γ and T-bet are on the top of the list (Table 1). Similarly, TH17-feature genes, such as IL-17, IL-17F, RORγt and RORα, were identified in the list including 411 genes specific to T H17 cells (Table 1). The TH-GM cell-specific gene list (“Genes preferentially upregulated in TH-GM” the TH-GM signature genes) contains 210 genes including the gene encoding GM-CSF as the top gene in the list (Table 1). A set of surface molecules which were selectively expressed at high level in TH-GM subset, and another set of surface molecules which were selectively expressed at low level in TH-GM subset compared with other subsets were identified (FIG. 19 and Table 1). These molecules (also TH-GM signature genes) can be used for further characterization by surface markers to identify the TH-GM subset of T cells. Several other genes of interest were also identified, including genes encoding cytokines and transcriptional factors, in particular IL-3. Various helper T cells were differentiated in vitro and confirmed that TH-GM cells are potent IL-3 producers as compared with T H1 and T H17 cells (FIGS. 20A, 20C and 20D). In addition, several other cytokines, including EBI-3, PENL and RANKL were found preferentially expressed in TH-GM cells (FIG. 20B), indicating diverse biological functions of TH-GM cells.
  • Example 8. TH-GM Cells are the Primary Pathogenic Population
  • To test the hypothesis that GM-CSF-expressing TH subset (TH-GM) was the primary encephalitogenic effector cells, adoptive transfer of different subsets of MOG35-55-specific CD4+ T cells was performed into Rag2−/− mice for EAE induction. As shown in FIG. 21, GM-CSF-expressing TH cells were preferentially able to induce EAE compared with T H17 and T H1 subsets.
  • Example 9. The Suppression of STAT5 Activity by Chemical Inhibitor Attenuates GM-CSF Expression by TH-GM and Ameliorates EAE
  • The effect of disrupting STAT5 activation by chemical inhibitor was examined to explore possible methods of treating autoimmune neuroinflammation. The phosphorylation on the key tyrosine residue in SH2 domain is crucial for STAT5 activation and function. A commercial STAT5 inhibitor (CAS 285986-31-4, Calbiochem) has been reported to selectively disrupt tyrosine phosphorylation and DNA binding of STAT5 (Muller et al., 2008). First, the inhibitory effect of this inhibitor on STAT5 activation upon IL-7 stimulation in CD4+ T cells was tested. At a concentration of 50 μM, the inhibitor had about 50% inhibitory effect, which was further enhanced with the increase of concentration (FIG. 22A). STAT5 inhibitor had low affinity and thus required a high concentration to fully block STAT5 activation, whereas JAK3 inhibitor showed potent inhibitory effect even at low concentration (FIG. 22B). The specificity of STAT5 inhibitor was next tested by examining its effect on the activation of STAT3 and STAT1. As shown in FIGS. 22C and 22D, this STAT5 inhibitor at relatively lower concentration (50 or 100 μM) showed minimal inhibitory effect on both STAT3 and STAT1 activation.
  • The effect of STAT5 inhibition on TH-GM differentiation was examined. As shown, STAT5 inhibitor suppressed TH-GM differentiation in a dosage-dependent manner (FIG. 22E). Reduced T H1 differentiation upon STAT5 inhibitor treatment was observed (data not shown), but T H17 differentiation was not suppressed by STAT5 inhibitor (data not shown).
  • To explore the therapeutic effect of targeting STAT5 activation in EAE disease, the commercial STAT5 inhibitor was administered to wild-type mice intraperitoneally every other clay after disease onset. Development of paralysis was assessed by daily assignment of clinical scores. STAT5 inhibition ameliorated EAE severity, associated with reduced immune cell infiltration in the CNS (FIGS. 23A and 23B). In contrast, although JAK3 inhibitor can potently block STAT5 activation (FIG. 22B), it showed detrimental effect on EAE (FIG. 23B). Of note, STAT5 inhibitor resulted in reduced GM-CSF production in CNS-infiltrating CD4+ T cells (FIGS. 23C and 23D). This study indicates that targeting STAT5 by chemical inhibitor is useful in therapeutic intervention in MS.
  • Example 10. GM-CSF-Producing TH Cells are Associated with Human RA
  • Plasma concentrations of GM-CSF and TNF-α in peripheral blood of RA patients were examined in comparison with gender/age-matched healthy control (HC), and found that both cytokines were elevated in RA (FIG. 24A). Ex vivo frequencies of IFN-γ-, IL-17- or GM-CSF-producing TH cells were quantified in RA and HC. High frequencies of IFN-γ- and/or GM-CSF-producing TH cells were detected in all samples, but observed low frequency (<1%) of IL-17-producing TH cells (FIG. 24B). GM-CSF-single-producing (GM-CSF+IFN-γ) TH cells represented a substantial population in both RA and HC (FIG. 24B). More importantly, the frequency of this population in peripheral blood of RA was significantly higher than that of HC (FIG. 24C). In contrast, neither GM-CSF/IFN-γ-double-producing nor IFN-γ-single-producing TH cells showed any significant difference in their frequencies between RA and HC (FIG. 24C). Therefore, the frequency of GM-CSF-single-producing TH cells in peripheral blood is selectively elevated in RA, suggesting a functional association of TH-cell-secreted GM-CSF with RA. Moreover, a significant correlation between plasma GM-CSF concentration and GM-CSF-single-producing TH cell frequency was observed in RA (FIG. 24D).
  • To further evaluate the association of GM-CSF-producing TH cells with RA, mononuclear cells were isolated from synovial fluid of RA patients and analyzed the abundance of these cells. A marked elevation of GM-CSF-producing TH cell frequency was observed in synovial fluid compared with peripheral blood, but most of these cells co-expressed IFN-γ (FIG. 24E). Similarly, both T H1 and T H17 frequencies were also increased in synovial fluid, with T H17 remaining to be a minor population compared with TH1 (FIG. 24E).
  • Example 11. GM-CSF Mediates Experimental Arthritis in a TNF-α-Independent Manner
  • The elevation of GM-CSF and TNF-α level in plasma of RA in comparison to HC may suggest a therapeutic approach by targeting these two cytokines. The efficacy of blocking both GM-CSF and TNF-α was tested in treating arthritic mice in antigen-induced arthritis (AIA) model, which is a T-cell driven RA model and is easily inducible in C57BL/6 strain with a rapid and synchronized disease onset, facilitating the exploration of RA pathogenesis. Either GM-CSF or TNF-α individual blockade attenuated MA development (FIG. 25A). Interestingly, the combination of GM-CSF- and TNF-α-specific neutralizing antibodies showed better efficacy in controlling arthritis development than individual treatments (FIG. 25A). That is, targeting GM-CSF may have beneficial efficacy in treating arthritis in a way independent of TNF-α activity. To further study the distinguishable effects of GM-CSF and TNF-α in mediating arthritis development, a mouse strain (Cd4-Cre; Stat5f/f, or Stat5−/− in short) with conditional Stat5 deletion was used in T cells for AIA induction. These conditional Stat5-knockout mice resisted arthritis development, as exemplified by milder joint selling, fewer immune cell infiltration in synovia, and reduced joint destruction (FIGS. 25B-25D), even though they had markedly increased level of serum TNF-α as well as IFN-γ (FIG. 25E). In contrast, serum level of GM-CSF was significantly reduced in knockout mice (FIG. 25E), which was likely the causal factor of the resistance to arthritis development as further supported by results described below. Consistent results were also observed in collagen-induced arthritis (CIA) model (FIGS. 26A-26D). Together, these findings suggest that GM-CSF is an important pathogenic mediator in RA and also indicate the promise of developing anti-GM-CSF drugs to treat RA patients who are anti-TNFα drugs unresponsive, marking GM-CSF-producing TH cells as a new biomarker for RA diagnosis.
  • Example 12. STAT5-Regulated GM-CSF Secretion by Autoreactive TH Cells Mediates Synovial Inflammation
  • On the basis of association of GM-CSF with RA, the cellular producers of GM-CSF and the regulatory mechanism underlying GM-CSF expression in arthritic mice were examined. Splenocytes were collected from wild-type AIA mice and separated cells into three fractions: splenocytes, splenocytes depleted of CD4+ T cells and CD4+ T cells; and stimulated each fraction at same cell numbers under various conditions. Splenocytes produced low but detectable level of GM-CSF without stimulation, which was markedly increased by PMA/Ionomycin or mBSA antigen stimulation (FIG. 28A). Under all conditions, splenocytes depleted of CD4+ T cells almost completely abrogated GM-CSF production (FIG. 28A). In contrast, CD4+ T cells produced dramatically elevated GM-CSF in comparison to splenocytes under all conditions (FIG. 28A). These results strongly support that CD4+ T cells are predominant producers of GM-CSF at least in spleens of arthritic mice, which is somehow consistent the observed correlation of plasma GM-CSF concentration with GM-CSF-single-producing TH cell frequency in RA (FIG. 24D). Thus, the functional significance of TH-cell-secreted GM-CSF was examined in arthritis development. Given T-cell-specific Csf2-knockout mice is not available and STAT5 is a key regulator of GM-CSF expression in TH cells, conditional Stat5-knockout mice was used, which showed decreased GM-CSF level and resistance to arthritis development as described above.
  • Consistent with a previous study (Burchill et al., 2007), similar frequencies of CD4+ T cells were observed in peripheral lymphoid tissues as well as in inflamed synovial tissues of STAT5-deficient mice compared with wild-type mice at day 7 after AIA induction (FIGS. 27A-27D), suggesting loss of STAT5 seems to not impair CD4+ T-cell generation in periphery and infiltration in synovial tissues. To determine the requirement of STAT5 for arthritogenic potential of CD4+ T cells, ex vivo-expanded antigen-reactive CD4+ T cells, derived from Stat5+/+ and Stat5−/− ALA mice, were transferred into wild-type naïve mice separately, followed by intra-articular injection of mBSA. Mice receiving Stat5+/+ CD4+ T cells displayed an abundant immune cell infiltration in synovial tissues at day 7 after AIA induction (FIG. 27E). In contrast, mice receiving Stat5−/− CD4+ T cells had marked reduction of synovial infiltrates (FIG. 27E). Therefore, STAT5-deficient CD4+ T cells are defective in arthritogenic potential.
  • Multiple lines of evidence support a central role of T cells in RA. However, the pathogenic mechanism of T cells remains insufficiently understood. Although T H1 is a predominant population among synovial infiltrating CD4+ T cells in human RA (Berner et al., 2000; Yamada et al., 2008), defective IFN-γ signaling results in increased disease susceptibility in animal models of arthritis (Guedez et al., 2001; Irmler et al., 2007; Manoury-Schwartz et al., 1997; Vermeire et al., 1997). In contrast, T H17 cells are proven crucial in animal models of arthritis (Pernis, 2009), but predominance of T H17 cells is limited in both peripheral blood and synovial compartment of human RA (Yamada et al., 2008) and (FIGS. 1B and 1E). As demonstrated herein, STAT5-regulated GM-CSF-producing TH cells potentiate arthritis pathogenesis.
  • To validate the regulatory role of STAT5 in GM-CSF production, splenocytes derived from AIA mice were stimulated with PMA/Ionomycin plus Golgiplug ex viva, followed by intracellular cytokine staining and flow cytometry. As expected, the frequency of GM-CSF-single-producing cells among CD4+CD44hi population was significantly decreased in Stat5−/− mice (FIG. 28 34B). Notably, no significant differences were observed in frequencies of IL-17-single-producing (TH17) or IFN-γ-single-producing (TH1) cells between two groups (FIG. 28B). Further study by combining mBSA restimulation and intracellular cytokine staining showed that the frequency of mBSA-specific GM-CSF-producing effector T cells was much lower in spleens of Stat5−/− mice than those in controls (FIG. 29A). In addition, AIA mice-derived splenocytes and inguinal lymph nodes (LNs) were restimulated with mBSA ex viva to measure cytokine concentrations in culture supernatants and found a significant reduction of GM-CSF with deletion of STAT5, but comparable levels of both IL-17 and IFN-γ between two groups (FIGS. 29B and 29C). Together, the results indicate that loss of STAT5 may specifically suppress GM-CSF-producing effector Th cells but not T H17 or T H1 cells in experimental arthritis.
  • To investigate the involvement of GM-CSF-producing TH cells and their regulation by STAT5 in synovial inflammation, synovial tissues were dissected from AIA mice and examined cytokine production by TH cells. In spite of multiple cellular sources of GM-CSF (Cornish et al., 2009). CD4+ TH cells were prominent producers of GM-CSF in synovial tissues of AIA mice (FIG. 28C), consistent with the observation in spleens (FIG. 28A). Moreover, a significantly lower percentage of synovial GM-CSF-producing TH cells was detected in Stat5−/− mice than Stat5+/+ mice (FIG. 28D). On the other hand, both T H1 and T H17 cells exhibited similar percentages between two groups (FIG. 28C). A decrease in GM-CSF level in synovial compartments of Stat5−/− mice in comparison to controls was expected. To address this, inflamed synovial tissues were harvested from AIA mice for RNA and protein extraction to examine cytokine level by qPCR and ELISA. Indeed, lower synovial GM-CSF but not IFN-γ or IL-17 was detected in Stat5−/− mice than Stat5+/+ mice at day 5 or 7 after arthritis induction (FIGS. 28E and 30A-30C). In addition, two important proinflammatory cytokines IL-6 and IL-1β were also found persistently and significantly reduced in STAT5-deficient mice (FIGS. 28E and 30A-30C), indicating the attenuated synovial inflammation. Notably. TNF-α production was reduced at day 7 but not at day 5 in STAT5-deficient mice (FIGS. 28E and 30A-C). Together, these results indicate that STAT5-regulated GM-CSF expression by arthritogenic TH cells is crucial for evoking synovial inflammation.
  • To determine the critical role of STAT5-regulated GM-CSF production by TH cells in mediating synovitis and arthritis development, GM-CSF was administered via intra-articular injection in mixture with mBSA to the left knee joints of mBSA/CFA-immunized mice, whereas mBSA was injected alone to the right knee joints. Injection with mBSA alone was sufficient to induce abundant immune cell infiltration in the synovial compartments of Stat5+/+ mice but failed to do so in Stat5−/− mice (FIG. 28F). Administration of GM-CSF together with mBSA efficiently restored synovial inflammation in Stat5−/− mice (FIG. 34F). Consistently, the Safranin-O/Fast Green staining revealed severe cartilage depletion upon GM-CSF/mBSA injection, but not mBSA alone, in Stat5−/− mice (FIG. 28G). These results therefore provide support for the notion that STAT5-regulated GM-CSF production by arthritogenic TH cells is essential for mediating arthritis pathogenesis.
  • Example 13. Th-Cell-Derived GM-CSF Mediates Neutrophil Accumulation in Synovial Tissues
  • The mechanism by which GM-CSF-producing Th cells evoke synovial inflammation and drive arthritis development was examined. Myeloid lineage-derived cells, including neutrophils, DCs and macrophages, express GM-CSF receptor and are common targets of GM-CSF (Hamilton, 2008). Importantly, those cells invade synovial compartments in RA patients and mouse arthritis models, and contribute to synovitis (McInnes and Schett, 2011). The infiltration of myeloid lineage-derived cells in synovial compartments of ALA mice was examined. CD11b+ myeloid cells represented a predominant population (˜70%) among synovial infiltrating leukocytes (FIG. 31B). Although CD4+ TH cell infiltration was not altered by STAT5 deletion, synovial CD11b+ cell infiltration was significantly reduced in Stat5−/− mice compared with Stat5+/+ mice when examined at day 7 after arthritis induction (FIG. 31B). This reduction is unlikely due to defective hematopoiesis, as similar frequencies of CD11b+ cells were detected in spleens of two group (FIG. 31A). Further, CD11b+ cells continuously increased in synovial tissues of wild-type mice, but not STAT5-deficient mice, over a 7-day time course (FIG. 31C). Notably, the selective ablation of synovial CD11b+ cell accumulation in STAT5-deficient mice can be partially restored by local administration of GM-CSF during arthritis induction (FIG. 31D). Together, these results indicate that myeloid cell accumulation in synovial compartments may be specifically dampened by T-cell-specific STAT5 deletion and resultant GM-CSF insufficiency.
  • Next, different populations of CD11b+ cells, including DCs, macrophages and neutrophils were analyzed. Monocyte-derived dendritic cells (MoDCs), characterized as CD11cintCD11bhiLy6C+/hiMHCIIhi, were recently reported to be involved in the mBSA/IL-1β arthritis model (Campbell et al., 2011). In the AIA model of the present study, MoDCs were identified at low abundance in spleens and synovial tissues (data not shown). Furthermore, comparable frequencies of MoDCs were detected in both peripheral lymphoid tissues and synovial tissues between Stat5+/+ and Stat5−/− mice (data not shown). These results are in agreement with a previous study showing a dispensable role of GM-CSF in MoDC differentiation (Greter et al., 2012).
  • Neutrophils have great cytotoxic potential and contribute to the RA initiation and progression in multiple ways (Wright et al., 2014). It has been suggested that RA disease activity and joint destruction directly correlates with neutrophil influx to joints (Wright et al., 2014). Based on the differential expression of Ly6C and Ly6G, CD11b+ myeloid cells can be classified into Ly6CloLy6Ghi population (neutrophils) and Ly6ChiLy6G population (monocytes/macrophages). The present study shows that Ly6CloLy6Ghi population continued to accumulate in synovial tissues over a 7-clay time course, and represented a predominant population among synovial CD11b+ cells in wild-type mice at day 7 after AIA induction, whereas this population was persistently and dramatically diminished in STAT5-deficient mice (FIG. 32A). Using Giemsa stain, it was validated that synovial-infiltrating Ly6CloLy6Ghi population were neutrophils, which displayed typical polymorphonuclear characteristics with ring-shaped nuclei (FIG. 32B). In contrast, synovial-infiltrating Ly6ChiLy6G population had mononuclear morphology and were likely monocytes/macrophages (FIG. 32B). Importantly, intraarticular administration of GM-CSF during arthritis induction efficiently restored neutrophil accumulation in synovial compartments of STAT5-deficient mice (FIG. 32C), suggesting a critical role of TH-cell-derived GM-CSF in mediating neutrophil accumulation to inflamed joints.
  • Neutrophils are recruited during inflammation, in which complex interactions between neutrophils and vascular endothelial cells direct neutrophil adhesion and transmigration from circulation to inflamed tissues (Kolaczkowska and Kubes, 2013). In an in vitro transmigration assay, neutrophil adhesion and migration across monolayers of endothelial cells was significantly enhanced by GM-CSF as chemoattractant (FIGS. 33A and 33B), suggesting GM-CSF may mediate neutrophil recruitment to inflamed joints in AIA. Effective neutrophil apoptosis is crucial for the resolution of inflammation. However, in synovitis, neutrophil apoptosis is delayed with a result of extended survival and persistent inflammation (Wright et al., 2014). Thus, the effect of GM-CSF on neutrophil survival was tested and found that GM-CSF had profound efficacy in delaying neutrophil apoptosis (FIG. 33C). Together, these results indicate that GM-CSF may mediate neutrophil recruitment and sustain neutrophil survival in synovial compartments and contribute to persistent synovitis. To determine the critical role of neutrophils in AIA, \ a neutralizing antibody (1A8) specific for Ly6G was used to deplete neutrophils in vivo. The administration of neutralizing antibody resulted in significant improvement of joint swelling in AIA (FIG. 32D). Thus, neutrophils accumulation mediated by TH-cell-derived GM-CSF are important for AIA development.
  • Example 14. GM-CSF Enhances Proinflammatory Cytokine Production by Myeloid Cells and Synovial Fibroblasts
  • Cytokines are important mediators in the cross-talk between innate and adaptive immunity. As shown herein, several proinflammatory cytokines (IL-6, IL-1β and TNF-α), which are in association with RA pathogenesis (Choy and Panayi, 2001), were significantly reduced in synovial tissues of STAT5-deficient AIA mice (FIGS. 28E and 30A-30C). To gain insights into the mechanism underlying the observed cytokine reduction, the cellular sources of these proinflammatory cytokines were examined by fractionating synovial cells into different populations based the differential expression of surface markers (FIG. 34A). Cytokine mRNA expression level in CD45+TCRβ+ population (T cells), CD45+TCRβCD11c CD11b+ population (mostly monocytes/macrophages and neutrophils) and CD45+TCRβ CD11c+ population (dendritic cells) was assessed by RT-PCR. GM-CSF, as similar to IL-17 (as a control), was predominantly produced by synovial T cells (FIG. 34B), further reinforcing the importance of GM-CSF-producing TH cells. In contrast, IL-6 and IL-1β were mainly produced by myeloid cells, e.g. CD11b+ population and CD11c+ population (FIG. 34B). TNF-α was expressed by all three populations, with relatively lower abundance in T cells (FIG. 34B). Based on the differential expression of Ly6C and Ly6G in CD11b+ population as discussed above, Ly6CloLy6Ghi population (neutrophils) and Ly6ChiLy6G population (monocytes/macrophages) were further analyzed, which showed that monocytes/macrophages were likely the major IL-6 producers whereas neutrophils seemed to be better producers of IL-1β and TNF-α (FIG. 34C). These results, together with the findings above (FIGS. 28E and 30A-30C), indicate a link that TH-cell-secreted GM-CSF elicits proinflammatory cytokine expression from myeloid cells in synovitis.
  • To test the regulatory role of GM-CSF in the expression of IL-6 and IL-1β, bone marrow-derived macrophages (BMDMs) and bone marrow-derived dendritic cells (BMDCs) were cultured, and stimulated with GM-CSF. Indeed, GM-CSF stimulation quickly upregulated mRNA expression of both IL-6 and IL-1β within 1 hour (FIGS. 34D and 34E). In addition, GM-CSF markedly increased the secretion of IL-6 from BMDMs in a dosage-dependent manner (FIG. 34F), and from BMDCs even at low dosage (FIG. 34G). To induce mature IL-1β secretion, BMDMs were primed with LPS for 6 h during which different concentrations of GM-CSF was added, followed by ATP stimulation. The addition of GM-CSF significantly enhanced the secretion of IL-1β into culture supernatant as measured by ELISA (FIG. 34H). Synovial fibroblasts, the active players in synovial inflammation (Muller-Ladner et al., 2007), also showed increased IL-1β mRNA expression upon GM-CSF stimulation (FIG. 34I). An inducible effect of GM-CSF on TNF-α expression was not observed in BMDMs, BMDCs or synovial fibroblasts (data not shown). Given the functional importance of IL-6 and IL-1β in arthritis development (Choy and Panayi, 2001), TH-cell-secreted GM-CSF may mediate synovial inflammation also via eliciting the expression of IL- and IL-1β from myeloid cells and synovial fibroblasts.
  • TABLE 1
    Summary of genes differentially expressed in TH1, TH17, and TH-GM cells
    Genes differentially Genes differentially
    expressed in TH1 expressed in TH17
    Gene Gene
    ID Gene Title ID Gene Title
    10366586 interferon gamma 10353415 interleukin 17F
    10598013 chemokine (C-C 10511779 ATPase, H+
    motif) receptor 5 /// transporting,
    chemokine (C-C lysosomal V0
    motif) receptor 2 subunit D2
    10523717 secreted 10345762 interleukin 1
    phosphoprotein 1 receptor, type 1
    10420308 granzyme B 10359697 chemokine (C
    motif) ligand 1
    10545135 interleukin 12 10587639 5′ nucleotidase,
    receptor, beta 2 ecto
    10531724 placenta-specific 8 10501860 formin binding
    protein 1-like
    10363070 glycoprotein 49 A /// 10345032 interleukin 17A
    leukocyte
    immunoglobulin-
    like receptor,
    subfamily B,
    member 4
    10363082 leukocyte 10446965 RAS, guanyl
    immunoglobulin- releasing protein 3
    like receptor,
    subfamily B,
    member 4
    10424683 lymphocyte antigen 10565990 ADP-
    6 complex, locus G ribosyltransferase
    2a
    10552406 natural killer cell 10465059 cathepsin W
    group 7 sequence
    10603151 glycoprotein m6b 10358476 proteoglycan 4
    (megakaryocyte
    stimulating factor,
    articular
    superficial zone
    protein)
    10360173 SLAM family 10471953 activin receptor
    member 7 IIA
    10455961 interferon inducible 10400006 aryl-hydrocarbon
    GTPase 1 receptor
    10400304 EGL nine homolog 10409876 cytotoxic T
    3 (C. elegans) lymphocyte-
    associated protein
    2 alpha
    10574023 metallothionein 2 10388591 carboxypeptidase
    D
    10493108 cellular retinoic 10390640 IKAROS family
    acid binding protein zinc finger 3
    II
    10375436 family with 10590623 chemokine (C-X-
    sequence similarity C motif) receptor
    71, member B 6
    10398039 serine (or cysteine) 10367734 uronyl-2-
    peptidase inhibitor, sulfotransferase
    clade A, member 3F ///
    serine (or
    cysteine) peptidase
    inhibitor, clade A,
    member 3G
    10349108 serine (or cysteine) 10500656 CD101 antigen
    peptidase inhibitor,
    clade B, member 5
    10607738 carbonic anhydrase 10347895 WD repeat
    5b, mitochondrial domain 69
    10496539 guanylate binding 10495854 protease, serine,
    protein 5 12 neurotrypsin
    (motopsin)
    10373918 leukemia inhibitory 10425049 apolipoprotein
    factor L9b ///
    apolipoprotein
    L9a
    10455954 predicted gene 4951 10378286 integrin alpha E,
    epithelial-
    associated
    10598976 tissue inhibitor of 10362896 CD24a antigen
    metalloproteinase 1
    10492136 doublecortin-like 10409866 cytotoxic T
    kinase 1 lymphocyte-
    associated protein
    2 beta
    10405211 growth arrest and 10400989 potassium
    DNA-damage- voltage-gated
    inducible 45 gamma channel,
    subfamily H (eag-
    related), member
    5
    10503202 chromodomain 10590242 chemokine (C-C
    helicase DNA motif) receptor 8
    binding protein 7
    10542275 ets variant gene 6 10407435 aldo-keto
    (TEL oncogene) reductase family
    1, member Cl8
    10556820 transmembrane 10592023 amyloid beta
    protein 159 (A4) precursor-
    like protein 2
    10444291 histocompatibility 10359480 dynamin 3
    2, class II antigen A,
    beta 1
    10439299 stefin A3 10475544 sema domain,
    transmembrane
    domain (TM), and
    cytoplasmic
    domain,
    (semaphorin) 6D
    10547641 solute carrier family 10409767 golgi membrane
    2 (facilitated glucose protein 1
    transporter),
    member 3
    10503200 chromodomain 10392464 family with
    helicase DNA sequence
    binding protein 7 similarity 20,
    member A
    10544320 RIKEN cDNA 10504891 transmembrane
    1810009J06 gene /// protein with EGF-
    predicted gene 2663 like and two
    follistatin-like
    domains 1
    10503218 chromodomain 10504817 transforming
    helicase DNA growth factor,
    binding protein 7 beta receptor 1
    10503198 chromodomain 10393559 tissue inhibitor of
    helicase DNA metalloproteinase
    binding protein 7 2
    10507594 solute earner family 10474419 leucine-rich
    2 (facilitated glucose repeat-containing
    transporter), G protein-coupled
    member 1 receptor 4
    10438626 ets variant gene 5 10456492 DNA segment,
    Chr 18, ERATO
    Doi 653,
    expressed
    10390328 T-box 21 10345241 dystonin
    10574027 metallothionein 1 10471555 angiopoietin-like
    2
    10493820 S100 calcium 10494821 tetraspanin 2
    binding protein A6
    (calcyclin)
    10376324 predicted gene 10542355 epithelial
    12250 membrane protein
    1
    10406852 calponin 3, acidic 10500295 pleckstrin
    homology domain
    containing, family
    O member 1
    10412076 gem (nuclear 10375402 a disintegrin and
    organelle) metallopeptidase
    associated protein 8 domain 19
    (meltrin beta)
    10496555 guanylate binding 10484227 SEC 14 and
    protein 1 /// spectrin domains
    guanylate binding 1
    protein 5
    10345074 centrin 4 10472097 formin-like 2
    10503194 chromodomain 10587829 procollagen
    helicase DNA lysine, 2-
    binding protein 7 oxoglutarate 5-
    dioxygenase 2
    10537561 RIKEN cDNA 10530536 tec protein
    1810009J06 gene /// tyrosine kinase
    predicted gene 2663
    10439895 activated leukocyte 10586700 RAR-related
    cell adhesion orphan receptor
    molecule alpha
    10459772 lipase, endothelial 10354191 ring finger
    protein 149
    10439762 S- 10438738 B-cell
    adenosylhomocysteine leukemia/lymphoma
    hydrolase 6
    10482030 stomatin 10347888 chemokine (C-C
    motif) ligand 20
    10459905 SET binding 10440131 G protein-
    protein 1 coupled receptor
    15
    10357833 ATPase, Ca++ 10453057 cytochrome P450,
    transporting, plasma family 1,
    membrane 4 subfamily b,
    polypeptide 1 ///
    RIKEN cDNA
    1700038P13 gene
    10475517 expressed sequence 10542140 killer cell lectin-
    AA467197 /// like receptor
    microRNA 147 subfamily B
    member 1F
    10585778 sema domain, 10471880 microRNA 181b-2
    immunoglobulin
    domain (Ig), and
    GPI membrane
    anchor,
    (semaphorin) 7A
    10354529 RIKEN cDNA 10542791 PTPRF
    1700019D03 gene interacting
    protein, binding
    protein 1 (liprin
    beta 1)
    10582275 solute carrier family 10583242 sestrin 3
    7 (cationic amino
    acid transporter, y+
    system), member 5
    10576034 interferon 10489569 phospholipid
    regulatory factor 8 transfer protein ///
    cathepsin A
    10503222 chromodomain 10523297 cyclin G2
    helicase DNA
    binding protein 7
    10503220 chromodomain 10381187 ATPase, H+
    helicase DNA transporting,
    binding protein 7 lysosomal V0
    subunit A1
    10503210 chromodomain 10346651 bone
    helicase DNA morphogenic
    binding protein 7 protein receptor,
    type II
    (serine/threonine
    kinase)
    10476945 cystatin F 10490159 prostate
    (leukocystatin) transmembrane
    protein, androgen
    induced 1
    10503216 chromodomain 10389581 yippee-like 2
    helicase DNA (Drosophila)
    binding protein 7
    10366983 transmembrane 10581992 avian
    protein 194 musculoaponeurotic
    fibrosarcoma
    (v-maf) AS42
    oncogene
    homolog
    10495675 coagulation factor 10413250 cytoplasmic
    III polyadenylated
    homeobox
    10421697 RIKEN cDNA 10555063 integrator
    9030625A04 gene complex subunit 4
    10445112 ubiquitin D 10406982 a disintegrin-like
    and
    metallopeptidase
    (reprolysin type)
    with
    thrombospondin
    type 1 motif, 6
    10530627 leucine rich repeat 10596303 acid phosphatase,
    containing 66 prostate
    10440019 transmembrane 10357472 chemokine (C-X-
    protein 45a C motif) receptor
    4
    10378783 ribosomal protein 10545130 growth arrest and
    L36 DNA-damage-
    inducible 45 alpha
    10447341 ras homolog gene 10436402 claudin domain
    family, member Q /// containing 1
    phosphatidylinositol
    glycan anchor
    biosynthesis, class F
    10373452 predicted gene 129 10539135 capping protein
    (actin filament),
    gelsolin-like
    10454286 microtubule- 10428534 trichorhinophalangeal
    associated protein, syndrome 1
    RP/EB family, (human)
    member 2
    10572497 interleukin 12 10368675 myristoylated
    receptor, beta 1 alanine rich
    protein kinase C
    substrate
    10368060 epithelial cell 10531910 hydroxysteroid
    transforming (17-beta)
    sequence 2 dehydrogenase 13
    oncogene-like
    10471457 ST6 (alpha-N- 10370303 adenosine
    acetyl-neuraminyl- deaminase, RNA-
    2,3-beta-galactosyl- specific, B1
    1,3)-N-
    acetylgalactosaminide
    alpha-2,6-
    sialyltransferase 4
    10374366 epidermal growth 10592888 chemochine (C-
    factor receptor X-C motif)
    receptor 5
    10450501 tumor necrosis 10503259 transformation
    factor related protein 53
    inducible nuclear
    protein 1
    10347291 chemokine (C-X-C 10446771 lysocardiolipin
    motif) receptor 2 acyltransferase 1
    10553501 solute carrier family 10428579 exostoses
    17 (sodium- (multiple) 1
    dependent inorganic
    phosphate
    cotransporter),
    member 6
    10345824 interleukin 18 10476314 prion protein
    receptor accessory
    protein
    10458314 transmembrane 10406598 serine
    protein 173 incorporator 5
    10388430 serine (or cysteine) 10461765 leupaxin
    peptidase inhibitor,
    clade F, member 1
    10496015 phospholipase A2, 10428536 trichorhinophalangeal
    group XIIA syndrome 1
    (human)
    10510391 spermidine synthase 10362245 erythrocyte
    protein band 4.1-
    like 2
    10486396 EH-domain 10604008 predicted gene
    containing 4 10058 ///
    predicted gene
    10230 ///
    predicted gene
    10486 ///
    predicted gene
    14632 ///
    predicted gene
    14819 ///
    predicted gene
    4836 ///
    predicted gene
    2012 ///
    predicted gene
    5169 ///
    predicted gene
    6121 /// Sycp3
    like X-linked ///
    predicted gene
    5168 ///
    predicted gene
    10488 ///
    predicted gene
    14525 ///
    predicted gene
    5935
    10368054 epithelial cell 10409857 RIKEN cDNA
    transforming 4930486L24 gene
    sequence 2
    oncogene-like
    10608637 na 10522368 NIPA-like
    domain containing
    1
    10595718 carbohydrate 10368720 solute carrier
    sulfotransferase 2 family 16
    (monocarboxylic
    acid transporters),
    member 10
    10496580 guanylate binding 10438639 diacylglycerol
    protein 3 kinase, gamma
    10594053 promyelocytic 10499431 synaptotagmin XI
    leukemia
    10544829 JAZF zinc finger 1 10565840 neuraminidase 3
    10601778 armadillo repeat 10494023 RAR-related
    containing, X-linked orphan receptor
    3 gamma
    10355967 adaptor-related 10391103 junction
    protein complex AP- plakoglobin
    1, sigma 3
    10592503 cytotoxic and 10417053 muscleblind-like
    regulatory T cell 2
    molecule
    10496023 caspase 6 10350341 microRNA 181b-1
    10599192 LON peptidase N- 10459071 RIKEN cDNA
    terminal domain and 2010002N04 gene
    ring finger 3
    10467578 phosphoinositide-3- 10463476 Kazal-type serine
    kinase adaptor peptidase inhibitor
    protein 1 domain 1
    10585703 ribonuclease P 25 10348537 receptor
    subunit (human) (calcitonin)
    activity modifying
    protein 1
    10365482 tissue inhibitor of 10348432 ArfGAP with
    metalloproteinase 3 GTPase domain,
    ankyrin repeat and
    PH domain 1
    10469151 inter-alpha 10576332 tubulin, beta 3 ///
    (globulin) inhibitor melanocortin 1
    H5 receptor
    10503192 chromodomain 10554094 insulin-like
    helicase DNA growth factor 1
    binding protein 7 receptor
    10593050 interleukin 10 10495794 phosphodiesterase
    receptor, alpha 5A, cGMP-
    specific
    10597648 myeloid 10569504 tumor necrosis
    differentiation factor receptor
    primary response superfamily,
    gene 88 member 23
    10538290 sorting nexin 10 10452516 ankyrin repeat
    domain 12
    10503204 chromodomain 10534596 cut-like
    helicase DNA homeobox 1
    binding protein 7
    10353707 protein tyrosine 10362073 serum/glucocorticoid
    phosphatase 4a1 /// regulated
    protein tyrosine kinase 1
    phosphatase 4a1-
    like
    10377010 SCO cytochrome 10408331 acyl-CoA
    oxidase deficient thioesterase 13
    homolog 1 (yeast)
    10440903 RIKEN cDNA 10415413 NYN domain and
    4932438H23 gene retroviral
    integrase
    containing
    10521205 SH3-domain 10598359 synaptophysin
    binding protein 2
    10604587 microRNA 363 10544114 homeodomain
    interacting protein
    kinase 2
    10571958 SH3 domain 10436128 myosin, heavy-
    containing ring chain 15
    finger 1
    10357553 interleukin 24 10408450 SRY-box
    containing gene 4
    10606730 armadillo repeat 10487011 glycine
    containing, X-linked amidinotransferase
    6 (L- arginine: glycine
    amidinotransferase)
    10564960 furin (paired basic 10378833 slingshot
    amino acid cleaving homolog 2
    enzyme) (Drosophila)
    10402585 tryptophanyl-tRNA 10521498 collapsin
    synthetase response mediator
    protein 1
    10417095 FERM, RhoGEF 10538939 eukaryotic
    (Arhgef) and translation
    pleckstrin domain initiation factor 2
    protein 1 alpha kinase 3
    (chondrocyte-
    derived)
    10442435 ribonucleic acid 10585276 POU domain,
    binding protein S1 class 2,
    associating factor
    1
    10394990 membrane bound 10512156 aquaporin 3
    O-acyltransferase
    domain containing 2
    10538753 atonal homolog 1 10469110 USP6 N-terminal
    (Drosophila) like
    10351667 signaling 10568392 regulator of G-
    lymphocytic protein signalling
    activation molecule 10
    family member 1
    10461844 guanine nucleotide 10603346 proteolipid
    binding protein, protein 2
    alpha q polypeptide
    10422057 ribosomal protein 10353947 transmembrane
    L7A protein 131
    10572897 heme oxygenase 10452633 TGFB-induced
    (decycling) 1 factor homeobox
    1
    10507784 palmitoyl-protein 10380289 monocyte to
    thioesterase 1 macrophage
    differentiation-
    associated
    10445702 ubiquitin specific 10521969 IMP1 inner
    peptidase 49 mitochondrial
    membrane
    peptidase-like
    (S. cerevisiae)
    10569057 ribonuclease/ 10521678 CD38 antigen
    angiogenin inhibitor 1
    10370471 1-acylglycerol-3- 10592515 ubiquitin
    phosphate O- associated and
    acyltransferase 3 SH3 domain
    containing, B
    10586591 carbonic 10512470 CD72 antigen
    anyhydrase 12
    10512701 translocase of outer 10587085 cDNA sequence
    mitochondrial BC031353
    membrane 5
    homolog (yeast)
    10462702 HECT domain 10492689 platelet-derived
    containing 2 growth factor, C
    polypeptide
    10552740 nucleoporin 62 /// 10514221 perilipin 2
    Nup62-Il4i1 protein
    10581996 chromodomain 10458247 leucine rich
    protein, Y repeat
    chromosome-like 2 transmembrane
    neuronal 2
    10363901 ets variant gene 5 10468898 lymphocyte
    transmembrane
    adaptor 1
    10520862 fos-like antigen 2 10555059 potassium
    channel
    tetramerisation
    domain containing
    14
    10526520 procollagen-lysine, 10408629 RIKEN cDNA
    2-oxoglutarate 5- 1300014106 gene
    dioxygenase 3
    10571274 glutathione 10546510 leucine-rich
    reductase repeats and
    immunoglobulin-
    like domains 1
    10351206 selectin, platelet 10544596 transmembrane
    protein 176B
    10493474 mucin 1, 10361748 F-box protein 30
    transmembrane
    10370000 glutathione S- 10356291 RIKEN cDNA
    transferase, theta 1 A530040E14 gene
    10500272 predicted gene 129 10581450 DEAD (Asp-Glu-
    Ala-Asp) box
    polypeptide 28
    10452815 xanthine 10414417 pellino 2
    dehydrogenase
    10393823 prolyl 4- 10372528 potassium large
    hydroxylase, beta conductance
    polypeptide calcium-activated
    channel,
    subfamily M, beta
    member 4 ///
    RIKEN cDNA
    1700058G18 gene
    10408280 leucine rich repeat 10408613 tubulin, beta 2B
    containing 16A
    10575685 nudix (nucleoside 10411274 synaptic vesicle
    diphosphate linked glycoprotein 2c
    moiety X)-type
    motif 7
    10599174 interleukin 13 10456357 phorbol-12-
    receptor, alpha 1 myristate-13-
    acetate-induced
    protein 1
    10458940 zinc finger protein 10511498 pleckstrin
    608 homology domain
    containing, family
    F (with FYVE
    domain) member
    2
    10476197 inosine 10402136 G protein-
    triphosphatase coupled receptor
    (nucleoside 68
    triphosphate
    pyrophosphatase)
    10419790 ajuba 10549990 vomeronasal 1
    receptor, G10 ///
    vomernasal 1
    receptor Vmn1r-
    ps4 ///
    vomeronasal 1
    receptor 3 ///
    vomeronasal 1
    receptor
    Vmn1r238 ///
    vomeronasal 1
    receptor 2
    10364909 ornithine 10554789 cathepsin C
    decarboxylase
    antizyme 1 ///
    ornithine
    decarboxylase
    antizyme 1
    pseudogene
    10503190 chromodomain 10427928 triple functional
    helicase DNA domain (PTPRF
    binding protein 7 interacting)
    10516932 sestrin 2 10549162 ST8 alpha N
    acetyl-
    neuraminide
    alpha-2,8-
    sialyltransferase 1
    10585338 KDEL (Lys-Asp- 10482109 mitochondrial
    Glu-Leu) containing ribosome
    2 recycling factor ///
    RNA binding
    motif protein 18
    10464425 G protein-coupled 10425092 cytohesin 4
    receptor kinase 5
    10441601 T-cell activation 10356866 programmed cell
    Rho GTPase- death 1
    activating protein
    10482059 glycoprotein 10554204 ATP/GTP
    galactosyltransferase binding protein-
    alpha 1,3 like 1
    10522411 cell wall biogenesis 10403229 integrin beta 8
    43 C-terminal
    homolog
    (S. cerevisiae)
    10369276 coiled-coil domain 10374529 expressed
    containing 109A sequence
    AV249152
    10368970 PR domain 10565434 ribosomal protein
    containing 1, with S13
    ZNF domain
    10369541 hexokinase 1 10431266 ceramide kinase
    10374236 uridine 10410124 cathepsin L
    phosphorylase 1
    10489660 engulfment and cell 10441003 runt related
    motility 2, ced-12 transcription
    homolog factor 1
    (C. elegans)
    10488797 peroxisomal 10555303 phosphoglucomutase
    membrane protein 4 2-like 1
    10558090 transforming, acidic 10530215 RIKEN cDNA
    coiled-coil 1110003E01 gene
    containing protein 2
    10409265 AU RNA binding 10480275 nebulette
    protein/enoyl-
    coenzyme A
    hydratase
    10374364 thymoma viral 10434302 kelch-like 24
    proto-oncogene 2 (Drosophila)
    10598575 LanC (antibiotic 10565002 CREB regulated
    synthetase transcription
    component C-like 3 coactivator 3
    (bacterial)
    10439514 growth associated 10413338 na
    protein 43
    10497842 Bardet-Biedl 10523670 AF4/FMR2
    syndrome 7 (human) family, member 1
    10462091 Kruppel-like factor 10478594 cathepsin A
    9 /// predicted gene
    9971
    10498024 solute carrier family 10514128 tetratricopeptide
    7 (cationic amino repeat domain
    acid transporter, y+ 39B
    system), member 11
    10483719 chimerin 10535956 StAR-related
    (chimaerin) 1 lipid transfer
    (START) domain
    containing 13
    10606694 Bruton 10503695 BTB and CNC
    agammaglobulinemia homology 2
    tyrosine kinase
    10443110 synaptic Ras 10584334 sialic acid
    GTPase activating acetylesterase
    protein 1 homolog
    (rat)
    10368062 epithelial cell 10502890 ST6 (alpha-N-
    transforming acetyl-
    sequence 2 neuraminyl-2,3-
    oncogene-like beta-galactosyl-
    1,3)-N-
    acetylgalactosaminide
    alpha-2,6-
    sialyltransferase 3
    10575693 vesicle amine 10564467 leucine rich
    transport protein 1 repeat containing
    homolog-like 28
    (T. californica)
    10562897 zinc finger protein 10345715 mitogen-activated
    473 /// vaccinia protein kinase
    related kinase 3 kinase kinase
    kinase 4
    10373709 eukaryotic 10568668 a disintegrin and
    translation initiation metallopeptidase
    factor 4E nuclear domain 12
    import factor 1 (meltrin alpha)
    10487238 histidine 10462406 RIKEN cDNA
    decarboxylase C030046E11 gene
    10594988 mitogen-activated 10472649 myosin IIIB
    protein kinase 6
    10422436 dedicator of 10363894 inositol
    cytokinesis 9 polyphosphate
    multikinase
    10459084 synaptopodin 10606058 chemokine (C-X-
    C motif) receptor
    3
    10567450 dynein, axonemal, 10439955 family with
    heavy chain 3 sequence
    similarity 55,
    member C
    10604751 fibroblast growth 10530615 OC1A domain
    factor 13 containing 2
    10584827 myelin protein 10528183 spermatogenesis
    zero-like 2 associated
    glutamate (E)-rich
    protein 4d ///
    spermatogenesis
    associated
    glutamate (E)-rich
    protein 4c ///
    spermatogenesis
    associated
    glutamate (E)-rich
    protein 4e ///
    predicted gene
    9758 /// RIKEN
    cDNA
    4930572O03 gene ///
    spermatogenesis
    associated
    glutamate (E)-rich
    protein 7,
    pseudogene 1 ///
    predicted gene
    7361
    10473356 ubiquitin- 10488507 abhydrolase
    conjugating enzyme domain containing
    E2L6 12
    10498350 purinergic receptor 10420668 microRNA 15a
    P2Y, G-protein
    coupled, 14
    10497862 transient receptor 10469951 ring finger
    potential cation protein 208
    channel, subfamily
    C, member 3
    10368056 epithelial cell 10501629 CDC14 cell
    transforming division cycle 14
    sequence 2 homolog A
    oncogene-like (S. cerevisiae)
    10425357 Smith-Magenis 10386789 Unc-51 like
    syndrome kinase 2
    chromosome region, (C. elegans)
    candidate 7-like
    (human)
    10498952 guanylate cyclase 1, 10401138 ATPase, H+
    soluble, alpha 3 transporting,
    lysosomal V1
    subunit D
    10548905 epidermal growth 10554118 family with
    factor receptor sequence
    pathway substrate 8 similarity 169,
    member B
    10579703 calcium 10603843 synapsin I
    homeostasis
    endoplasmic
    reticulum protein ///
    RIKEN cDNA
    1700030K09 gene
    10404630 RIO kinase 1 10575184 WW domain
    (yeast) containing E3
    ubiquitin protein
    ligase 2
    10518069 EF hand domain 10537712 glutathione S-
    containing 2 transferase kappa
    1
    10469672 glutamic acid 10511541 dpy-19-like 4
    decarboxylase 2 (C. elegans)
    10526941 RIKEN cDNA 10394816 predicted gene
    D830046C22 gene 9282
    10567448 dynein, axonemal, 10587503 SH3 domain
    heavy chain 3 binding glutamic
    acid-rich protein
    like 2
    10437885 myosin, heavy 10411359 proteolipid
    polypeptide 11, protein 2
    smooth muscle
    10600122 X-linked 10579939 ubiquitin specific
    lymphocyte- peptidase 38 ///
    regulated 3B /// X- predicted gene
    linked lymphocyte- 9725
    regulated 3C /// X-
    linked lymphocyte-
    regulated 3A
    10587665 RIKEN cDNA 10370242 poly(rC) binding
    4930579C12 gene protein 3
    10350753 glutamate-
    ammonia ligase
    (glutamine
    synthetase)
    10456296 mucosa
    associated
    lymphoid tissue
    lymphoma
    translocation gene
    1
    10380571 guanine
    nucleotide binding
    protein (G
    protein), gamma
    transducing
    activity
    polypeptide 2 ///
    ABI gene family,
    member 3
    10369413 sphingosine
    phosphate lyase 1
    10552276 ubiquitin-
    conjugating
    enzyme E2H ///
    predicted gene
    2058
    10394532 ubiquitin-
    conjugating
    enzyme E2F
    (putative) ///
    ubiquitin-
    conjugating
    enzyme E2F
    (putative)
    pseudogene
    10556463 aryl hydrocarbon
    receptor nuclear
    translocator-like
    10471994 kinesin family
    member 5C
    10395328 sorting nexin 13
    10599348 glutamate
    receptor,
    ionotropic,
    AMPA3 (alpha 3)
    10601595 RIKEN cDNA
    3110007F17 gene ///
    predicted gene
    6604 ///
    predicted gene
    5167 ///
    predicted gene
    2411 ///
    predicted gene
    14957
    10372891 SLIT-ROBO Rho
    GTPase activating
    protein 1
    10355024 islet cell
    autoantigen 1-like
    10518147 podoplanin
    10473537 olfactory receptor
    1123
    10424411 tumor
    susceptibility gene
    101
    10439960 centrosomal
    protein 97
    10551852 CAP-GLY
    domain containing
    linker protein 3
    10599291 reproductive
    homeobox 4E ///
    reproductive
    homeobox 4G ///
    reproductive
    homeobox 4F ///
    reproductive
    homeobox 4A ///
    reproductive
    homeobox 4C ///
    reproductive
    homeobox 4B ///
    reproductive
    homeobox 4D
    10587315 glutathione S-
    transferase, alpha
    4
    10447167 metastasis
    associated 3
    10480288 nebulette
    10491300 SKl-like
    10596637 mitogen-activated
    protein kinase-
    activated protein
    kinase 3
    10518019 DNA-damage
    inducible protein
    2 /// regulatory
    solute carrier
    protein, family 1,
    member 1
    10384685 RIKEN cDNA
    1700093K21 gene
    (0439483 Rho GTPase
    activating protein
    31
    10353844 neuralized
    homolog 3
    homolog
    (Drosophila)
    10459604 RIKEN cDNA
    4933403F05 gene
    10488892 transient receptor
    potential cation
    channel,
    subfamily C,
    member 4
    associated protein
    10542822 RAB15 effector
    protein
    10553354 neuron navigator
    2
    10425966 ataxin 10
    10360506 thymoma viral
    proto-oncogene 3
    10531610 RasGEF domain
    family, member
    1B
    10417787 guanine
    nucleotide binding
    protein (G
    protein), gamma 2
    10381588 granulin
    10437080 tetratricopeptide
    repeat domain 3
    10509560 ribosomal protein
    L38
    10466886 na
    10580457 NEDD4 binding
    protein 1
    10451061 runt related
    transcription
    factor 2
    10433953 yippee-like 1
    (Drosophila)
    10447461 stonin 1
    10501909 methyltransferase
    like 14 /// Sec24
    related gene
    family, member D
    (S. cerevisiae)
    10519693 sema domain,
    immunoglobulin
    domain (Ig), short
    basic domain,
    secreted,
    (semaphorin) 3D
    10385557 CCR4-NOT
    transcription
    complex, subunit
    6
    10413047 plasminogen
    activator,
    urokinase
    10406663 arylsulfatase B
    10430113 Rho GTPase
    activating protein
    39
    10475830 mitochondrial
    ribosomal protein
    S5
    10410892 RAS p21 protein
    activator 1
    10515994 stromal
    membrane-
    associated
    GTPase-activating
    protein 2
    10410099 CDC14 cell
    division cycle 14
    homolog B
    (S. cerevisiae)
    10428157 ring finger
    protein 19A
    10563643 tumor
    susceptibility gene
    101
    10412260 follistatin ///
    thyroid hormone
    receptor
    associated protein
    3
    10386539 similar to
    ubiquitin A-52
    residue ribosomal
    protein fusion
    product 1
    10415574 cyclin 1
    10494978 protein tyrosine
    phosphatase, non-
    receptor type 22
    (lymphoid)
    10511416 thymocyte
    selection-
    associated high
    mobility group
    box
    10562500 dpy-19-like 3
    (C. elegans)
    10568135 proline-rich
    transmembrane
    protein 2 ///
    RIKEN cDNA
    2900092E17 gene
    10514466 Jun oncogene
    10500847 membrane
    associated
    guanylate kinase,
    WW and PDZ
    domain containing
    3
    10549760 zinc finger
    protein 580
    10549377 RIKEN cDNA
    1700034J05 gene
    10430174 apolipoprotein L9a ///
    apolipoprotein L9b
    10474333 elongation
    protein 4 homolog
    (S. cerevisiae)
    10560791 predicted gene,
    EG381936 ///
    predicted gene
    6176
    10407159 ankyrin repeat
    domain 55
    10603659 mediator complex
    subunit 14
    10576854 cortexin 1
    10353775 BEN domain
    containing 6
    10573865 predicted gene
    3579
    10356886 solute carrier
    organic anion
    transporter family,
    member 4C1
    10507273 phosphatidylinositol
    3 kinase,
    regulatory
    subunit,
    polypeptide 3
    (p55)
    10424252 WDYHV motif
    containing 1
    10518735 splA/ryanodine
    receptor domain
    and SOCS box
    containing 1
    10562576 pleckstrin
    homology domain
    containing, family
    F (with FYVE
    domain) member
    1
    10375667 ring finger
    protein 130
    10528268 protein tyrosine
    phosphatase, non-
    receptor type 12
    10593205 REX2, RNA
    exonuclease 2
    homolog
    (S. cerevisiae)
    10576056 microtubule-
    associated protein
    1 light chain 3
    beta
    10547916 parathymosin
    10377689 gamma-
    aminobutyric acid
    receptor
    associated protein
    10602307 ovary testis
    transcribed ///
    predicted gene
    15085 ///
    predicted gene
    15127 ///
    predicted gene,
    OTTMUSG00000019001 ///
    leucine zipper
    protein 4 ///
    predicted gene
    15097 ///
    predicted gene
    15091 ///
    predicted gene
    10439 ///
    predicted gene
    15128
    10426835 DIP2 disco-
    interacting protein
    2 homolog B
    (Drosophila)
    10439798 DAZ interacting
    protein 3, zinc
    finger
    10375614 glutamine
    fructose-6-
    phosphate
    transaminase 2
    10361882 NHS-like 1
    10419274 glia maturation
    factor, beta
    10424781 glutamate
    receptor,
    ionotropic, N-
    methyl D-
    aspartate-
    associated protein
    1 (glutamate
    binding)
    10546960 na
    10514713 WD repeat
    domain 78
    10394954 grainyhead-like 1
    (Drosophila)
    10437205 Purkinje cell
    protein 4
    10464251 attractin like 1
    10496251 3-
    hydroxybutyrate
    dehydrogenase,
    type 2
    10396383 solute carrier
    family 38,
    member 6
    10585794 cytochrome P450,
    family 11,
    subfamily a,
    polypeptide 1
    10385719 Sec24 related
    gene family,
    member A
    (S. cerevisiae)
    10407358 polyadenylate
    binding protein-
    interacting protein
    1
    10498775 golgi integral
    membrane protein
    4
    10584435 von Willebrand
    factor A domain
    containing 5A
    10466304 deltex 4 homolog
    (Drosophila)
    10598292 forkhead box P3 ///
    RIKEN cDNA
    4930524L23 gene ///
    coiled-coil
    domain containing
    22
    10472440 Taxi (human T-
    cell leukemia
    virus type I)
    binding protein 3
    10398455 protein
    phosphatase 2,
    regulatory subunit
    B (B56), gamma
    isoform
    10493076 SH2 domain
    protein 2A
    10409152 RIKEN cDNA
    1110007C09 gene
    10542880 RIKEN cDNA
    4833442J19 gene
    10378523 Smg-6 homolog,
    nonsense
    mediated mRNA
    decay factor
    (C. elegans)
    10531560 anthrax toxin
    receptor 2
    10467319 retinol binding
    protein 4, plasma
    10395978 predicted gene
    527
    10471715 mitochondrial
    ribosome
    recycling factor
    10511755 WW domain
    containing E3
    ubiquitin protein
    ligase 1
    10353754 zinc finger
    protein 451
    10477572 chromatin
    modifying protein
    4B
    10359161 sterol O-
    acyltransferase 1
    10462035 lactate
    dehydrogenase B
    10543319 family with
    sequence
    similarity 3,
    member C
    10579052 predicted gene
    10033
    10475532 sulfide quinone
    reductase-like
    (yeast)
    10428857 metastasis
    suppressor 1
    10475144 calpain 3 ///
    glucosidase,
    alpha; neutral C
    10396645 zinc finger and
    BTB domain
    containing 1
    10428302 Kruppel-like
    factor 10
    10577882 heparan-alpha-
    glucosaminide N-
    acetyltransferase
    10548069 dual-specificity
    tyrosine-(Y)-
    phosphorylation
    regulated kinase 4
    10436053 developmental
    pluripotency
    associated 2
    10401564 RIKEN cDNA
    1110018G07 gene
    10471535 family with
    sequence
    similarity 129,
    member B
    10349404 mannoside
    acetylglucosaminyltransferase 5
    10520173 amiloride-
    sensitive cation
    channel 3
    10508860 solute carrier
    family 9
    (sodium/hydrogen
    exchanger),
    member 1
    10374500 vacuolar protein
    sorting 54 (yeast)
    10387723 RIKEN cDNA
    2810408A11 gene
    10488020 thioredoxin-
    related
    transmembrane
    protein 4
    10411126 junction-
    mediating and
    regulatory protein
    10345706 DNA segment,
    Chr 1, Brigham &
    Womens Genetics
    0212 expressed
    10364375 cystatin B
    10480379 mitochondrial
    ribosomal protein
    S5
    10521243 G protein-
    coupled receptor
    kinase 4
    10497920 ankyrin repeat
    domain 50
    10593723 acyl-CoA
    synthetase
    bubblegum family
    member 1
    10375634 mitogen-activated
    protein kinase 9
    10384555 aftiphilin
    10468113 Kv channel-
    interacting protein
    2
    10423363 progressive
    ankylosis
    10538150 transmembrane
    protein 176A
    10396485 synaptic nuclear
    envelope 2
    10401007 protein
    phosphatase 2,
    regulatory subunit
    B (B56), epsilon
    isoform
    10419151 eosinophil-
    associated,
    ribonuclease A
    family, member 1
    10390768 SWI/SNF related,
    matrix associated,
    actin dependent
    regulator of
    chromatin,
    subfamily e,
    member 1
    10478145 protein
    phosphatase 1,
    regulatory
    (inhibitor) subunit
    16B
    10433057 calcium binding
    and coiled coil
    domain 1
    10545921 MAX
    dimerization
    protein 1
    10392449 WD repeat
    domain,
    phosphoinositide
    interacting 1
    10545608 sema domain,
    immunoglobulin
    domain (Ig), TM
    domain, and short
    cytoplasmic
    domain
    10567219 ADP-ribosylation
    factor-like 6
    interacting protein
    1
    10471201 c-abl oncogene 1,
    receptor tyrosine
    kinase
    10505841 predicted gene
    13271 ///
    predicted gene
    13290 ///
    predicted gene
    13277 ///
    predicted gene
    13276
    10414360 lectin, galactose
    binding, soluble 3
    10403258 guanosine
    diphosphate
    (GDP)
    dissociation
    inhibitor 2
    10476759 Ras and Rab
    interactor 2
    10430866 cytochrome P450,
    family 2,
    subfamily d,
    polypeptide 10
    10432619 POU domain,
    class 6,
    transcription
    factor 1
    10521972 protocadherin 7
    10350646 ER degradation
    enhancer,
    mannosidase
    alpha-like 3
    10440993 regulator of
    calcineurin 1
    10505008 solute carrier
    family 44,
    member 1
    10566670 olfactory receptor
    478
    10356172 phosphotyrosine
    interaction domain
    containing 1
    10418506 stabilin 1
    10419429 olfactory receptor
    723 /// olfactory
    receptor 724
    10581434 dipeptidase 2
    10401365 zinc finger,
    FYVE domain
    containing 1
    10591188 olfactory receptor
    843
    10565846 signal peptidase
    complex subunit 2
    homolog
    (S. cerevisiae)
    10467258 myoferlin
    10548547 predicted gene
    6600
    10523012 deoxycytidine
    kinase
    10348547 ubiquitin-
    conjugating
    enzyme E2F
    (putative)
    10483667 corepressor
    interacting with
    RBPJ, 1
    10584071 PR domain
    containing 10
    10585249 protein
    phosphatase 2
    (formerly 2A),
    regulatory subunit
    A (PR 65), beta
    isoform
    10546137 ankyrin repeat
    and BTB (POZ)
    domain containing
    1
    10484720 olfactory receptor
    1166
    10571415 vacuolar protein
    sorting 37 A
    (yeast)
    10595189 solute carrier
    family 17
    (anion/sugar
    transporter),
    member 5
    10584426 olfactory receptor
    910
    10585986 myosin IXa
    10401753 VPS33B
    interacting
    protein, apical-
    basolateral
    polarity regulator
    10349793 dual
    serine/threonine
    and tyrosine
    protein kinase
    10527528 SWI/SNF related,
    matrix associated,
    actin dependent
    regulator of
    chromatin,
    subfamily e,
    member 1
    10485767 olfactory receptor
    1277
    10557459 mitogen-activated
    protein kinase 3
    10471486 endoglin
    10420846 frizzled homolog
    3 (Drosophila)
    10405849 olfactory receptor
    466
    10568691 RIKEN cDNA
    A130023I24 gene
    10351111 dynamin 3,
    opposite strand ///
    microRNA 214 ///
    microRNA 199a-2
    10540785 RIKEN cDNA
    6720456B07 gene
    10540923 makorin, ring
    finger protein, 2
    10413416 interleukin 17
    receptor D
    10386636 ubiquitin specific
    peptidase 22
    10383799 transcobalamin 2
    Genes differentially Genes upregulated Genes downregulated on
    upregulated in TH-GM cells on TH-GM surface TH-GM cells surface
    Gene Gene Gene
    ID Gene Title ID Gene Title Symbol Gene Title
    10385918 interleukin 3 10435704 CD80 antigen Ly6a lymphocyte
    antigen 6 complex,
    locus A
    10511363 preproenkephalin 10548409 killer cell lectin- Cd27 CD27 antigen
    like receptor
    subfamily C,
    member 1
    10497878 interleukin 2 10421737 tumor necrosis Sell selectin,
    factor (ligand) lymphocyte
    superfamily,
    member 11
    10385912 colony 10597420 chemokine (C-C Ctsw cathepsin W
    stimulating factor motif) receptor 4
    2 (granulocyte-
    macrophage)
    10404422 serine (or 10441633 chemokine (C-C Ltb lymphotoxin B
    cysteine) motif) receptor 6
    peptidase
    inhibitor, clade
    B, member 6b
    10408689 neuritin 1 10365933 early endosome Gngt2 guanine nucleotide
    antigen 1 binding protein (G
    protein), gamma
    transducing activity
    polypeptide 2 ///
    ABI gene family,
    member 3
    10467979 stearoyl- 10404840 CD83 antigen Gpr18 G protein-coupled
    Coenzyme A receptor 18
    desaturase 1
    10469312 phosphotriesterase 10359434 Fas ligand (TNF Igfbp4 insulin-like growth
    related /// superfamily, factor binding
    Clq-like 3 member 6) protein 4
    10435704 CD80 antigen 10344966 lymphocyte Il17ra interleukin 17
    antigen 96 receptor A
    10502655 cysteine rich 10345752 interleukin 1 Il18r1 interleukin 18
    protein 61 receptor, type II receptor 1
    10350159 ladinin 10439527 T cell Klrd1 killer cell lectin-
    immunoreceptor like receptor,
    with Ig and ITIM subfamily D,
    domains member 1
    10548409 killer ceil lectin- 10494595 Notch gene Mctp2 multiple C2
    like receptor homolog 2 domains,
    subfamily C, (Drosophila) transmembrane 2
    member 1
    10571399 zinc finger, 10597279 chemokine (C-C Ms4a6b membrane-
    DHHC domain motif) receptor- spanning 4-
    containing 2 like 2 domains, subfamily
    A, member 6B
    10538791 TNFAIP3 10485405 CD44 antigen Pld3 phospholipase D
    interacting family, member 3
    protein 3
    10407126 polo-like kinase 10561104 AXL receptor Pyhin1 pyrin and HIN
    2 (Drosophila) tyrosine kinase domain family,
    member 1
    10355984 serine (or 10585048 cell adhesion S1pr1 sphingosine-1-
    cysteine) molecule 1 phosphate receptor
    peptidase 1
    inhibitor, clade
    E, member 2
    10421737 tumor necrosis Slc44a2 solute carrier
    factor (ligand) family 44, member
    superfamily, 2
    member 11
    10503023 cystathionase
    (cystathionine
    gamma-lyase)
    10389207 chemokine (C-C
    motif) ligand 5
    10361887 PERP, TP53
    apoptosis
    effector
    10530841 insulin-like
    growth factor
    binding protein 7
    10504838 nuclear receptor
    subfamily 4,
    group A, member
    3
    10482762 isopentenyl-
    diphosphate delta
    isomerase
    10597420 chemokine (C-C
    motif) receptor 4
    10441633 chemokine (C-C
    motif) receptor 6
    10595402 family with
    sequence
    similarity 46,
    member A
    10480139 Clq-like 3 ///
    phosphotriesterase
    related
    10540472 basic helix-loop-
    helix family,
    member e40
    10404429 serine (or
    cysteine)
    peptidase
    inhibitor, clade
    B, member 9
    10595404 family with
    sequence
    similarity 46,
    member A
    10365933 early endosome
    antigen 1
    10384373 fidgetin-like 1
    10400072 scinderin
    10377938 enolase 3, beta
    muscle
    10589994 eomesodermin
    homolog
    (Xenopus laevis)
    10404840 CD83 antigen
    10485624 proline rich Gla
    (G-
    carboxyglutamic
    acid) 4
    (transmembrane)
    10369102 predicted gene
    9766
    10505030 fibronectin type
    III and SPRY
    domain
    containing 1-like
    10606868 brain expressed
    gene 1
    10501832 ATP-binding
    cassette, sub-
    family D (ALD),
    member 3
    10457225 mitogen-
    activated protein
    kinase kinase
    kinase 8
    10554521 phosphodiesterase
    8A
    10446229 tumor necrosis
    factor (ligand)
    superfamily,
    member 9
    10593842 tetraspanin 3
    10407211 phosphatidic
    acid phosphatase
    type 2A
    10488655 BCL2-like 1
    10470182 brain expressed
    myelocytomatosis
    oncogene
    10445977 Epstein-Barr
    virus induced
    gene 3
    10587495 interleukin-1
    receptor-
    associated kinase
    1 binding protein
    1
    10419082 RIKEN cDNA
    5730469M10
    gene
    10472212 plakophilin 4
    10487588 interleukin 1
    alpha
    10359434 Fas ligand (TNF
    superfamily,
    member 6)
    10351015 serine (or
    cysteine)
    peptidase
    inhibitor, clade C
    (antithrombin),
    member 1
    10344966 lymphocyte
    antigen 96
    10488415 cystatin C
    10598771 monoamine
    oxidase A
    10345752 interleukin 1
    receptor, type II
    10588577 cytokine
    inducible SH2-
    containing
    protein
    10439527 T cell
    immunoreceptor
    with Ig and ITIM
    domains
    10511258 family with
    sequence
    similarity 132,
    member A
    10403584 nidogen 1
    10399973 histone
    deacetylase 9
    10494595 Notch gene
    homolog 2
    (Drosophila)
    10346168 signal transducer
    and activator of
    transcription 4
    10350630 family with
    sequence
    similarity 129,
    member A
    10564667 neurotrophic
    tyrosine kinase,
    receptor, type 3
    10419288 GTP
    cyclohydrolase 1
    10407535 ribosomal
    protein L10A ///
    ribosomal protein
    L10A,
    pseudogene 2
    10468945 acyl-Coenzyme
    A binding
    domain
    containing 7
    10435271 HEG homolog 1
    (zebrafish)
    10576639 neuropilin 1
    10505059 T-cell acute
    lymphocytic
    leukemia 2
    10457091 neuropilin
    (NRP) and
    tolloid (TLL)-
    like 1
    10428081 heat-responsive
    protein 12
    10435712 CD80 antigen
    10597279 chemokine (C-C
    motif) receptor-
    like 2
    10485405 CD44 antigen
    10436662 microRNA 155
    10562044 zinc finger and
    BTB domain
    containing 32
    10463599 nuclear factor of
    kappa light
    polypeptide gene
    enhancer in B-
    cells 2, p49/p100
    10456005 CD74 antigen
    (invariant
    polypeptide of
    major
    histocompatibility
    complex, class
    II antigen-
    associated)
    10490903 carbonic
    anhydrase 13
    10468762 RIKEN cDNA
    4930506M07
    gene
    10470316 na
    10363195 heat shock factor
    2
    10596652 HemK
    methyltransferase
    family member 1
    10435693 cytochrome c
    oxidase, subunit
    XVII assembly
    protein homolog
    (yeast)
    10544660 oxysterol
    binding protein-
    like 3
    10384725 reticuloendotheliosis
    oncogene
    10408600 serine (or
    cysteine)
    peptidase
    inhibitor, clade
    B, member 6a
    10391444 RUN domain
    containing 1 ///
    RIKEN cDNA
    1700113I22 gene
    10561516 nuclear factor of
    kappa light
    polypeptide gene
    enhancer in B-
    cells inhibitor,
    beta
    10566846 DENN/MADD
    domain
    containing 5A
    10435048 Tctex1 domain
    containing 2
    10470175 lipocalin 13
    10586250 DENN/MADD
    domain
    containing 4A
    10512774 coronin, actin
    binding protein
    2A
    10366546 carboxypeptidase
    M
    10354286 KDEL (Lys-
    Asp-Glu-Leu)
    containing 1
    10547621 apolipoprotein B
    mRNA editing
    enzyme, catalytic
    polypeptide 1
    10440419 B-cell
    translocation
    gene 3 /// B-cell
    translocation
    gene 3
    pseudogene
    10407467 aldo-keto
    reductase family
    1, member E1
    10558580 undifferentiated
    embryonic cell
    transcription
    factor 1
    10544644 na
    10424543 WNT1 inducible
    signaling
    pathway protein
    1
    10507137 PDZK1
    interacting
    protein 1
    10384691 RIKEN cDNA
    0610010F05
    gene
    10565315 fumarylacetoacetate
    hydrolase
    10586248 DENN/MADD
    domain
    containing 4A
    10561104 AXL receptor
    tyrosine kinase
    10385837 interleukin 13
    10440393 SAM domain,
    SH3 domain and
    nuclear
    localization
    signals, 1
    10401987 potassium
    channel,
    subfamily K,
    member 10
    10453715 RAB18, member
    RAS oncogene
    family
    10496466 alcohol
    dehydrogenase 4
    (class II),
    pipolypeptide
    10396712 fucosyltransferase 8
    10603708 calcium/calmodulin-
    dependent
    serine protein
    kinase (MAGUK
    family)
    10352178 saccharopine
    dehydrogenase
    (putative) ///
    similar to
    Saccharopine
    dehydrogenase
    (putative)
    10349081 PH domain and
    leucine rich
    repeat protein
    phosphatase 1
    10364950 growth arrest
    and DNA-
    damage-
    inducible 45 beta
    10566877 SET binding
    factor 2
    10575160 nuclear factor of
    activated T-cells
    5
    10458090 receptor
    accessory protein
    5
    10439845 predicted gene
    5486
    10461558 solute carrier
    family 15,
    member 3
    10586254 DENN/MADD
    domain
    containing 4A
    10574166 copine II
    10598467 proviral
    integration site 2
    10447084 galactose
    mutarotase
    10366346 pleckstrin
    homology-like
    domain, family
    A, member 1
    10355567 transmembrane
    BAX inhibitor
    motif containing
    1
    10407420 neuroepithelial
    cell transforming
    gene 1
    10411882 neurolysin
    (metallopeptidase
    M3 family)
    10585048 cell adhesion
    molecule 1
    10538890 hypothetical
    protein
    LOC641050
    10406681 adaptor-related
    protein complex
    3, beta 1 subunit
    10455647 tumor necrosis
    factor, alpha-
    induced protein 8
    10447521 transcription
    factor B1,
    mitochondrial ///
    T-cell lymphoma
    invasion and
    metastasis 2
    10523772 leucine rich
    repeat containing
    8D
    10417759 ubiquitin-
    conjugating
    enzyme E2E 2
    (UBC4/5
    homolog, yeast)
    10586244 DENN/MADD
    domain
    containing 4A
    10436500 glucan (1,4-
    alpha),
    branching
    enzyme 1
    10556297 adrenomedullin
    10593492 zinc finger
    CCCH type
    containing 12C
    10373358 interleukin 23,
    alpha subunit p19
    10358583 hemicentin 1
    10567995 nuclear protein 1
    10512030 RIKEN cDNA
    3110043021
    gene
    10594652 lactamase, beta
    10344960 transmembrane
    protein 70
    10399908 protein kinase,
    cAMP dependent
    regulatory, type
    II beta
    10605766 melanoma
    antigen, family
    D, 1
    10474141 solute carrier
    family 1 (glial
    high affinity
    glutamate
    transporter),
    member 2
    10461909 cDNA sequence
    BC016495
    10548030 CD9 antigen
    10525473 transmembrane
    protein 120B
    10435266 HEG homolog 1
    (zebrafish)
    10593483 ferredoxin 1
    10476569 RIKEN cDNA
    2310003L22
    gene
    10526718 sperm motility
    kinase 3A ///
    sperm motility
    kinase 3B ///
    sperm motility
    kinase 3C
    10547613 ribosomal
    modification
    protein rimK-like
    family member B
    10511446 aspartate-beta-
    hydroxylase
    10375137 potassium large
    conductance
    calcium-activated
    channel,
    subfamily M,
    beta member 1
    10528154 predicted gene
    6455 /// RIKEN
    cDNA
    4933402N22
    gene
    10514173 ribosomal
    protein L34 ///
    predicted gene
    10154 ///
    predicted
    pseudogene
    10086 ///
    predicted gene
    6404
    10586227 DENN/MADD
    domain
    containing 4A
    10402648 brain protein 44-
    like
    10575745 ATM interactor
    10346255 ORM1-like 1
    (S. cerevisiae)
    10400405 nuclear factor of
    kappa light
    polypeptide gene
    enhancer in B-
    cells inhibitor,
    alpha
    10528527 family with
    sequence
    similarity 126,
    member A
    10472738 DDB 1 and
    CUL4 associated
    factor 17
    10368534 nuclear receptor
    coactivator
    7
    10407543 GTP binding
    protein
    4
    10376555 COP9
    (constitutive
    photomorphogenic)
    homolog,
    subunit 3
    (Arabidopsis
    thaliana)
    10567297 inositol 1,4,5-
    triphosphate
    receptor
    interacting
    protein-like 2
    10589886 RIKEN cDNA
    4930520004
    gene
    10423593 lysosomal-
    associated
    protein
    transmembrane
    4B
    10577954 RAB11 family
    interacting
    protein 1 (class I)
    10604528 muscleblind-like
    3 (Drosophila)
    10432675 RIKEN cDNA
    I730030J21 gene
    10385747 PHD finger
    protein
    15
    10398240 echinoderm
    microtubule
    associated
    protein like 1
    10511803 RIKEN cDNA
    2610029I01 gene
    10466606 annexin A1
    10520304 ARP3 actin-
    related protein 3
    homolog B
    (yeast)
    10425903 na
    10488709 RIKEN cDNA
    8430427H17
    gene
    10376096 acyl-CoA
    synthetase long-
    chain family
    member
    6
    10429491 activity
    regulated
    cytoskeletal-
    associated
    protein
    10439710 pleckstrin
    homology-like
    domain, family
    B, member 2
    10467110 expressed
    sequence
    AI747699
    10536898 interferon
    regulatory factor
    5
    10505044 fukutin
    10605370 membrane
    protein,
    palmitoylated
    10363669 DnaJ (Hsp40)
    homolog,
    subfamily C,
    member 12
    10496727 dimethylarginine
    dimethylaminohydrolase
    1
    10587683 B-cell
    leukemia/lymphoma
    2 related
    protein A1a ///
    B-cell
    leukemia/lymphoma
    2 related
    protein A1d ///
    B-cell
    leukemia/lymphoma
    2 related
    protein A1b ///
    B-cell
    leukemia/lymphoma
    2 related
    protein A1e
    10458816 toll like receptor
    adaptor molecule
    2
    10513008 Kruppel-like
    factor 4 (gut)
    10550906 plasminogen
    activator,
    urokinase
    receptor
    10362674 U3A small
    nuclear RNA
    10473190 DnaJ (Hsp40)
    homolog,
    subfamily C,
    member 10
    10477581 ribosomal
    protein L5
    10571774 aspartylglucosaminidase
    10395356 anterior gradient
    homolog 3
    (Xenopus laevis)
    10392440 solute carrier
    family 16
    (monocarboxylic
    acid
    transporters),
    member 6
    10352815 interferon
    regulatory factor
    6
  • REFERENCES
    • Afkarian, M., Sedy, J. R., Yang, J., Jacobson, N. G., Cereb, N., Yang, S. Y., Murphy. T. L., and Murphy, K. M. (2002). T-bet is a STAT1-induced regulator of IL-12R expression in naive CD4+ T cells. Nature immunology 3, 549-557.
    • Ansel, K. M., Djuretic, I., Tanasa, B., and Rao. A, (2006). Regulation of Th2 differentiation and 114 locus accessibility. Annual review of immunology 24, 607-656.
    • Baron. J. L., Madri, J. A., Ruddle, N. H., Hashim, G., and Janeway, C. A., Jr. (1993). Surface expression of alpha 4 integrin by CD4 T cells is required for their entry into brain parenchyma. J Exp Med 177, 57-68.
    • Bell, A. L., Magill. M. K., McKane, W. R., Kirk, F., and Irvine, A. E. (1995). Measurement of colony-stimulating factors in synovial fluid: potential clinical value. Rheumatol Int 14, 177-182.
    • Berner, B., Akca, D., Jung, T., Muller, G. A., and Reuss-Borst, M. A. (2000). Analysis of Th1 and Th2 cytokines expressing CD4+ and CD8+ T cells in rheumatoid arthritis by flow cytometry. J Rheumatol 27, 1128-1135.
    • Bettelli, E., Carrier, Y., Gao, W., Korn, T., Strom, T. B., Oukka, M., Weiner, H. L., and Kuchroo, V. K. (2006). Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235-238.
    • Brustic, A., Heink, S., Huber, M., Rosenplanter, C., Stadelmann, C., Yu. P., Arpaia, E., Mak, T. W., Kamradt, T., and Lohoff, M. (2007). The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4. Nature immunology 8, 958-966.
    • Burchill, M. A., Yang, J., Vogtenhuber, C., Blazar, B. R., and Farmr, M. A. (2007). IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J Immunol 178, 280-290.
    • Campbell. I. K., Rich, M. J., Bischof, R. J., Dunn. A. R., Grail. D., and Hamilton, J. A. (1998). Protection from collagen-induced arthritis in granulocyte-macrophage colony-stimulating factor-deficient mice. J Immunol 161, 3639-3644.
    • Campbell, I. K., van Nieuwenhuijze, A., Segura, E., O'Donnell, K., Coghill, E., Hommel, M., Gerondakis, S., Villadangos. J. A., and Wicks, I. P. (2011). Differentiation of inflammatory dendritic cells is mediated by NF-kappaB1-dependent GM-CSF production in CD4 T cells. J Immunol 186, 5468-5477.
    • Choy, E H., and Panayi, G. S. (2001). Cytokine pathways and joint inflammation in rheumatoid arthritis. N Engl J Med 344, 907-916.
    • Codarri, L., Gyulveszi, G., Tosevski, V., Hesske, L., Fontana. A., Magnenat, L., Suter, T., and Becher. B. (2011). RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol 12, 560-567.
    • Cook, A. D., Braine, E. L., Campbell, I. K., Rich, M. J., and Hamilton, J. A. (2001). Blockade of collagen-induced arthritis post-onset by antibody to granulocyte-macrophage colony-stimulating factor (GM-CSF): requirement for GM-CSF in the effector phase of disease. Arthritis Res 3, 293-298.
    • Cope, A. P., Schulze-Koops, H., and Aringer, M. (2007). The central role of T cells in rheumatoid arthritis. Clin Exp Rheumatol 25, S4-11.
    • Cornish, A. L., Campbell, I. K., McKenzie, B. S., Chatfield, S., and Wicks, I. P. (2009). G-CSF and GM-CSF as therapeutic targets in rheumatoid arthritis. Nat Rev Rheumatol 5, 554-559.
    • Croxford, A. L., Mair, F., and Becher, B. (2012). IL-23: one cytokine in control of autoimmunity. European journal of immunology 42, 2263-2273.
    • Cua, D. J., Sherlock, J., Chen, Y., Murphy, C. A., Joyce, B., Seymour, B., Lucian, L., To, W., Kwan. S., Churakova, T., et al. (2003). Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 421, 744-748.
    • El-Behi, M., Ciric, B., Dai, H., Yan, Y., Cullimore, M., Safavi, F., Zhang, G. X., Dittel, B. N., and Rostami, A. (2011). The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol 12, 568-575.
    • Gran. B., Zhang, G. X., Yu, S., Li. J., Chen, X. H., Ventura, E. S., Kamoun, M., and Rostami, A. (2002). IL-12p35-deficient mice are susceptible to experimental autoimmune encephalomyelitis: evidence for redundancy in the IL-12 system in the induction of central nervous system autoimmune demyelination. J Immunol 69, 7104-7110.
    • Gregory, S. G., Schmidt, S., Seth. P., Oksenberg, J. R., Hart, J., Prokop, A., Caillier, S. J., Ban, M., Goris, A., Barcellos, L. F., et al. (2007). Interleukin 7 receptor alpha chain (IL7R) shows allelic and functional association with multiple sclerosis. Nature genetics 39, 1083-1091.
    • Greter, M., Helft, J., Chow, A., Hashimoto, D., Mortha, A., Agudo-Cantcro, J., Bogunovic, M., Gautier, E. L., Miller, J., Leboeuf, M., et al., (2012). GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity 36, 1031-1046.
    • Guedez, Y. B., Whittington, K. B., Clayton, J. L., Joosten, L. A., van de Loo, F. A., van den Berg, W. B., and Rosloniec, E. F. (2001). Genetic ablation of interferon-gamma upregulates interleukin-1 beta expression and enables the elicitation of collagen-induced arthritis in a nonsusceptible mouse strain. Arthritis Rheum 44, 2413-2424.
    • Gutcher, I., and Becher, B. (2007). APC-derived cytokines and T cell polarization in autoimmune inflammation. J Clin Invest 117, 1119-1127.
    • Haak, S., Croxford, A. L., Kreymborg, K., Heppner, F. L., Pouly, S., Becher, B., and Waisman, A. (2009). IL-17A and IL-17F do not contribute vitally to autoimmune neuroinflammation in mice. J Clin Invest 119, 61-69.
    • Hamilton, J. A. (2008). Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol 8, 533-544.
    • Harrington, L. E., Hatton, R. D., Mangan, P. R., Turner, H., Murphy, T. L., Murphy,
    • K. M., and Weaver, C. T. (2005). Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 6, 1123-1132.
    • Hofstetter, 1-1.1-1., Ibrahim, S. M., Koczan, D., Kruse, N., Weishaupt, A., Toyka, K. V., and Gold, R. (2005). Therapeutic efficacy of IL-17 neutralization in murine experimental autoimmune encephalomyelitis. Cell Immunol 237, 123-130.
    • Irmler, I. M., Gajda, M., and Brauer. R. (2007). Exacerbation of antigen-induced arthritis in IFN-gamma-deficient mice as a result of unrestricted IL-17 response. J Immunol 179, 6228-6236.
    • Ivanov, I I. McKenzie, B. S., Zhou. L., Tadokoro, C. E., Lepelley, A., Lafaille, J. J., Cua, D. J., and Littman, D. R. (2006). The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+T helper cells. Cell 126, 1121-1133.
    • Kaplan, M. N., Schindler, U., Smiley, S. T., and Grusby, M. J. (1996a). Stat6 is required for mediating responses to IL-4 and for development of Th2 cells. Immunity 4, 313-319.
    • Kaplan, M. H., Sun, Y. L., Hoey, T., and Grusby, M. J. (1996b). Impaired IL-12 responses and enhanced development of Th2 cells in Stat4-deficient mice. Nature 382, 174-177.
    • Kolaczkowska, E., and Kubes, P. (2013). Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol 13, 159-175.
    • Komatsu, N., and Takayanagi, H. (2012). Autoimmune arthritis: the interface between the immune system and joints. Adv Immunol 115, 45-71.
    • Korn. T., Bettelli, E., Gao, W., Awasihi, A., Jager. A., Strom, T. B., Oukka, M., and Kuchroo, V. K. (2007). IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature 448, 484-487.
    • Langrish, C. L., Chen, Y., Blumenschein, W. M., Mattson, J., Basham, B., Sedgwick, J. D., McClanahan, T., Kastelein, R. A., and Cua, D. J. (2005). IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 201, 233-240.
    • Laurence, A., Tato. C. M., Davidson, T. S., Kanno, Y., Chen. Z., Yao, Z., Blank, R. B., Meylan, F., Siegel. R., Hennighausen, L., et al. (2007). Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 26, 371-381.
    • Lawlor, K. E., Wong, P. K., Campbell, P. K., van Rooijen, N., and Wicks, I. P. (2005). Acute CD4+T lymphocyte-dependent interleukin-1-driven arthritis selectively requires interleukin-2 and interleukin-4, joint macrophages, granulocyte-macrophage colony-stimulating factor, interleukin-6, and leukemia inhibitory factor. Arthritis Rheum 52, 3749-3754.
    • Lee, L. F., Logronio, K., Tu, G. H., Zhai, W., Ni, I., Mei, L., Dilley, J., Yu, J., Rajpal, A., Brown, C., et al. (2012). Anti-IL-7 receptor-alpha reverses established type 1 diabetes in nonobese diabetic mice by modulating effector T-cell function. Proc Natl Acad Sci USA 109, 12674-12679.
    • Leonard, J. P., Waldburger, K. E., and Goldman, S. J. (1995). Prevention of experimental autoimmune encephalomyelitis by antibodies against interleukin 12. J Exp Med 181.381-386.
    • Lighvani, A. A., Frucht, D. M., Jankovic, D., Yamanc, H., Aliberti, J., Hissong, B. D., Nguyen, B. V., Gadina, M., Sher. A., Paul, W. E., and O'Shea. J. J. (2001). T-bet is rapidly induced by interferon-gamma in lymphoid and myeloid cells. Proc Natl Acad Sci USA 98, 15137-15142.
    • Liu, X., Lee, Y. S., Yu, C. R., and Egwuagu, C. E. (2008). Loss of STAT5 in CD4+ T cells prevents development of experimental autoimmune diseases. J Immunol 180, 6070-6076.
    • Lock, C., Hermans, G., Pedotti, R., Brendolan, A., Schadt, E., Garren, H., Langer-Gould, A., Stroher, S., Cannella, B., Allard, J., et al. (2002). Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med 8, 500-508.
    • Lundmark, F., Duvefelt. K., Iacobaeus, E., Kockum, I., Wallstrom, E., Khademi, M., Oturai, A., Ryder, L. P., Saatchi, J., Harbo, H. F., et al. (2007). Variation in interleukin 7 receptor alpha chain (IL7R) influences risk of multiple sclerosis: Nature genetics 39, 1108-1113.
    • Manoury-Schwartz, B., Chiocchia, G., Bcssis, N., Ahchsira-Amar, O., Batteux, F., Muller, S., Huang, S., Boissier, M. C., and Fournier, C. (1997). High susceptibility to collagen-induced arthritis in mice lacking IFN-gamma receptors. J Immunol 158, 5501-5506.
    • MeGeachy, M. J., Chen, Y., Tato, C. M., Laurence, A., Joyce-Shaikh, B., Blumenschein, W. M., McClanahan, T. K., O'Shea, J. J., and Cua, D. J. (2009). The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat Immunol 10, 314-324.
    • McInnes, I. B., and Schen, G. (2007). Cytokines in the pathogenesis of rheumatoid arthritis. Nat Rev Immunol 7, 429-442.
    • McInnes, I. B., and Schen, G. (2011). The pathogenesis of rheumatoid arthritis. N Engl J Med 365, 2205-2219.
    • Muller-Ladner, U., Ospelt, C., Gay, S., Distler, O., and Pap, T. (2007). Cells of the synovium in rheumatoid arthritis. Synovial fibroblasts. Arthritis Res Ther 9, 223.
    • Muller, J., Sperl, B., Reindl, W., Kiessling, A., and Berg, T. (2008). Discovery of chromone-based inhibitors of the transcription factor STAT5. Chembiochem: a European journal of chemical biology 9, 723-727.
    • Nurieva, R., Yang, X. O., Martinez, G., Zhang, Y., Panopoulos, A. D., Ma, L., Schluns, K., Tian, Q., Watowich, Jetten. A. M., and Dong, C. (2007). Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature 448, 480-483.
    • Okada. Y., Wu, D., Trynka, G., Raj, T., Terao, C., Ikari, K., Kochi, Y., Ohmura, K., Suzuki, A., Yoshida, S., et al. (2014). Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506, 376-381.
    • Pernis, A. B. (2009). Th17 cells in rheumatoid arthritis and systemic lupus erythematosus. J Intern Med 265, 644-652.
    • Plater-Zyberk. C., Joosten, L. A., Helsen, M. M., Hepp, J., Baeuerle, P. A., and van den Berg, W. B. (2007). GM-CSF neutralisation suppresses inflammation and protects cartilage in acute streptococcal cell wall arthritis of mice. Ann Rheum Dis 66, 452-457.
    • Ponomarev, E. D., Shriver, L. P., Maresz, K., Pedras-Vasconcelos, J., Verthelyi, D., and Dittel, B. N. (2007). GM-CSF production by autoreactive T cells is required for the activation of microglial cells and the onset of experimental autoimmune encephalomyelitis. J Immunol 178, 39-48.
    • Reboldi, A., Coisne, C., Baumjohann, D., Benvenuto, F., Bottinelli, D., Lira, S., Uccclli, A., Lanzavecchia, A., Engelhardt. B., and Sallusto, F. (2009). C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat Immunol 10, 514-523.
    • Segal, B. M., and Shevach, E. M. (1996). IL-12 unmasks latent autoimmune disease in resistant mice. J Exp Med 184, 771-775.
    • Shimoda, K., van Deursen, J., Sangster, M. Y., Sarawar, S. R., Carson, R. T., Tripp, R. A., Chu, C., Quelle, F. W., Nosaka, T., Vignali, D. A., et al. (1996). Lack of IL-4-induced Th2 response and IgE class switching in mice with disrupted Stat6 gene. Nature 380, 630-633.
    • Sonderegger, I., Iezzi, G., Maier, R., Schmitz, N., Kurrer, M., and Kopf, M. (2008). GM-CSF mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival. J Exp Med 205, 2281-2294.
    • Steinman, L. (2007). A brief history of T(H)17, the first major revision in the T(H)1/T(H)2 hypothesis of T cell-mediated tissue damage. Nat Med 13, 139-145.
    • Stritesky, G. L., Ych, N., and Kaplan. M. H. (2008). IL-23 promotes maintenance but not commitment to the Th17 lineage. J Immunol 181, 5948-5955.
    • Szabo, S. J., Sullivan, B. M., Stemmann, C., Satoskar, A. R., Sleckman, B. P., and Glimcher, L. H. (2002). Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells. Science 295, 338-342.
    • Takeda, K., Tanaka, T., Shi, W., Matsumoto, M., Minami, M., Kashiwamura, S., Nakanishi, K., Yoshida. N., Kishimoto, T., and Akira. S. (1996). Essential role of Stat6 in IL-4 signalling. Nature 380, 627-630.
    • Thierfelder. W. E., van Deursen. J. M., Yamamoto, K., Tripp, R. A., Sarawar, S. R., Carson, R. T., Sangster, M. Y., Vignali, D. A., Doherty, P. C., Grosveld, G. C., and Elle, J. N. (1996). Requirement for Stat4 in interleukin-12-mediated responses of natural killer and T cells. Nature 382, 171-174.
    • Veldhoen, M., Hirota, K., Westendorf, A. M., Buer, J., Dumoutier, L., Renauld, J. C., and Stockinger. B. (2008). The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453, 106-109.
    • Veldhoen, M., Hocking, R. J., Atkins, C. J., Locksley, R. M., and Stockinger, B. (2006). TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 24, 179-189.
    • Vermeire, K., Heremans, H., Vandeputte, M., Huang, S., Billiau, A., and Matthys, P. (1997). Accelerated collagen-induced arthritis in IFN-gamma receptor-deficient mice. J Immunol 158, 5507-5513.
    • Willenborg, D. O., Fordham, S., Bernard, C. C., Cowden, W. B., and Ramshaw, I. A. (1996). IFN-gamma plays a critical down-regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. J Immunol 157, 3223-3227.
    • Wright, H. L., Moots, R. J., and Edwards, S. W. (2014). The multifactoral role of neutrophils in rheumatoid arthritis. Nat Rev Rheumatol 10, 593-601.
    • Yamada, H., Nakashima, Y., Okazaki, K., Mawatari, T., Fukushi, J. I., Kaibara, N., Hori, A., Iwamoto, Y., and Yoshikai, Y. (2008). Th1 but not Th17 cells predominate in the joints of patients with rheumatoid arthritis. Ann Rheum Dis 67, 1299-1304.
    • Yang, X. O., Panopoulos, A. D., Nurieva, R., Chang, S. H., Wang, D., Watowich, S. S., and Dong, C. (2007a). STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. The Journal of biological chemistry 282, 9358-9363.
    • Yang, X. O., Pappu, B. P., Nurieva, R., Akimzhanov, A., Kang. H. S., Chung, Y., Ma, L., Shah, B., Panopoulos, A. D., Schluns, K. S., et al. (2008). T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity 28, 29-39.
    • Yang, X. P., Ghoreschi, K., Steward-Tharp, S. M., Rodriguez-Canales, J., Zhu, J., Grainger, J. R., Hirahara, K., Sun, H. W., Wei, L., Vahedi. G., et al. (2011). Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat Immunol 12, 247-254.
    • Yang, Y., Ochando, J. C., Bromberg, J. S., and Ding, Y. (2007b). Identification of a distant T-bet enhancer responsive to IL-12/Stat4 and IFNgamma/Stat1 signals. Blood 110, 2494-2500.
    • Yang, Y. H., and Hamilton. J. A. (2001). Dependence of interleukin-1-induced arthritis on granulocyte-macrophage colony-stimulating factor. Arthritis Rheum 44, 111-119.
    • Yao, Z., Cui, Y., Watford, W. T., Bream, J. H., Yamaoka, K., Hissong, B. D., Li, D., Durum, S. K., Jiang, Q., Bhandoola, A., et al. (2006). Stat5a/h are essential for normal lymphoid development and differentiation. Proc Natl Acad Sci USA 103, 1000-1005.
    • Zhang, G. X., Gran, B., Yu, S., Li. J., Siglienti, L., Chen, X., Kamoun, M., and Rostami, A. (2003). Induction of experimental autoimmune encephalomyelitis in IL-12 receptor-beta 2-deficient mice: IL-12 responsiveness is not required in the pathogenesis of inflammatory demyelination in the central nervous system. J Immunol 170, 2153-2160.
    • Zhou, L., Ivanov, I I, Spolski, R., Min, R., Shenderov, K., Egawa, T., Levy, D. E., Leonard. W. J., and Littman. D. R. (2007). IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nature immunology 8, 967-974.
    • Zhu, J., and Paul, W. E. (2010). Peripheral CD4+ T-cell differentiation regulated by networks of cytokines and transcription factors. Immunological reviews 238, 247-262.
  • While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (20)

1.-33. (canceled)
34. A method of treating an inflammatory disease associated with a TNF-α-independent inflammatory pathway, comprising administering to a subject in need thereof an effective amount of a signal transducer and activator of transcription 5 (STAT5) inhibitor, wherein the inflammatory disease is mediated by granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting T-helper (Th-GM) cells, wherein the STAT5 inhibitor reduces serum level of GM-CSF in the subject and thereby providing a therapeutic benefit to the subject in a TNF-α-independent manner.
35. The method of claim 34, wherein the inflammatory disease is rheumatoid arthritis.
36. The method of claim 34, wherein the inflammatory disease is multiple sclerosis.
37. The method of claim 34, wherein the TH-GM cells are differentiated from precursor CD4+ cells in the presence of activated STAT5 and IL-7.
38. The method of claim 34, wherein the STAT5 inhibitor is pimozide.
39. The method of claim 34, wherein the STAT5 inhibitor is CAS 285986-31-4.
40. The method of claim 34, wherein the STAT5 inhibitor is an antibody that specifically binds to STAT5.
41. The method of claim 34, wherein the STAT5 inhibitor is an antisense nucleic acid, small interfering RNA, short hairpin RNA, or microRNA.
42. A method of treating rheumatoid arthritis, comprising administering to a subject in need thereof an effective amount of (A) a STAT5 inhibitor and (B) a therapeutic agent that inhibits the TNF-α inflammatory pathway.
43. The method of claim 42, wherein the STAT5 inhibitor is pimozide.
44. The method of claim 42, wherein the STAT5 inhibitor is CAS 285986-31-4.
45. The method of claim 42, wherein the STAT5 inhibitor is an antibody that specifically binds to STAT5.
46. The method of claim 42, wherein the STAT5 inhibitor is an antisense nucleic acid, small interfering RNA, short hairpin RNA, or microRNA.
47. The method of claim 42, wherein the therapeutic agent that inhibits the TNF-α inflammatory pathway is an etanercept, adalimumab, infliximab, golimumab, or certolizumab pegol.
48. The method of claim 42, wherein the therapeutic agent that inhibits the TNF-α inflammatory pathway is prednisone, methotrexate, or tofacitinib.
49. The method of claim 42, Wherein the therapeutic agent that inhibits the TNF-α inflammatory pathway is anakinra, abatacept, rituximab, or tocilizumab.
50. The method of claim 42, wherein (A) and (B) are administered sequentially.
51. The method of claim 42, wherein (A) and (B) are administered simultaneously.
52. The method of claim 42, wherein (A) and (B) are administered in a single dose.
US16/902,189 2014-09-26 2020-06-15 Methods and Compositions for Modulating TH-GM Cell Function Pending US20200408757A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/902,189 US20200408757A1 (en) 2014-09-26 2020-06-15 Methods and Compositions for Modulating TH-GM Cell Function

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
SG10201406130P 2014-09-26
SG10201406130P 2014-09-26
PCT/SG2015/050344 WO2016048247A1 (en) 2014-09-26 2015-09-25 Methods and compositions for modulating th-gm cell function
US201715514644A 2017-03-27 2017-03-27
US16/902,189 US20200408757A1 (en) 2014-09-26 2020-06-15 Methods and Compositions for Modulating TH-GM Cell Function

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/SG2015/050344 Continuation WO2016048247A1 (en) 2014-09-26 2015-09-25 Methods and compositions for modulating th-gm cell function
US15/514,644 Continuation US10690666B2 (en) 2014-09-26 2015-09-25 Methods and compositions for modulating TH-GM cell function

Publications (1)

Publication Number Publication Date
US20200408757A1 true US20200408757A1 (en) 2020-12-31

Family

ID=55581587

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/514,644 Active 2036-03-31 US10690666B2 (en) 2014-09-26 2015-09-25 Methods and compositions for modulating TH-GM cell function
US16/902,189 Pending US20200408757A1 (en) 2014-09-26 2020-06-15 Methods and Compositions for Modulating TH-GM Cell Function

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/514,644 Active 2036-03-31 US10690666B2 (en) 2014-09-26 2015-09-25 Methods and compositions for modulating TH-GM cell function

Country Status (9)

Country Link
US (2) US10690666B2 (en)
EP (2) EP3739039A1 (en)
JP (3) JP6729911B2 (en)
KR (3) KR102507694B1 (en)
CN (1) CN107002037A (en)
AU (3) AU2015322125B2 (en)
CA (2) CA2962757C (en)
SG (1) SG11201702475VA (en)
WO (1) WO2016048247A1 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11612595B2 (en) 2017-05-16 2023-03-28 Generos Biopharma Ltd. Compositions and methods for treating rheumatoid arthritis
JP2022528481A (en) * 2019-04-09 2022-06-10 ジェネラス バイオファーマ リミテッド Pharmaceutical compositions of pimozide and methotrexate, and their use
WO2020216335A1 (en) * 2019-04-26 2020-10-29 健艾仕生物医药有限公司 Use of compound in preparation of drug for treating atherosclerosis
WO2020216378A1 (en) 2019-04-26 2020-10-29 健艾仕生物医药有限公司 Heterocyclic compound, application thereof, and composition containing same
CN116369249B (en) * 2023-01-13 2024-05-17 四川轻化工大学 Construction method of zebra fish enteritis model

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012164398A2 (en) * 2011-06-01 2012-12-06 Netherlands Cancer Institute Modulation of the ubiquitin-proteasome system (ups)

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2002241303A (en) * 2001-02-13 2002-08-28 Hisamitsu Pharmaceut Co Inc Gm-csf of epithelial cell comprising ets transcription factor or gene coding for the same and/or defensin protein manifestation-controlling agent
WO2003038072A1 (en) * 2001-10-31 2003-05-08 Universite Libre De Bruxelles Generation and use of new types of dendritic cells
JP4711963B2 (en) 2003-09-15 2011-06-29 オクラホマ メディカル リサーチ ファウンデーション Methods using cytokine assays to diagnose, treat and evaluate inflammatory and autoimmune diseases
US7776313B2 (en) 2006-05-25 2010-08-17 University Health Network Methods of diagnosing and treating rheumatoid arthritis and osteoarthritis
US20080286234A1 (en) * 2007-05-15 2008-11-20 Eyink Daniel A Method for treating demyelinating diseases
WO2009039199A2 (en) * 2007-09-17 2009-03-26 Intradigm Corporation Compositions comprising stat5 sirna and methods of use thereof
TW200918553A (en) * 2007-09-18 2009-05-01 Amgen Inc Human GM-CSF antigen binding proteins
EA020766B1 (en) * 2008-07-08 2015-01-30 Борд Оф Риджентс, Дзе Юниверсити Оф Техас Систем Inhibitors of proliferation and activation of signal transducer and activator of transcription (stats)
EP2193790A1 (en) 2008-12-04 2010-06-09 Klinikum der Universität Regensburg IL-3 Inhibitors in use for treatment of rheumatoid arthritis in an early stage
JP2013520208A (en) * 2010-02-24 2013-06-06 ザ・ボード・オブ・トラスティーズ・オブ・ザ・レランド・スタンフォード・ジュニア・ユニバーシティ Diagnosis, prognosis and treatment of autoimmune diseases
KR20120128492A (en) * 2011-05-17 2012-11-27 연세대학교 산학협력단 Pharmaceutical Compositions for Preventing or Treating Bone Disorders
KR20140103122A (en) * 2011-11-17 2014-08-25 넨키 인스티튜트 오브 익스페리멘탈 바이올로지 Compositions and methods for treating glioma
EP2669295A1 (en) * 2012-05-29 2013-12-04 Universitätsklinikum Regensburg Diagnostic method for determining the presence and amount of human interleukin-3 in a sample using novel IL-3 antibodies

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012164398A2 (en) * 2011-06-01 2012-12-06 Netherlands Cancer Institute Modulation of the ubiquitin-proteasome system (ups)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Dougan et al (Immunity, 2019; 50:796-811) (Year: 2019) *
Freire et al (Periodontol 2000, 2013; 63(1):149-164) (Year: 2000) *
Tabas et al (Science, 2013; 339(6116):166-172) (Year: 2013) *

Also Published As

Publication number Publication date
AU2021201241B2 (en) 2022-08-04
AU2022256995A1 (en) 2022-11-24
KR20220025115A (en) 2022-03-03
JP7106604B2 (en) 2022-07-26
CA2962757C (en) 2022-03-22
CA2962757A1 (en) 2016-03-31
AU2015322125B2 (en) 2021-01-07
KR102360824B1 (en) 2022-02-09
US20170219581A1 (en) 2017-08-03
KR20210033037A (en) 2021-03-25
JP2022140482A (en) 2022-09-26
AU2015322125A1 (en) 2017-04-20
AU2021201241A1 (en) 2021-03-11
KR102507694B1 (en) 2023-03-08
CN107002037A (en) 2017-08-01
EP3198007A4 (en) 2018-08-08
JP2020165991A (en) 2020-10-08
KR20170066451A (en) 2017-06-14
WO2016048247A1 (en) 2016-03-31
EP3198007B1 (en) 2023-11-01
EP3198007A1 (en) 2017-08-02
KR102221013B1 (en) 2021-03-02
SG11201702475VA (en) 2017-04-27
CA3147152A1 (en) 2016-03-31
EP3739039A1 (en) 2020-11-18
JP6729911B2 (en) 2020-07-29
JP2017531792A (en) 2017-10-26
US10690666B2 (en) 2020-06-23

Similar Documents

Publication Publication Date Title
US20200408757A1 (en) Methods and Compositions for Modulating TH-GM Cell Function
Garo et al. Smad7 controls immunoregulatory PDL2/1-PD1 signaling in intestinal inflammation and autoimmunity
Medina et al. Ebi3 prevents Trypanosoma cruzi-induced myocarditis by dampening IFN-γ-driven inflammation
US20170233816A1 (en) Markers of immune response
Julián et al. CD26/DPPIV inhibition alters the expression of immune response-related genes in the thymi of NOD mice
US20140234843A1 (en) Methods and compositions for expanding cells and improving engraftment
정다운 E prostanoid receptor 4 expressing macrophages mediate mucosal repair by production of Cxcl1 during colitis
Marciniak Involvement of Notch pathway in synovial fibroblasts and hematopoietic cells in experimental models of arthritis
Fu The Transcriptional Regulation and Function of IL-9 in Allergic Lung Inflammation and Tumor Growth
US20220018853A1 (en) Use of gilz as a biomarker in sepsis
Lin et al. Increased Expression of TLR8 in Peripheral Blood CD16+ Monocytes Positively Correlates with Disease Activity in Anti-CCP+ Rheumatoid Arthritis Patients
Vázquez Targeting the IL-23/Th17 pathway to treat Myasthenia Gravis
Clark Studies on Myeloid Cell Functions in Periodontitis
Jaufmann Regulation of innate and adaptive B-cell responses by the proteins SLy2 and CXCR1
CA3180476A1 (en) Markers and cellular antecedents of rheumatoid arthritis flares
Waddell Role of macrophages and eosinophils in inflammatory bowel diseases
Ridgley Cytokine mediated CD4+ T-cell dysregulation in early rheumatoid arthritis
Mohamad Disease mechanism in two novel primary immunodeficiencies characterized by increased viral susceptibility and immune dysregulation
Polletti Reorganization of the macrophage epigenome during sustained stimulation
Malmhäll T cell subsets in asthma and allergy-role of glucocorticoids, plasticity and microRNA-155
Verjans et al. The cAMP response element modulator (CREM) regulates T
Campbell Harding Regulation of IL-4 mediated signalling in primary human bronchial fibroblasts by IL-13Rα2

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL UNIVERSITY OF SINGAPORE, SINGAPORE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FU, XIN-YUAN;SHENG, WANQIANG;ZHANG, YONGLIANG;AND OTHERS;REEL/FRAME:052944/0582

Effective date: 20151009

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED