WO2010003002A9 - Modulation of follicular helper t cells - Google Patents
Modulation of follicular helper t cells Download PDFInfo
- Publication number
- WO2010003002A9 WO2010003002A9 PCT/US2009/049421 US2009049421W WO2010003002A9 WO 2010003002 A9 WO2010003002 A9 WO 2010003002A9 US 2009049421 W US2009049421 W US 2009049421W WO 2010003002 A9 WO2010003002 A9 WO 2010003002A9
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- mice
- cell
- cxcr5
- thi
- Prior art date
Links
- 230000003325 follicular Effects 0.000 title description 13
- 210000002443 helper t lymphocyte Anatomy 0.000 title description 12
- 108010074108 interleukin-21 Proteins 0.000 claims abstract description 74
- 108090001005 Interleukin-6 Proteins 0.000 claims abstract description 42
- 108010017324 STAT3 Transcription Factor Proteins 0.000 claims abstract description 40
- 102000004495 STAT3 Transcription Factor Human genes 0.000 claims abstract description 40
- 230000004069 differentiation Effects 0.000 claims abstract description 22
- 238000000034 method Methods 0.000 claims abstract description 18
- 102100030703 Interleukin-22 Human genes 0.000 claims abstract description 9
- 239000000556 agonist Substances 0.000 claims abstract description 9
- 239000005557 antagonist Substances 0.000 claims abstract description 8
- 210000004027 cell Anatomy 0.000 claims description 193
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 45
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 14
- 239000003795 chemical substances by application Substances 0.000 claims description 11
- 230000021633 leukocyte mediated immunity Effects 0.000 claims description 11
- 201000010099 disease Diseases 0.000 claims description 8
- -1 IL- 21 Proteins 0.000 claims description 7
- 230000019491 signal transduction Effects 0.000 claims description 3
- 108010002350 Interleukin-2 Proteins 0.000 claims 1
- 241000699670 Mus sp. Species 0.000 description 101
- 102100030704 Interleukin-21 Human genes 0.000 description 69
- 102100031658 C-X-C chemokine receptor type 5 Human genes 0.000 description 49
- 101000922405 Homo sapiens C-X-C chemokine receptor type 5 Proteins 0.000 description 49
- 230000014509 gene expression Effects 0.000 description 49
- 108010045069 keyhole-limpet hemocyanin Proteins 0.000 description 40
- 102000004889 Interleukin-6 Human genes 0.000 description 38
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 29
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 29
- 108090000623 proteins and genes Proteins 0.000 description 28
- 230000003053 immunization Effects 0.000 description 24
- 238000002649 immunization Methods 0.000 description 24
- 210000003719 b-lymphocyte Anatomy 0.000 description 23
- 102000013691 Interleukin-17 Human genes 0.000 description 20
- 108050003558 Interleukin-17 Proteins 0.000 description 20
- 230000011664 signaling Effects 0.000 description 20
- 230000002950 deficient Effects 0.000 description 18
- 238000001727 in vivo Methods 0.000 description 18
- 108090000978 Interleukin-4 Proteins 0.000 description 17
- 238000011740 C57BL/6 mouse Methods 0.000 description 15
- 210000001280 germinal center Anatomy 0.000 description 15
- 230000006870 function Effects 0.000 description 14
- 238000010186 staining Methods 0.000 description 14
- 102000004127 Cytokines Human genes 0.000 description 13
- 108090000695 Cytokines Proteins 0.000 description 13
- 101001019455 Homo sapiens ICOS ligand Proteins 0.000 description 13
- 102100034980 ICOS ligand Human genes 0.000 description 13
- 238000000338 in vitro Methods 0.000 description 13
- 238000002965 ELISA Methods 0.000 description 12
- 210000000612 antigen-presenting cell Anatomy 0.000 description 12
- 210000001102 germinal center b cell Anatomy 0.000 description 12
- 230000001404 mediated effect Effects 0.000 description 11
- 241000699666 Mus <mouse, genus> Species 0.000 description 10
- 230000011712 cell development Effects 0.000 description 10
- 230000000694 effects Effects 0.000 description 10
- 238000011282 treatment Methods 0.000 description 10
- 238000011161 development Methods 0.000 description 9
- 230000018109 developmental process Effects 0.000 description 9
- 208000023275 Autoimmune disease Diseases 0.000 description 8
- 108091008731 RAR-related orphan receptors α Proteins 0.000 description 8
- 238000002474 experimental method Methods 0.000 description 8
- 102100029822 B- and T-lymphocyte attenuator Human genes 0.000 description 7
- 101710144268 B- and T-lymphocyte attenuator Proteins 0.000 description 7
- 206010028980 Neoplasm Diseases 0.000 description 7
- 108010090804 Streptavidin Proteins 0.000 description 7
- 238000004458 analytical method Methods 0.000 description 7
- 230000008901 benefit Effects 0.000 description 7
- 230000033228 biological regulation Effects 0.000 description 7
- 238000006243 chemical reaction Methods 0.000 description 7
- 230000001419 dependent effect Effects 0.000 description 7
- 239000003814 drug Substances 0.000 description 7
- 208000015181 infectious disease Diseases 0.000 description 7
- 210000000952 spleen Anatomy 0.000 description 7
- 230000003393 splenic effect Effects 0.000 description 7
- 229940124597 therapeutic agent Drugs 0.000 description 7
- 101100381525 Mus musculus Bcl6 gene Proteins 0.000 description 6
- 208000035475 disorder Diseases 0.000 description 6
- 239000012636 effector Substances 0.000 description 6
- 230000004727 humoral immunity Effects 0.000 description 6
- 238000004519 manufacturing process Methods 0.000 description 6
- 238000002493 microarray Methods 0.000 description 6
- 102000004169 proteins and genes Human genes 0.000 description 6
- 102000013968 STAT6 Transcription Factor Human genes 0.000 description 5
- 108010011005 STAT6 Transcription Factor Proteins 0.000 description 5
- 230000005875 antibody response Effects 0.000 description 5
- 239000000427 antigen Substances 0.000 description 5
- 108091007433 antigens Proteins 0.000 description 5
- 102000036639 antigens Human genes 0.000 description 5
- 230000005784 autoimmunity Effects 0.000 description 5
- 230000000903 blocking effect Effects 0.000 description 5
- 230000036039 immunity Effects 0.000 description 5
- 230000016784 immunoglobulin production Effects 0.000 description 5
- 230000001965 increasing effect Effects 0.000 description 5
- 108010074109 interleukin-22 Proteins 0.000 description 5
- 230000035755 proliferation Effects 0.000 description 5
- 238000012755 real-time RT-PCR analysis Methods 0.000 description 5
- 230000001105 regulatory effect Effects 0.000 description 5
- 230000001225 therapeutic effect Effects 0.000 description 5
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 4
- 102100037792 Interleukin-6 receptor subunit alpha Human genes 0.000 description 4
- 102000005886 STAT4 Transcription Factor Human genes 0.000 description 4
- 108010019992 STAT4 Transcription Factor Proteins 0.000 description 4
- 210000004241 Th2 cell Anatomy 0.000 description 4
- 238000000540 analysis of variance Methods 0.000 description 4
- 230000016396 cytokine production Effects 0.000 description 4
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 4
- 238000009472 formulation Methods 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- 239000000203 mixture Substances 0.000 description 4
- 230000007935 neutral effect Effects 0.000 description 4
- 238000003753 real-time PCR Methods 0.000 description 4
- 230000002441 reversible effect Effects 0.000 description 4
- 238000013207 serial dilution Methods 0.000 description 4
- 210000004989 spleen cell Anatomy 0.000 description 4
- 238000012360 testing method Methods 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- 108091008773 RAR-related orphan receptors γ Proteins 0.000 description 3
- 101150099493 STAT3 gene Proteins 0.000 description 3
- 238000000692 Student's t-test Methods 0.000 description 3
- 210000004970 cd4 cell Anatomy 0.000 description 3
- 230000003915 cell function Effects 0.000 description 3
- 239000003153 chemical reaction reagent Substances 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 150000002148 esters Chemical class 0.000 description 3
- 230000006698 induction Effects 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 108040006858 interleukin-6 receptor activity proteins Proteins 0.000 description 3
- 230000003834 intracellular effect Effects 0.000 description 3
- 210000004698 lymphocyte Anatomy 0.000 description 3
- 230000003472 neutralizing effect Effects 0.000 description 3
- 238000001543 one-way ANOVA Methods 0.000 description 3
- 108090000765 processed proteins & peptides Proteins 0.000 description 3
- 239000000651 prodrug Substances 0.000 description 3
- 229940002612 prodrug Drugs 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 150000003839 salts Chemical class 0.000 description 3
- 238000012353 t test Methods 0.000 description 3
- ZRKFYGHZFMAOKI-QMGMOQQFSA-N tgfbeta Chemical compound C([C@H](NC(=O)[C@H](C(C)C)NC(=O)CNC(=O)[C@H](CCC(O)=O)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@H]([C@@H](C)O)NC(=O)[C@H](CC(C)C)NC(=O)CNC(=O)[C@H](C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](C)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](N)CCSC)C(C)C)[C@@H](C)CC)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](C)C(=O)N[C@@H](CC=1C=CC=CC=1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](C)C(=O)N[C@@H](CC(C)C)C(=O)N1[C@@H](CCC1)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CO)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CC(C)C)C(O)=O)C1=CC=C(O)C=C1 ZRKFYGHZFMAOKI-QMGMOQQFSA-N 0.000 description 3
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 2
- 108010085238 Actins Proteins 0.000 description 2
- 102000007469 Actins Human genes 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 238000000018 DNA microarray Methods 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101000713602 Homo sapiens T-box transcription factor TBX21 Proteins 0.000 description 2
- 101000819111 Homo sapiens Trans-acting T-cell-specific transcription factor GATA-3 Proteins 0.000 description 2
- 102100037850 Interferon gamma Human genes 0.000 description 2
- 108010074328 Interferon-gamma Proteins 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 108091008778 RORγ2 Proteins 0.000 description 2
- 102100036840 T-box transcription factor TBX21 Human genes 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- 102100021386 Trans-acting T-cell-specific transcription factor GATA-3 Human genes 0.000 description 2
- 239000004480 active ingredient Substances 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 235000020958 biotin Nutrition 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 210000002798 bone marrow cell Anatomy 0.000 description 2
- 201000011510 cancer Diseases 0.000 description 2
- 230000024245 cell differentiation Effects 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 230000036755 cellular response Effects 0.000 description 2
- 230000009025 developmental regulation Effects 0.000 description 2
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 2
- 210000004602 germ cell Anatomy 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 238000002991 immunohistochemical analysis Methods 0.000 description 2
- 230000001771 impaired effect Effects 0.000 description 2
- 230000002757 inflammatory effect Effects 0.000 description 2
- 230000005764 inhibitory process Effects 0.000 description 2
- QWTDNUCVQCZILF-UHFFFAOYSA-N isopentane Chemical compound CCC(C)C QWTDNUCVQCZILF-UHFFFAOYSA-N 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 206010025135 lupus erythematosus Diseases 0.000 description 2
- 210000001165 lymph node Anatomy 0.000 description 2
- 108090000629 orphan nuclear receptors Proteins 0.000 description 2
- 102000004164 orphan nuclear receptors Human genes 0.000 description 2
- 230000008506 pathogenesis Effects 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 239000008194 pharmaceutical composition Substances 0.000 description 2
- 239000006187 pill Substances 0.000 description 2
- 239000002464 receptor antagonist Substances 0.000 description 2
- 229940044551 receptor antagonist Drugs 0.000 description 2
- 230000028327 secretion Effects 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 239000012453 solvate Substances 0.000 description 2
- 210000004988 splenocyte Anatomy 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 210000001519 tissue Anatomy 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 1
- 108090000672 Annexin A5 Proteins 0.000 description 1
- 102000004121 Annexin A5 Human genes 0.000 description 1
- 208000033241 Autosomal dominant hyper-IgE syndrome Diseases 0.000 description 1
- 102100021631 B-cell lymphoma 6 protein Human genes 0.000 description 1
- 238000011725 BALB/c mouse Methods 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 102100036301 C-C chemokine receptor type 7 Human genes 0.000 description 1
- 102100032912 CD44 antigen Human genes 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 102000003712 Complement factor B Human genes 0.000 description 1
- 108090000056 Complement factor B Proteins 0.000 description 1
- 230000007067 DNA methylation Effects 0.000 description 1
- 108010040476 FITC-annexin A5 Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 101000971234 Homo sapiens B-cell lymphoma 6 protein Proteins 0.000 description 1
- 101000716065 Homo sapiens C-C chemokine receptor type 7 Proteins 0.000 description 1
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 description 1
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 1
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 1
- 101000599048 Homo sapiens Interleukin-6 receptor subunit alpha Proteins 0.000 description 1
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 1
- 206010020772 Hypertension Diseases 0.000 description 1
- 101150054533 IL21 gene Proteins 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 208000029462 Immunodeficiency disease Diseases 0.000 description 1
- 108060003951 Immunoglobulin Proteins 0.000 description 1
- 102000053646 Inducible T-Cell Co-Stimulator Human genes 0.000 description 1
- 108700013161 Inducible T-Cell Co-Stimulator Proteins 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- 101100179071 Mus musculus Icoslg gene Proteins 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 208000030852 Parasitic disease Diseases 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 102000034442 RING-type E3 ubiquitin transferases Human genes 0.000 description 1
- 108030001238 RING-type E3 ubiquitin transferases Proteins 0.000 description 1
- 239000013614 RNA sample Substances 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 238000012952 Resampling Methods 0.000 description 1
- 102100038043 Roquin-1 Human genes 0.000 description 1
- 101710168637 Roquin-1 Proteins 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000006275 Ubiquitin-Protein Ligases Human genes 0.000 description 1
- 108010083111 Ubiquitin-Protein Ligases Proteins 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 229940030600 antihypertensive agent Drugs 0.000 description 1
- 239000002220 antihypertensive agent Substances 0.000 description 1
- 238000003782 apoptosis assay Methods 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 238000010804 cDNA synthesis Methods 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000009134 cell regulation Effects 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 208000037976 chronic inflammation Diseases 0.000 description 1
- 230000006020 chronic inflammation Effects 0.000 description 1
- 230000004186 co-expression Effects 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 108091008034 costimulatory receptors Proteins 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 230000007812 deficiency Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- AFABGHUZZDYHJO-UHFFFAOYSA-N dimethyl butane Natural products CCCC(C)C AFABGHUZZDYHJO-UHFFFAOYSA-N 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 238000012137 double-staining Methods 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 230000002500 effect on skin Effects 0.000 description 1
- 210000003162 effector t lymphocyte Anatomy 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 201000002491 encephalomyelitis Diseases 0.000 description 1
- 208000010227 enterocolitis Diseases 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 230000012178 germinal center formation Effects 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 230000008348 humoral response Effects 0.000 description 1
- 208000014796 hyper-IgE recurrent infection syndrome 1 Diseases 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 230000007813 immunodeficiency Effects 0.000 description 1
- 102000018358 immunoglobulin Human genes 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 239000003112 inhibitor Substances 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 108040002099 interleukin-21 receptor activity proteins Proteins 0.000 description 1
- 102000008640 interleukin-21 receptor activity proteins Human genes 0.000 description 1
- 238000010212 intracellular staining Methods 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- PGHMRUGBZOYCAA-ADZNBVRBSA-N ionomycin Chemical compound O1[C@H](C[C@H](O)[C@H](C)[C@H](O)[C@H](C)/C=C/C[C@@H](C)C[C@@H](C)C(/O)=C/C(=O)[C@@H](C)C[C@@H](C)C[C@@H](CCC(O)=O)C)CC[C@@]1(C)[C@@H]1O[C@](C)([C@@H](C)O)CC1 PGHMRUGBZOYCAA-ADZNBVRBSA-N 0.000 description 1
- PGHMRUGBZOYCAA-UHFFFAOYSA-N ionomycin Natural products O1C(CC(O)C(C)C(O)C(C)C=CCC(C)CC(C)C(O)=CC(=O)C(C)CC(C)CC(CCC(O)=O)C)CCC1(C)C1OC(C)(C(C)O)CC1 PGHMRUGBZOYCAA-UHFFFAOYSA-N 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 230000007762 localization of cell Effects 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 201000001441 melanoma Diseases 0.000 description 1
- 238000010208 microarray analysis Methods 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 210000000440 neutrophil Anatomy 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 230000003076 paracrine Effects 0.000 description 1
- 244000045947 parasite Species 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 230000005522 programmed cell death Effects 0.000 description 1
- 238000011321 prophylaxis Methods 0.000 description 1
- 238000010807 real-time PCR kit Methods 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000009711 regulatory function Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 238000012552 review Methods 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 238000011285 therapeutic regimen Methods 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000001131 transforming effect Effects 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/1703—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- A61K38/1709—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/177—Receptors; Cell surface antigens; Cell surface determinants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/19—Cytokines; Lymphokines; Interferons
- A61K38/20—Interleukins [IL]
- A61K38/204—IL-6
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/461—Cellular immunotherapy characterised by the cell type used
- A61K39/4611—T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/46—Cellular immunotherapy
- A61K39/464—Cellular immunotherapy characterised by the antigen targeted or presented
- A61K39/4643—Vertebrate antigens
- A61K39/4644—Cancer antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0636—T lymphocytes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
Definitions
- Th cells A fundamental function of Th cells is to provide "help" to B cells and regulate their proliferation and immunoglobulin class-switching, especially in the germinal center structures.
- ThI and Th2 cells have been shown to regulate B cell responses.
- IFN ⁇ regulates IgG2a production while IL-4 is critical in IgE class-switching.
- T fh an additional Th subset called follicular helper T (T fh ) cells are present in germinal centers and are characterized by their expression of chemokine (C-X-C motif) receptor 5 (CXCR5).
- C-X-C motif chemokine receptor 5
- T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78.
- IL-21 is expressed by both ThI 7 and T f1 , cells SUMMARY OF THE INVENTION
- T follicular helper cells provides a helper function to B cells. They stimulate the differentiation of B cells into antibody-forming cells.
- T f1 generation is regulated by IL-6, IL-21 and STAT3.
- Tfh cells have a distinct gene expression profile from THl, TH2 and TH 17 lineages as demonstrated by microarray analysis (The microarray data have been deposited into NCBI GEO with the accession number GSEl 1924).
- T fh cells can contribute to pathogenesis antibody-mediated autoimmune diseases (Systemic lupus erythematosus (SLE)). In this case targeting of specific T f1 , factors will help to prevent this autoimmune disease.
- T f11 cells can be beneficial during parasite, viral and bacterial infection. During infection antibody is required to confer protection against infectious diseases by neutralizing pathogenic factors on the microorganisms (antibody- mediated protection). In this case T fh cells will help to clear infection.
- FIGS 1A-1D show that T fh cells express distinct genes from ThI, Th2 and Thl7 cells.
- FIGS. 2A-2D depict the generation of T f ] 1 cells as independent of ThI and Th2 lineages.
- Figures 3A-3E show B7H expressed on B cells as required for generation of T f ] 1 cells.
- Figures 4A-4D show IL-21 is necessary for T f ] 1 cell development.
- Figures 5A-5C show the generation of T fh cells requires IL-6 and STAT3.
- Figures 6A-6C show the generation of Ta cells is independent of THl 7 lineage
- Figures 7A-7G show IL-21, in the absence of IL-4, IFN ⁇ and TGF-b signaling, generates Ta cells.
- Figure 8 depicts follicular helper T cells co-express CXCR5 and BTLA.
- Figures 9 A and 9B show FDR estimated from ANOVA tests using resampled samples.
- Figure 10 shows antigen-specific cytokine production by Tn 1 cells.
- Figures 1 IA-I ID shows T fl , cell generation is independent of THl and TH2 cells.
- Figure 12 shows regulation of antibody responses by STAT3.
- Figures 13 A and 13B show normal germinal center reaction in IL-17 KO, IL- 17F
- Figure 14 depicts a schematic demonstration that development of T ⁇ cells, independent of THl, TH2 or TH 17 lineages, is regulated by IL-6 and IL-21.
- Na ⁇ ve CD4 + helper T (Th) cells upon encountering their cognate antigens presented on professional antigen-presenting cells (APC), differentiate into effector cells that are characterized by their distinct cytokine production profiles and immune regulatory functions.
- APC professional antigen-presenting cells
- ThI 7 a third subset of Th cells, has been recently identified.
- ThI 7 cells produce IL-17, IL-17F and IL-22 and regulate inflammatory responses by tissue cells.
- ThI 7 differentiation is initiated by transforming growth factor ⁇ (TGF ⁇ ) and IL-6, possibly via regulating the chromatin remodeling of the III 7-1117 f ' locus.
- TGF ⁇ transforming growth factor ⁇
- Bettelli, E., et al. Reciprocal Developmental Pathways for the Generation of Pathogenic Effector TH 17 and Regulatory T Cells, Nature , 2006, 441 :235-238; Mangan, P.R., et al., Transforming Growth Factor-beta Induces Development of the T(H)17 Lineage, Nature, 2006, 441 :231 -234; Veldhoen, M., et al., TGFbeta in the Context of an Inflammatory Cytokine Milieu Supports de novo Differentiation of IL- 17 -producing T Cells, Immunity, 2006, 24:179-189; Akimzhanov, A.M., et al., Chromatin Remodeling of Interleukin
- IL-21 is reportedly an autocrine factor induced by IL-6 to regulate ThI 7 differentiation.
- Korn, T., et al., IL-21 Initiates an Alternative Pathway to Induce Proinflammatory TH 17 Cells, Nature, 2007, 448:484-487; Yang, X.O., et al., STAT3 Regulates Cytokine-mediated Generation of Inflammatory Helper T Cells, J Biol Chem, 2007, 282:9358-9363; Nurieva, R., et al., Essential Autocrine Regulation by IL-21 in the Generation of Inflammatory T Cells, Nature, 2007, 448:480-483; Zhou, L., et al., IL-6 Programs TH-17 Cell Differentiation by Promoting Sequential Engagement of the IL-21 and IL-23 Pathways, Nat Immunol, 2007, 8:967-974.
- TGF ⁇ signaling mediates ThI 7 differentiation in vivo.
- Bettelli, E., et al. Reciprocal Developmental Pathways for the Generation of Pathogenic Effector THl 7 and Regulatory T Cells, Nature , 2006, 441 :235-238; Mangan, P.R., et al., Transforming Growth Factor-beta Induces Development of the T(H)17 Lineage, Nature, 2006, 441 :231-234; Veldhoen, M., et al., Signals Mediated by Transforming Growth Factor-beta Initiate Autoimmune Encephalomyelitis, but Chronic Inflammation is Needed to Sustain Disease, Nat Immunol, 2006, 7:1151-1156.
- Thl7 cell development is dependent on STAT3, which functions to upregulate the expression of two Thl7-specific orphan nuclear receptors ROR ⁇ t and ROR ⁇ and ultimately determines ThI 7 terminal differentiation.
- STAT3 functions to upregulate the expression of two Thl7-specific orphan nuclear receptors ROR ⁇ t and ROR ⁇ and ultimately determines ThI 7 terminal differentiation.
- activated T cells may transiently express CXCR5, the T fh cell subset demonstrates more stable expression of this chemokine receptor. These cells also regulate humoral immunity, especially germinal center reactions. Consistent with this notion, CXCR5 has been shown to be important for proper T and B cell localization in immune responses and antibody production.
- IL-21 in the CXCR5+ Follicular B Helper T Cell Maintenance In Vivo, J Immunol, 2005, 175:2340-2348.
- IL-21 is also expressed in T f1 , cells and may serve as an important regulator of humoral responses by Tn, cells.
- IL-21 directly regulates B cell proliferation and class-switching.
- IL-21 R deficiency results in defective antibody responses and impaired germinal center formation. Spolski, R., et al., Interleukin-21 : Basic Biology and Implications for Cancer and Autoimmunity, Annu Rev Immunol, 2008, 26.
- sanroque mice which have a mutation in a RING-type E3 ubiquitin ligase, Roquin, developed spontaneous autoantibody production and lupus-like autoimmunity, associated with greatly increased numbers of CXCR5 + CD4 + T cells and enhanced expression of IL-21 and ICOS. Vinuesa, C. G., et al., A RING-type Ubiquitin Ligase Family Member Required to Repress Follicular Helper T Cells and Autoimmunity, Nature, 2005, 435:452-458.
- T f1 cells have a divergent gene expression profile from ThI, Th2 and ThI 7 cells, and develop in vivo independent of these lineages.
- IL-21, IL-6 and STAT3 are critical in the generation of T f1 , cells.
- T cells activated in vitro in the presence of IL-21 but without TGF ⁇ signaling preferentially acquired T f1 , gene expression and functioned to promote humoral immunity in vivo.
- T f1 cells represent a distinct Th lineage and suggest a reciprocal relationship between the Ta and ThI 7 lineages.
- Ta cells are strongly implicated in humoral immunity, including antibody production and antibody-mediated autoimmunity, and germinal center reactions.
- T f1 cells are distinct from ThI, Th2 or Th 17 cells in their gene expression and developmental regulation.
- Generation of T fh cells requires IL-21, IL-6 and STAT3.
- IL- 21, IL-6 and STAT3, their signaling, expression and activity are targets in modulating Tf 1 , cell- mediated immune response.
- Methods of treating disease therefore, include administering an agent that modulates IL-6, IL-21, and/or STAT3 signaling, expression or activity in an amount effective to modulate the differentiation of T f1 , cells.
- Useful agents may modulate the production, growth or activity of Tn, cells.
- the agent may also modulate the expression of genes expressed by T fh cells and/or be used in combination with other agents.
- C et al.
- Regulation of Immune and Autoimmune Responses by ICOS J Autoimmun, 2003, 21 :255-260.
- This costimulatory pathway is also important in generation OfT f1 , cells in mouse.
- Akiba, H., et al. The Role of ICOS in the CXCR5+ Follicular B Helper T Cell Maintenance In Vivo, J Immunol, 2005, 175:2340-2348.
- impaired negative regulation of ICOS by Roquin E3 ubiquitin ligase leads to increased numbers of CXCR5 + T f1 , cells and IL-21 hyperproduction.
- Vinuesa C.G., et al., A RING-type Ubiquitin Ligase Family Member Required to Repress Follicular Helper T Cells and Autoimmunity, Nature, 2005a, 435:452-458; Yu, D., et al., Roquin Represses Autoimmunity by Limiting Inducible T-cell Co-stimulator Messenger RNA, Nature, 2007, 450:299-303.
- ICOS-B7h interaction is necessary for IL-21 expression by T cells.
- ICOS regulates Tn, cells through production of IL-21.
- IL-21 regulation by calcium signaling and NFAT factors has been shown. Kim, H.P., et al., Calcium-dependent Activation of Interleukin-21 Gene Expression in T Cells, J Biol Chem, 2005, 280:25291-25297.
- ICOS, together with TcR and CD28 has been shown to increase the expression of NFATcI through a PI-3 kinase- Itk-calcium pathway.
- ICOS can act through NFATcI to regulate IL-21 expression. Since IL-21 regulates ThI 7 differentiation, an IL-21 defect can account for the impairment in IL- 17 expression in the ICOS-deficient animals.
- T fh cells are distinct from ThI and Th2 cells. Chtanova, T., et al., T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78; Kim, CH. , et al., Unique Gene Expression Program of Human Germinal Center T Helper Cells, Blood, 2004, 104:1952-1960. The data reported here also reveals that T f1 , cells do not produce ThI or Th2 cytokines.
- T ⁇ cells share common regulators with ThI 7 cells. Both subsets express IL-21 and their development depends similarly on IL-6, IL-21 and STAT3.
- T f h cells differ from ThI 7 cells in the following aspects. First, they are distinct in their gene expression profiles. Second, T fh cells do not produce IL-17, IL-17F or IL-22. Additionally, T fh cell development does not require ROR ⁇ or ROR ⁇ t. Thus, we conclude that T f1 , cells develop independent of the Th 17 lineage.
- ThI 7 development in mouse is not only mediated by the IL-6-IL-21 axis, but also by TGF ⁇ .
- IL-21 can be induced in T cells independent of TGF ⁇ signaling.
- T cells activated in the presence of IL-21 but in the absence of IL-4, IFN ⁇ and TGF ⁇ signaling produced IL-21 but not IL-4, IFN ⁇ , IL-17, IL-17F or IL-22.
- these cells acquired expression of CXCR5, Bcl-6, IL-6R and IL-6st, genes expressed by in vivo generated T fh cells, suggesting that T fh cells may be generated in vitro under the above condition.
- T fh -Hke cells generated in vitro preferentially expressed CXCR5 in vivo and functioned to promote humoral immunity, similar to in vivo-generated T f ) 1 cells.
- a novel approach and methods for generating T f h cells in vitro is provided herein.
- TGF ⁇ signaling is not essential for T f1 , cells, although required for
- T fh cell development is independent of ThI, Th2 and Thl7 cells, and IL-21 serves as critical factor for generation of this lineage.
- T f1 , cells also enhanced the T f11 cell generation in recipient mice, suggesting that IL-21 may function in a paracrine fashion to regulate T fh cell development.
- T fh cells are distinct in their gene expression and immune function and develop via a pathway that is dependent on IL-21 or IL-6 but independent of ThI, Th2 or ThI 7 lineages ( Figure 14).
- mice defective in T fh cells there were still detectable amounts of antigen-specific antibodies, suggesting that other Th subsets may independently regulate the humoral immunity.
- This knowledge may be applied to treat disease, e.g., antibody-mediated autoimmune diseases or other diseases associated with T f1 , cell-mediated immune response.
- T f1 , cell function and/or generation may be modulated by use of an agent such as an antagonist of one or more of IL-6, IL-21, and STAT3, and/or an agonist of one or more of IL-6, IL-21, and STAT3.
- an agent such as an antagonist of one or more of IL-6, IL-21, and STAT3, and/or an agonist of one or more of IL-6, IL-21, and STAT3.
- Use of such an agent in an amount effective to inhibit or induce the differentiation of Ta cells, and/or to modulate the expression of genes expressed by Ta cells can affect T f1 , cell-mediated immune response.
- antibody-mediated autoimmune diseases may be treated by administration of a antagonist of one or more of IL-6, IL-21, and STAT3 in an amount effective to inhibit the differentiation of T f1 , cells and/or modulate gene expression by T f1 , cells.
- an agonist of one or more of IL-6, IL- 21, and STAT3 may be used to increase antibody production against infections and cancers by administering the agonist in an amount effective to induce the differentiation of T f1 , cells and/or modulate the gene expression by T f1 , cells.
- T f1 Differentiation Of T f1 , cells can be modulated via one or more of the IL-6, IL-21, and STAT3 signaling pathways.
- Antagonists that may interrupt these pathways may be small molecule inhibitors, antibodies and the like.
- IL-6, IL-21, and/or STAT3 antagonists which are suitable agents to modulate T fh cell mediated immune response include molecules that bind to any of these signalling factors and/or their corresponding receptors. Examples of antagonists that may be suitable include soluble receptor antagonists and antibodies that prevent binding of IL-6, IL-21, and/or STAT3 to its receptor.
- inhibition of the activity of any of IL-6, IL-21, and STAT3 may be accomplished by binding the corresponding receptor with a therapeutically effective amount of a receptor antagonist such as a blocking antibody.
- a receptor antagonist such as a blocking antibody.
- Functions of T fh cells which may be reduced by interrupting a IL-6, IL-21 , and STAT3 signaling pathway include, for example, expansion of Tn, cells and production of factors produced by T f11 cells.
- IL-6, IL-21, and/or STAT3 may be useful for the treatment of disorders or a wide variety of conditions where decreased Ta cell mediated immune response is useful.
- Disorders or conditions advantageously treated by these methods include autoimmune diseases, which involve Ta cell-mediated immune response.
- disorders or conditions which may be treated by decreasing Ta cell-mediated immune response include, antibody-mediated auto immune diseases such as lupus.
- Ta cell -mediated immune response through induction of IL-6, IL-21, and/or STAT3 activity and/or expression include administering a therapeutically effective amount of IL-6, IL-21, and/or STAT3 or an agonist for IL-6, IL-21, and/or STAT3 to an individual in need thereof.
- the IL-6, IL-21, and/or STAT3, or agonist thereof increases the generation, differentiation and function of T ⁇ cells in the individual, thereby increasing T ⁇ cell-mediated response to infection or, in some cases, tumors.
- Examples of the types of infections or tumors that may benefit from increased Ta, cell mediated immune response include immunity against excellular bacteria, fungus, viruses, and tumors including melanoma.
- IL-6, IL-21, and/or STAT3 agonists or similar include mimicry fragments, small molecules, and molecules and proteins of similar function.
- the phrase "therapeutically effective" is intended to qualify the amount of active ingredients used in the treatment of a disease or disorder. This amount will achieve the goal of reducing or eliminating the said disease or disorder.
- patient means all mammals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, pigs, and rabbits. Preferably, the patient is a human.
- the pharmaceutical formulation may include the molecule or a pharmaceutically acceptable salt, ester, prodrug or solvate thereof, where appropriate, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients.
- the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences.
- formulations of use molecules include those suitable for oral, parenteral
- formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods may include the step of bringing into association the molecule or a pharmaceutically acceptable salt, ester, prodrug or solvate thereof ("active ingredient") with the carrier which constitutes one or more accessory ingredients.
- At least one molecule or a pharmaceutically acceptable salt, ester, or prodrug thereof in combination with another therapeutic agent.
- another therapeutic agent such as a pharmaceutically acceptable salt, ester, or prodrug thereof.
- the therapeutic effectiveness of one molecule may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced).
- the benefit of experienced by a patient may be increased by administering one molecule as described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
- Multiple therapeutic agents may be administered in any order or even simultaneously. If simultaneously, the multiple therapeutic agents may be provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). One of the therapeutic agents may be given in multiple doses, or both may be given as multiple doses. If not simultaneous, the timing between the multiple doses may be any duration of time ranging from a few minutes to four weeks.
- T FH T follicular helper cells
- Tfh T follicular helper cells
- T FH cell generation is regulated by B7h expressed on B cells and, similar to TH 17 cell development, is dependent on IL-21, IL-6 and STAT3.
- differentiation of TFH cells unlike THl 7 cells, does not require TGF ⁇ signaling or TH17-specific orphan nuclear receptors ROR ⁇ and ROR ⁇ in vivo.
- na ⁇ ve T cells activated in vitro in the presence of IL-21 but not TGF ⁇ signaling preferentially acquire TFH gene expression and function to promote germinal center reactions in vivo.
- TFH is thus demonstrated as a distinct lineage of effector TH differentiation, and IL-6, IL-21, and STAT3 are shown to be important regulators of these cells.
- Antagonists of these factors can be used in the treatment of autoimmune diseases, while agonists of these factors can be used to promote generation and expansion of TFH cells, which can help antibody production against infections and cancers.
- mice IL-4-, IFN ⁇ - and IL-6-def ⁇ cient mice on C57BL/6 background and STAT6 and STAT4-deficient mice on BALB/c background were purchased from Jackson Laboratories and C57BL/6, B6.SJL (CD45.1) and BALB/c mice were used as controls.
- Rord t/st , Rord l/sl Ror ⁇ ' ⁇ and wild-type bone marrow chimeras were generated as described by Yang et al.
- ICOSL-BKO mice were created by breeding ICOSL flox mice with CD19-cre mice.
- Nurieva, R.I., et al., B7h is Required for T Cell Activation, Differentiation, and Effector Function, Proc Natl Acad Sci U S A, 2003, 100:14163-14168; Rickert, R.C., et al., Impairment of T-cell-dependent B-cell Responses and B-I Cell Development in CD 19 -deficient Mice, Nature, 1995, 376:352-355.
- IL- 17- and IL-17F- deficient mice were recently generated in the lab. Yang, X., et al., Regulation of Inflammatory Responses by 1L-17F, J Exp Med In press, 2008. Mice were housed in the SPF animal facility at M. D. Anderson Cancer Center and the animal experiments were performed at the age of 6-10 weeks using protocols approved by Institutional Animal Care and Use Committee.
- IA-B was performed as previously described. Chung, Y., et al., Expression and Regulation of IL-22 in the IL- 17 -Producing CD4+ T Lymphocytes, Cell Res, 2006, 16:902-907.
- the cytokine stimuli for Thl7 differentiation were 100 ng/ml of IL-21, 5 ng/ml of TGF ⁇ , and 10 ⁇ g/ml of anti-IL-4 and 10 ⁇ g/ml of anti-IFN ⁇ , and for generation of T fh cells were 50 ng/ml of IL-21, 10 ⁇ g/ml anti-IFN ⁇ , 10 ⁇ g/ml anti-IL-4 and 20 ⁇ g/ml TGF- ⁇ (lDl l) neutralizing antibodies.
- IL-4, IL-6, IL- 12 and TGF ⁇ were purchased from Peprotech.
- IL-21, IL-23 and TGF- ⁇ (IDl 1) neutralizing antibodies were purchased from R&D.
- plate- bound anti-CD3 5 ⁇ g/ml
- KLH Keyhole Limpet Hemocyanin
- the T f1 , cell induction was determined by staining with PerCP-labeled anti-CD4 mAb (Pharmingen) and biotinylated anti- CXCR5 mAb (Pharmingen), followed by APC-labeled streptavidin (Jackson ImmunoResearch Laboratories, Inc.).
- sera from immunized mice were collected, and antigen- specific IgM, and IgG antibodies were measured by using ELISA. Briefly, serum samples were added in a 3 -fold serial dilution onto plates precoated with 10 ⁇ g/ml KLH or Ova protein.
- Antigen-specific antibodies were detected with biotinylated goat antimouse IgM or rat anti- mouse IgG antibodies (Southern Biotechnology Associates).
- spleen cells from KLH-immunized mice were stimulated in 96-well plates as triplicates with or without KLH. Effector cytokines (IFN- ⁇ and IL-21) were analyzed 4 days later by ELISA (Pharmingen).
- IFN- ⁇ and IL-21 Effector cytokines
- Statistical analysis of microarray data The DNA microarray analysis was carried out at the Institute for Systems Biology microarray core facility using Affymetrix Mouse 430 2.0 chips. The total RNA samples were labeled according to manufacturer's instruction using One-Cycle Target Labeling method, which consists of oligo-dT primed cDNA synthesis followed by in vitro transcription that incorporates biotinylated nucleotides. The microarray data were normalized using GCRMA.
- CD4 + T cells from OT-II mice (CD45.2) were intravenously transferred into C57BL/6 (CD45.1 + ) mice (3x10 6 cells/mouse) (3 groups; 3 mice per group). 2 groups of recipient mice were immunized subcutaneously with 100 ⁇ g Ova protein emulsified in CFA and treated with a 300 ⁇ g of control rat Ig or anti-IFN ⁇ and anti-IL-4 mAbs at the time of immunization (day 0) and on days 2 and 4.
- C57BL/6 mice that did not receive T cells was used as a control (No transfer).
- CXCR5 + CD44 hl cells were sorted from B6.SJL (CD45.1) mice immunized with KLH. These cells were transferred into C57BL/6 (CD45.2 + ) mice (5x10 6 cells/mouse) (3 mice per group). Second group did not receive cells. All mice were immunized subcutaneously with 1000 ⁇ g KLH.
- lymphoid cells from the draining lymph nodes of the recipient mice were isolated and stained with FITC-labeled anti-CD45.1 mAb and PerCP-labeled anti-CD4 mAb plus biotinylated anti- CXCR5, followed by APC-labeled streptavidin, or stained with FITC-labeled PNA and PerCP- labeled anti-B220.
- Bcl6 forward CACACCCGTCCATCATTGAA, reverse: TGTCCTCACGGTGCCTTTTT
- IL6R forward GGTGGCCCAGTACCAATGC, reverse: GGACCTGGACCACGTGCT
- CXCR5 forward ACTCCTTACCACAGTGC ACCTT, reverse: GGAAACGGGAGGTGAACCA
- IL-6st forward ATT TGT GTG CTG AAG GAG GC, reverse: AAA GGA CAG GAT GTT GCA GG.
- T ⁇ As a first step toward understanding T ⁇ , cell regulation, we compared the gene expression profiles of ThI, Th2 and ThI 7 cells differentiated in vitro with in- vivo generated T fh cells. Taking advantage of the co-expression of B and T lymphocyte attenuator (BTLA) by CXCR5 + Ta cells (Figure 8), CD4 + CD44 hi CXCR5 + BTLA + were FACS sorted from splenocytes of C57BL/6 mice seven days after immunization with keyhole limpet hemocyanin (KLH). These cells as well as ThI, Th2 and ThI 7 cells were restimulated with anti-CD3 for 4 hours and subject to gene profiling analysis in duplicates using Affimetrix gene chips.
- KLH keyhole limpet hemocyanin
- the microarray data were normalized using GCRMA and the genes whose expression was changed across the ThI, Th2, Th 17 and T fh cells were then selected using a False Discovery Rate (FDR) estimation method. Then, the expression levels of 8350 probe-sets showing differential expressions among the four types of cells were used for hierarchical clustering, which revealed that T fh cells have a very distinct gene expression profile ( Figure IA, Figure 9A).
- FDR False Discovery Rate
- T f1 cells did not express the typical markers for ThI (IFN ⁇ and T-bet) or Th2 (IL-4 and GATA3) cells ( Figure IB), consistent with a previous report on human T f ] 1 cells.
- T f11 cells shared IL-21 expression with ThI 7 cells, they did not express IL-17, IL-17F, IL-22 or ROR ⁇ t (Figure IB). Instead, similar to their human counterparts, mouse Ta cells express mRNAs for CXCR5 as well as Bcl-6 ( Figure IB). Chtanova, T., et al., T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78; Kim, CH. , et al., Unique Gene Expression Program of Human Germinal Center T Helper Cells, Blood, 2004, 104:1952-1960.
- T f11 cells preferentially expressed mRNAs for IL-6R and IL-6st (gpl30) and also upregulated the expression of IL-21R (Figure IB), suggesting possible regulation Of T f11 cells by IL-6 and IL-21.
- Figure IB the expression of IL-21R
- Ta cells after they were activated ex vivo with anti-CD3 and anti-CD28 for 24 hours. High expressions of IL-21 but not IL-4, IL-10, IFN ⁇ or IL- 17 were observed in T fh cells ( Figure 1C).
- purified Ta cells were activated with KLH and irradiated splenic APC. Consistent with above results, Ta cells preferentially produced IL-21, but not ThI, Th2 and Th 17 cytokines ( Figure 10).
- intracellular analysis on CXCR5 + and CXCR5 " cells following PMA and ionomycin restimulation also revealed that T fh cells did not express IFN ⁇ or IL- 17 ( Figure ID).
- Ta cells from IL-4- or IFN ⁇ -deficient mice produced similar amount of IL-21, but did not express IL-4, IL-10, IFN ⁇ or IL-17 (Supplementary Figure 4D), supporting that they were Ta cells. Thus, we conclude that CXCR5 + CD4 + Ta cells develop independent of the ThI and Th2 lineages.
- Inducible costimulator is the third member of the CD28 family with an important role in regulation of T-dependent antibody responses and germinal center reactions.
- ICOS was previously shown to be expressed at high levels on human tonsillular CXCR5+ T cells within the light zone of germinal centers and efficiently supported the immunoglobulin production.
- T fh cells are regarded as regulators of the germinal center reaction by providing help to activated B cells that also express CXCR5. Because B cells constitutively express ICOSL, we asked whether the generation of Ta cells may require B cell help via engagement of ICOS receptor on T cells. We thus bred mice carrying the ICOSL conditional flox (f) allele with CD19-cre mice. Nurieva, R.
- EXAMPLE 4 IL-21 and IL-6 are required for generation of T fh cells, which is dependent on STAT3
- IL-21 has been recently shown to be induced by IL-6 and to autoregulate its own expression during ThI 7 differentiation.
- Nurieva, R., et al. Essential Autocrine Regulation by IL-21 in the Generation of Inflammatory T Cells, Nature, 2007, 448:480-483.
- TQ 1 T cells produced a greater amount of IL-21 compared to ThI and Th2 subsets, and induced the differentiation of autologous B cells into Ig-secreted plasma cells through IL-21.
- T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78; Bryant, V.L., et al., Cytokine-mediated Regulation of Human B Cell Differentiation Into Ig-secreting Cells: Predominant Role of IL-21 Produced by CXCR5+ T Follicular Helper Cells, J Immunol, 2007, 179:8180-8190. Becouse IL-21 is also expressed in Ta cells, we assessed if IL-21 is important for T f1 , cell generation.
- I121 +/+ ' +/ ⁇ and ⁇ ' ⁇ mice were immunized with KLH and splenic T f11 cells were analyzed in these mice.
- Il21 +/ ⁇ mice exhibited reduced number of T fh cells, which was further reduced in I121 '1' mice ( Figure 4A).
- PNA + germinal center B cells were also greatly reduced in 1121 ⁇ ' ⁇ mice ( Figure 4B-C).
- CD4 T cells from 112 f' ⁇ and ⁇ ' ⁇ mice showed normal proliferation and IFN ⁇ expression after re-stimulation with KLH ex vivo. ( Figure 5D).
- IL-21 is necessary for T f11 cell development.
- T fh cell generation is independent of ThI 7 differentiation or function
- ThI 7 differentiation requires also TGF ⁇ in addition to IL-6 or IL-21
- TGF ⁇ signaling is required for T f11 cell generation.
- C57BL/6 mice were immunized with KLH in the absence or presence of TGF ⁇ blocking antibody as previously described.
- Veldhoen, M., et al. Signals Mediated by Transforming Growth Factor-beta Initiate Autoimmune Encephalomyelitis, but Chronic Inflammation is Needed to Sustain Disease, Nat Immunol, 2006, 7:1151-1156.
- IL-6 and IL-21 are required for both T fh and Th 17 differentiation, these two subsets appear to have distinct genetic program and differ in their dependency on TGF ⁇ signaling.
- IL-21 is sufficient to drive Tfh cell development in vitro in the absence of TGF ⁇ signaling.
- Na ⁇ ve OT-II cells were activated by Ova peptide and splenic APC in the absence (neutral condition) or presence of IL-21, TGF ⁇ and antibodies to IL-4 and IFN ⁇ (Th 17 condition) or IL-21 plus antibodies to IL-4, IFN ⁇ and TGF ⁇ .
- T cells cultured under Th 17 condition highly expressed TH17-specific genes, including genes encoding IL-17, IL-17F, IL-22, ROR ⁇ and ROR ⁇ t ( Figure 7A).
- T cells treated with IL-21 in the absence of TGF ⁇ signaling upregulated genes that are specifically expressed in T ⁇ , cells, including those encoding CXCR5, Bcl-6, IL-6R and IL-6st ( Figure 7A). They also upregualted IL-21R expression but did not express Th 17 genes ( Figure 7A).
- CD4 + T cells from OT-II mice were transferred into C57BL/6 (CD45.1) mice which were subsequently divided into 2 groups (3 mice per group). Mice were immunized subcutaneously with Ova protein emulsified in CFA and treated with a 300 ⁇ g of control rat Ig or anti-IFN ⁇ and anti-IL-4 mAbs. Seven days after the immunization, experimental mice were sacrificed and splenic CD45.1 + and CD45.2 + CD4 cells were stained with biotinylated CXCR5 mAb, followed by APC-labeled streptavidin. Numbers in dot plot quadrants represent the percentages.
- CD44hiCXCR5 + and CD44MCXCR5 " cells from immunized mice were purified and real-time RT-PCR analysis of Ta specific genes were performed.
- B-C IU ' ' ' , Ifn ⁇ 1' (B), Stat6 ⁇ ' ⁇ (C) and Stat4 ⁇ A (D) and their appropriate controls (WT, 3 mice per group) were immunized with KLH emulsified in CFA. Seven days after the immunization, experimental mice were sacrificed and the germinal center B cells were determined by staining with FITC-labeled PNA and PerCP-labeled B220 mAb.
- T ft cells were analyzed by staining with PerCP-labeled CD4 mAb and biotinylated CXCR5 mAb, followed by APC-labeled streptavidin. Numbers in dot plot quadrants represent the percentages. The experiments were repeated three times with consistent results.
- P values were calculated with the t-test by comparing the CXCR5 + cells and B220 + PNA + cells between wild-type and ICOSL deficient mice and are indicated as followed: * P ⁇ 0.005; #, P ⁇ 0.001. Numbers in dot plot quadrants represent the percentages.
- B Splenic B220 + B cells from ICOSL germline deficient mice (Icosl ' ' ' ), B cell specific ICOSL deficient mice (ICOSL-BKO, cre+) and the ere- controls were analyzed for ICOSL expression.
- C-E Wild-type (WT) and B cell specific ICOSL deficient mice (3 mice per group) were immunized with KLH in CFA.
- mice Seven days after the immunization, experimental mice were sacrificed and analyzed as in A . P values were calculated with the t-test by comparing the CXCR5 + cells and B220 + PNA + cells between wild-type and B cell specific B7h deficient mice and are indicated as followed: ** P ⁇ 0.001 ; ##, P ⁇ 0.001.
- C KLH specific antibodies (IgM and IgG) were measured in the sera by ELISA. The sera from WT and B cell specific ICOSL deficient mice were subject to a 3-fold serial dilution, and the concentrations of KLH-specific IgM and IgG were analyzed by ELISA and averaged for each group.
- E KLH specific antibodies
- GC in the spleens of KLH -immunized WT and B cell specific ICOSL deficient mice were identified by PNA staining (brown).
- T and B cells were identified by staining with anti-CD4 (red) and anti- B220 (blue). The data represent at least three independent experiments with consistent results.
- I121 +/+ , Il21 +/ ⁇ and 1121 '1' mice (3 mice per group) were immunized subcutaneously with KLH emulsified in CFA. Seven days after the immunization, experimental mice were sacrificed and Ta cells (A) and the germinal center B cells (B) were analyzed. Numbers in dot plot quadrants represent the percentages. Germinal centers were determined by immunohistochemical analysis (C). Spleen cells from immunized mice were stimulated in 96-well plates as triplicates with the indicated concentration of KLH peptide. Proliferation was assayed after 3 days of treatment by adding [ 3 H]thymidine to the culture for the last 8 h. IFN- ⁇ was measured after 4 days of treatment. The experiments were repeated twice with consistent results.
- I16 'A (A) or T-cell specific Stat3 'A mice (C) and their appropriate controls (WT, 3 mice per group) were immunized subcutaneously with KLH emulsified in CFA. Seven days after the immunization, experimental mice were sacrificed and Ta cells and the germinal center B cells were analyzed. Numbers in dot plot quadrants represent the percentages.
- B C57BL/6 mice were immunized with KLH in CFA. Seven days later, CD4 + CD44 hi CXCR5 + and CD4 + CD44 hi CXCR5 " cells were sorted and restimulated with anti- CD3 and anti-CD28 with or without IL-6 or IL-21 for 48 hours.
- mice As shown in Figure 6, Ragl '1' reconstituted with WT and Rord tl$t /Ror ⁇ ⁇ ' ⁇ bone marrow cells (A) or IUT' ' and Ill7f ⁇ mice and their controls (C) (3 mice per group) were immunized subcutaneously with KLH in CFA. Seven days after the immunization, experimental mice were sacrificed and the Tn 1 cells were determined. Numbers in dot plot quadrants represent the percentages. (B). C57BL/6 mice were immunized with KLH emulsified in CFA with 100 ⁇ g of isotype control antibodies or TGF ⁇ blocking antibodies (3 mice per group).
- mice Seven days later, experimental mice were sacrificed and splenic Ta cells and germinal center B cells were analyzed. Splenocytes were restimulated with KLH for overnight and the production of IL- 17 and IFN ⁇ was analyzed in CD4 + gate by intracellular cytokine staining. The results represent one of three individuals with similar results.
- FIG. 7 FACS-sorted CD62 hi CD44 Io CD25 neg CD4 + T cells from CD45.1 + OT-II mice were cultured with irradiated splenic APC plus OVA 323-339 peptide under ThO, T fh (IL-21 plus antibodies to IL-4, IFN ⁇ and TGF ⁇ ) or TH 17 condition for 5 days. After 5 days, CD4 + T cells were restimulated with anti-CD3 for 4 hours for real-time PCR analysis (A) or for 24 hours for cytokine measurement by ELISA (B). (C-F).
- mice Germinal center in the spleens of the recipient mice were identified by PNA staining (brown). The results are a representative of multiple mice of two independent experiments with similar results.
- follicular helper T cells co-express CXCR5 and BTLA.
- mice were immunized with KLH in CFA. Seven days after the immunization, experimental mice were sacrificed and the T f1 , cell induction was determined by staining CD4 cells with PE-labeled anti-BTLA mAb and biotinylated anti-CXCR5 mAb, followed by APC-labeled streptavidin.
- CD4 + CD44 h 'CXCR5 + (T ft ) and CD4 + CD44hiCXCR5 ⁇ (non-Ta) cells were sorted and restimulated with KLH and irradiated APC for 48 hours for cytokine measurement by ELISA.
- T fh cell generation is independent of THl and TH2 cells.
- A-C Basal levels of CXCR5 expression in non-immunized IL-4 KO, IFN ⁇ KO, STAT6 KO and STAT4 KO mice were analyzed by staining with PerCP-labeled anti-CD4 mAb and biotinylated anti-CXCR5 mAb, followed by APC-labeled streptavidin. Numbers in dot plot quadrants represent the percentages.
- D IL-4 KO, IFN ⁇ KO and their appropriate controls (WT, 3 mice per group) were immunized with KLH emulsified in CFA.
- CD4 + CD44 hl CXCR5 + (T ft ) and CD4 + CD44hiCXCR5 " (non-Ta) cells were sorted and restimulated with anti-CD3 for 4 hours for real-time RT-PCR analysis of TH subset-specific genes.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Epidemiology (AREA)
- Zoology (AREA)
- Cell Biology (AREA)
- Biomedical Technology (AREA)
- Microbiology (AREA)
- Organic Chemistry (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Biotechnology (AREA)
- Mycology (AREA)
- Oncology (AREA)
- Hematology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Marine Sciences & Fisheries (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
Methods of modulating the differentiation of Tf1, cells by administering an effective amount of an agonist and/or antagonist of IL-6, IL-21, or STAT3.
Description
MODULATION OF FOLLICULAR HELPER T CELLS
CROSS-REFERENCE TO RELATED APPLICATIONS [0001] None.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
[0002] This disclosure was developed at least in part using funding from the National
Institutes of Health, Regulation of Inflammatory Genes in Rheumatoid Arthritis, Grant No. ROl- AR-50772. The U.S. government has certain rights in this invention..
THE NAMES OF THE PARTIES TO A JOINT RESEARCH AGREEMENT
[0003] None.
REFERENCE TO SEQUENCE LISTING [0004] None.
BACKGROUND OF THE INVENTION
[0005] A fundamental function of Th cells is to provide "help" to B cells and regulate their proliferation and immunoglobulin class-switching, especially in the germinal center structures. ThI and Th2 cells have been shown to regulate B cell responses. For example, IFNγ regulates IgG2a production while IL-4 is critical in IgE class-switching. However, an additional Th subset called follicular helper T (Tfh) cells are present in germinal centers and are characterized by their expression of chemokine (C-X-C motif) receptor 5 (CXCR5). Vinuesa, C. G., et al., Follicular B Helper T Cells in Antibody Responses and Autoimmunity, Nature Reviews Immunology Nat Rev Immunol, 2005b, 5:853-865.
[0006] Human Tf1, cells express distinct genes from ThI or Th2 cells. Chtanova, T., et al.,
T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78. Despite their potential importance in humoral immunity and immunopathology, the developmental regulation of Tn1 cells and their relationship with other Th subsets has been unclear to date. For example, IL-21 is expressed by both ThI 7 and Tf1, cells
SUMMARY OF THE INVENTION
[0007] A subset of T cells, named T follicular helper cells (Tfh), provides a helper function to B cells. They stimulate the differentiation of B cells into antibody-forming cells. We found that Tf1, generation is regulated by IL-6, IL-21 and STAT3. Tfh cells have a distinct gene expression profile from THl, TH2 and TH 17 lineages as demonstrated by microarray analysis (The microarray data have been deposited into NCBI GEO with the accession number GSEl 1924). Finally in vitro generated Tf1, cells acquire Tf1, gene expression and function as in vivo generated Tfh cells. Antibody-producing B cells need to be tightly controlled. Any dysregulation of T cell function can have a significant effect on the antibody-producing B cells. For example, excessive function of Tfh cells can contribute to pathogenesis antibody-mediated autoimmune diseases (Systemic lupus erythematosus (SLE)). In this case targeting of specific Tf1, factors will help to prevent this autoimmune disease. However, Tf11 cells can be beneficial during parasite, viral and bacterial infection. During infection antibody is required to confer protection against infectious diseases by neutralizing pathogenic factors on the microorganisms (antibody- mediated protection). In this case Tfh cells will help to clear infection.
BRIEF DESCRIPTION OF THE DRAWINGS
[0008] The foregoing summary as well as the following detailed description of the preferred embodiment of the invention will be better understood when read in conjunction with the appended drawings. It should be understood, however, that the invention is not limited to the precise arrangements and instrumentalities shown herein.
[0009] For a more complete understanding of the present invention, and the advantages thereof, reference is now made to the following descriptions taken in conjunction with the accompanying drawings, in which:
[0010] Figures 1A-1D show that Tfh cells express distinct genes from ThI, Th2 and Thl7 cells.
[0011] Figures 2A-2D depict the generation of Tf]1 cells as independent of ThI and Th2 lineages.
[0012] Figures 3A-3E show B7H expressed on B cells as required for generation of Tf]1 cells.
[0013] Figures 4A-4D show IL-21 is necessary for Tf]1 cell development.
[0014] Figures 5A-5C show the generation of Tfh cells requires IL-6 and STAT3.
[0015] Figures 6A-6C show the generation of Ta cells is independent of THl 7 lineage
[0016] Figures 7A-7G show IL-21, in the absence of IL-4, IFNγ and TGF-b signaling, generates Ta cells.
[0017] Figure 8 depicts follicular helper T cells co-express CXCR5 and BTLA.
[0018] Figures 9 A and 9B show FDR estimated from ANOVA tests using resampled samples.
[0019] Figure 10 shows antigen-specific cytokine production by Tn1 cells.
[0020] Figures 1 IA-I ID shows Tfl, cell generation is independent of THl and TH2 cells.
[0021] Figure 12 shows regulation of antibody responses by STAT3.
[0022] Figures 13 A and 13B show normal germinal center reaction in IL-17 KO, IL- 17F
KO and RORα/γ KO mice.
[0023] Figure 14 depicts a schematic demonstration that development of T^ cells, independent of THl, TH2 or TH 17 lineages, is regulated by IL-6 and IL-21.
DETAILED DESCRIPTION
[0024] Naϊve CD4+ helper T (Th) cells, upon encountering their cognate antigens presented on professional antigen-presenting cells (APC), differentiate into effector cells that are characterized by their distinct cytokine production profiles and immune regulatory functions.
[0025] In addition to ThI and Th2 cells, ThI 7, a third subset of Th cells, has been recently identified. ThI 7 cells produce IL-17, IL-17F and IL-22 and regulate inflammatory responses by tissue cells. Dong, C, et al., Cell Fate Decision: T-helper 1 and 2 Subsets in Immune Responses, Arthritis Res, 2000, 2:179-188; Dong, C, THl 7 Cells in Development: An Updated View of Their Molecular Identity and Genetic Programming, Nat Rev Immunol, 2008, 8:337-348. In mouse, ThI 7 differentiation is initiated by transforming growth factor β (TGFβ) and IL-6, possibly via regulating the chromatin remodeling of the III 7-1117 f 'locus. Bettelli, E., et al., Reciprocal Developmental Pathways for the Generation of Pathogenic Effector TH 17 and Regulatory T Cells, Nature , 2006, 441 :235-238; Mangan, P.R., et al., Transforming Growth Factor-beta Induces Development of the T(H)17 Lineage, Nature, 2006, 441 :231 -234; Veldhoen, M., et al., TGFbeta in the Context of an Inflammatory Cytokine Milieu Supports de novo Differentiation of IL- 17 -producing T Cells, Immunity, 2006, 24:179-189; Akimzhanov, A.M., et al., Chromatin Remodeling of Interleukin-17 (IL-17J-IL-17F Cytokine Gene Locus During Inflammatory Helper T Cell Differentiation, J Biol Chem, 2007, 282:5969-972. While IL-6 is
necessary for Th 17 differentiation, IL-21 is reportedly an autocrine factor induced by IL-6 to regulate ThI 7 differentiation. Korn, T., et al., IL-21 Initiates an Alternative Pathway to Induce Proinflammatory TH 17 Cells, Nature, 2007, 448:484-487; Yang, X.O., et al., STAT3 Regulates Cytokine-mediated Generation of Inflammatory Helper T Cells, J Biol Chem, 2007, 282:9358-9363; Nurieva, R., et al., Essential Autocrine Regulation by IL-21 in the Generation of Inflammatory T Cells, Nature, 2007, 448:480-483; Zhou, L., et al., IL-6 Programs TH-17 Cell Differentiation by Promoting Sequential Engagement of the IL-21 and IL-23 Pathways, Nat Immunol, 2007, 8:967-974. On the other hand, TGFβ signaling mediates ThI 7 differentiation in vivo. Bettelli, E., et al., Reciprocal Developmental Pathways for the Generation of Pathogenic Effector THl 7 and Regulatory T Cells, Nature , 2006, 441 :235-238; Mangan, P.R., et al., Transforming Growth Factor-beta Induces Development of the T(H)17 Lineage, Nature, 2006, 441 :231-234; Veldhoen, M., et al., Signals Mediated by Transforming Growth Factor-beta Initiate Autoimmune Encephalomyelitis, but Chronic Inflammation is Needed to Sustain Disease, Nat Immunol, 2006, 7:1151-1156. Thl7 cell development is dependent on STAT3, which functions to upregulate the expression of two Thl7-specific orphan nuclear receptors RORγt and RORα and ultimately determines ThI 7 terminal differentiation. Laurence, A., et al., Interleukin- 2 Signaling Via STAT5 Constrains T Helper 17 Cell Generation, Immunity, 2007, 26:371-381 ; Yang, X.O., et al., STAT3 Regulates Cytokine-mediated Generation of Inflammatory Helper T Cells, J Biol Chem, 2007, 282:9358-9363; Ivanov, II, et al., The Orphan Nuclear Receptor RORgammat Directs the Differentiation Program of Proinflammatory IL-17+ T Helper Cells, Cell, 2006, 126:1121-1133.
[0026] Although activated T cells may transiently express CXCR5, the Tfh cell subset demonstrates more stable expression of this chemokine receptor. These cells also regulate humoral immunity, especially germinal center reactions. Consistent with this notion, CXCR5 has been shown to be important for proper T and B cell localization in immune responses and antibody production. Haynes, N.M., et al., Role of CXCR5 and CCR7 in Follicular Th Cell Positioning and Appearance of a Programmed Cell Death Gene-lHigh Germinal Center-associated Subpopulation, J Immunol, 2007, 179:5099-5108; Junt, T., et al., CXCR5- Dependent Seeding of Follicular Niches by B and Th Cells Augments Antiviral B Cell Responses, J Immunol, 2005, 175:7109-7116. In addition to CXCR5, other markers have been also reported for TfI1 cells, such as inducible costimulatory receptor (ICOS), IL-21 cytokine and Bcl-6 transcription factor.
[0027] ICOS is essential for generation of Ta cells in vivo. Akiba, H., et al., The Role of
ICOS in the CXCR5+ Follicular B Helper T Cell Maintenance In Vivo, J Immunol, 2005, 175:2340-2348. In addition to ThI 7 cells, IL-21 is also expressed in Tf1, cells and may serve as an important regulator of humoral responses by Tn, cells. IL-21 directly regulates B cell proliferation and class-switching. IL-21 R deficiency results in defective antibody responses and impaired germinal center formation. Spolski, R., et al., Interleukin-21 : Basic Biology and Implications for Cancer and Autoimmunity, Annu Rev Immunol, 2008, 26. On the other hand, sanroque mice, which have a mutation in a RING-type E3 ubiquitin ligase, Roquin, developed spontaneous autoantibody production and lupus-like autoimmunity, associated with greatly increased numbers of CXCR5+ CD4+ T cells and enhanced expression of IL-21 and ICOS. Vinuesa, C. G., et al., A RING-type Ubiquitin Ligase Family Member Required to Repress Follicular Helper T Cells and Autoimmunity, Nature, 2005, 435:452-458.
[0028] Tf1, cells have a divergent gene expression profile from ThI, Th2 and ThI 7 cells, and develop in vivo independent of these lineages. IL-21, IL-6 and STAT3 are critical in the generation of Tf1, cells. Moreover, T cells activated in vitro in the presence of IL-21 but without TGFβ signaling preferentially acquired Tf1, gene expression and functioned to promote humoral immunity in vivo. Thus, as shown herein, our data indicate that Tf1, cells represent a distinct Th lineage and suggest a reciprocal relationship between the Ta and ThI 7 lineages.
[0029] Ta cells are strongly implicated in humoral immunity, including antibody production and antibody-mediated autoimmunity, and germinal center reactions. As shown in the figures, Tf1, cells are distinct from ThI, Th2 or Th 17 cells in their gene expression and developmental regulation. Generation of Tfh cells requires IL-21, IL-6 and STAT3. Thus, IL- 21, IL-6 and STAT3, their signaling, expression and activity are targets in modulating Tf1, cell- mediated immune response. Methods of treating disease, therefore, include administering an agent that modulates IL-6, IL-21, and/or STAT3 signaling, expression or activity in an amount effective to modulate the differentiation of Tf1, cells. Useful agents may modulate the production, growth or activity of Tn, cells. The agent may also modulate the expression of genes expressed by Tfh cells and/or be used in combination with other agents.
[0030] Inducible costimulator (ICOS)-Inducible costimulator ligand (ICOSL) interaction regulates humoral immunity and germinal center reactions. Dong, C, et al., Regulation of Immune and Autoimmune Responses by ICOS, J Autoimmun, 2003, 21 :255-260. This costimulatory pathway is also important in generation OfTf1, cells in mouse. Akiba, H., et al., The Role of ICOS in the CXCR5+ Follicular B Helper T Cell Maintenance In Vivo, J Immunol, 2005,
175:2340-2348. ICOS deficiency in human patients causes a severe reduction of Tfh cells. Bossaller, L., et al., ICOS Deficiency is Associated With a Severe Reduction of CXCR5+CD4 Germinal Center Th Cells, J Immunol, 2006, 177:4927-4932. Conversely, impaired negative regulation of ICOS by Roquin E3 ubiquitin ligase leads to increased numbers of CXCR5+ Tf1, cells and IL-21 hyperproduction. Vinuesa, C.G., et al., A RING-type Ubiquitin Ligase Family Member Required to Repress Follicular Helper T Cells and Autoimmunity, Nature, 2005a, 435:452-458; Yu, D., et al., Roquin Represses Autoimmunity by Limiting Inducible T-cell Co-stimulator Messenger RNA, Nature, 2007, 450:299-303.
[0031] The mechanism by which ICOS and ICOSL regulates Tfh cell development has been studied by the inventors. ICOS-B7h interaction is necessary for IL-21 expression by T cells. ICOS regulates Tn, cells through production of IL-21. IL-21 regulation by calcium signaling and NFAT factors has been shown. Kim, H.P., et al., Calcium-dependent Activation of Interleukin-21 Gene Expression in T Cells, J Biol Chem, 2005, 280:25291-25297. ICOS, together with TcR and CD28 has been shown to increase the expression of NFATcI through a PI-3 kinase- Itk-calcium pathway. Nurieva, R.I., et al., A Costimulation-initiated Signaling Pathway Regulates NFATcI Transcription in T Lymphocytes, J Immunol, 2007b, 179:1096-1103; Nurieva, R.I., et al., Transcriptional Regulation of Th2 Differentiation by Inducible Costimulator, Immunity, 2003a, 18:801-811. ICOS can act through NFATcI to regulate IL-21 expression. Since IL-21 regulates ThI 7 differentiation, an IL-21 defect can account for the impairment in IL- 17 expression in the ICOS-deficient animals. Dong, C, et al., Regulation of Immune and Autoimmune Responses by ICOS, J Autoimmun, 2003, 21 :255-260. As shown, using a B cell-specific ICOSL deficient mouse, ICOSL expression on B cells is required for the generation of Tf]1 cells and IL-21 expression. This result not only substantiates the importance of ICOS-ICOSL interaction in Tfh cell development, but also suggests an important function of B cells in vivo as APCs in the generation or maintenance of Tf1, cells. Consistent with our findings, Ebert et al previously showed that human B cells regulate Tfh cell phenotypes. Ebert, Lisa M., et al., B Cells Alter the Phenotype and Function of Follicular-homing CXCR5+ T Cells, Eur J Immunol, 2004, 34:3562-3571.
[0032] Previous gene expression analysis has revealed that human Tfh cells are distinct from ThI and Th2 cells. Chtanova, T., et al., T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78; Kim, CH. , et al., Unique Gene Expression Program of Human Germinal Center T Helper Cells, Blood, 2004, 104:1952-1960. The data reported here
also reveals that Tf1, cells do not produce ThI or Th2 cytokines. Although human Tfh cells appear to express IL-10 and CXCLl 3, a ligand for CXCR5, mouse Tf1, cells do not. Chtanova, T., et al., T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78; Ebert, Lisa M., et al., B Cells Alter the Phenotype and Function of Follicular-homing CXCR5+ T Cells, Eur J Immunol, 2004, 34:3562-3571; Kim, C.H., et al., Unique Gene Expression Program of Human Germinal Center T Helper Cells, Blood, 2004, 104:1952-1960; Kim, C.H., et al., Subspecialization of CXCR5+ T Cells: B Helper Activity is Focused in a Germinal Center-localized Subset of CXCR5+ T Cells, J Exp Med, 2001, 193:1373-1381. The data reported here show that naϊve T cells differentiate into Ta cells in vivo, independent of IFNγ, IL- 4 and STAT6. Hence, T^ cells develop independent of ThI and Th2 lineages.
[0033] On the other hand, T^ cells share common regulators with ThI 7 cells. Both subsets express IL-21 and their development depends similarly on IL-6, IL-21 and STAT3. However, Tfh cells differ from ThI 7 cells in the following aspects. First, they are distinct in their gene expression profiles. Second, Tfh cells do not produce IL-17, IL-17F or IL-22. Additionally, Tfh cell development does not require RORα or RORγt. Thus, we conclude that Tf1, cells develop independent of the Th 17 lineage.
[0034] ThI 7 development in mouse is not only mediated by the IL-6-IL-21 axis, but also by TGFβ. As shown, IL-21 can be induced in T cells independent of TGFβ signaling. T cells activated in the presence of IL-21 but in the absence of IL-4, IFNγ and TGFβ signaling produced IL-21 but not IL-4, IFNγ, IL-17, IL-17F or IL-22. Furthermore, these cells acquired expression of CXCR5, Bcl-6, IL-6R and IL-6st, genes expressed by in vivo generated Tfh cells, suggesting that Tfh cells may be generated in vitro under the above condition. Moreover, these Tfh-Hke cells generated in vitro preferentially expressed CXCR5 in vivo and functioned to promote humoral immunity, similar to in vivo-generated Tf)1 cells. Thus, a novel approach and methods for generating Tfh cells in vitro is provided herein.
[0035] Furthermore, TGFβ signaling, is not essential for Tf1, cells, although required for
IL- 17 expression in vivo, indicating a reciprocal relationship of Tf1, and Th 17 cells. Interestingly, Bcl-6 has recently been shown as a repressor of TGFβ-SMAD signaling. Wang, D., et al., BCL6 Represses Smad Signaling in Transforming Growth Factor-B Resistance, Cancer Research, 2008. The lack of TGFβ signaling favors Tfi, cell development. Tfh cell development is independent of ThI, Th2 and Thl7 cells, and IL-21 serves as critical factor for generation of this
lineage. Our in vitro generated Tf1, cells also enhanced the Tf11 cell generation in recipient mice, suggesting that IL-21 may function in a paracrine fashion to regulate Tfh cell development.
[0036] In summary, an extensive characterization of the developmental regulation of Ta cells and useful applications of this information are disclosed herein. The data shows that Tfh cells are distinct in their gene expression and immune function and develop via a pathway that is dependent on IL-21 or IL-6 but independent of ThI, Th2 or ThI 7 lineages (Figure 14). In mice defective in Tfh cells, there were still detectable amounts of antigen-specific antibodies, suggesting that other Th subsets may independently regulate the humoral immunity. This knowledge may be applied to treat disease, e.g., antibody-mediated autoimmune diseases or other diseases associated with Tf1, cell-mediated immune response.
[0037] As a result, Tf1, cell function and/or generation may be modulated by use of an agent such as an antagonist of one or more of IL-6, IL-21, and STAT3, and/or an agonist of one or more of IL-6, IL-21, and STAT3. Use of such an agent in an amount effective to inhibit or induce the differentiation of Ta cells, and/or to modulate the expression of genes expressed by Ta cells, can affect Tf1, cell-mediated immune response. For example, antibody-mediated autoimmune diseases may be treated by administration of a antagonist of one or more of IL-6, IL-21, and STAT3 in an amount effective to inhibit the differentiation of Tf1, cells and/or modulate gene expression by Tf1, cells. On the other hand, an agonist of one or more of IL-6, IL- 21, and STAT3 may be used to increase antibody production against infections and cancers by administering the agonist in an amount effective to induce the differentiation of Tf1, cells and/or modulate the gene expression by Tf1, cells.
[0038] Differentiation Of Tf1, cells can be modulated via one or more of the IL-6, IL-21, and STAT3 signaling pathways. Antagonists that may interrupt these pathways may be small molecule inhibitors, antibodies and the like. IL-6, IL-21, and/or STAT3 antagonists which are suitable agents to modulate Tfh cell mediated immune response include molecules that bind to any of these signalling factors and/or their corresponding receptors. Examples of antagonists that may be suitable include soluble receptor antagonists and antibodies that prevent binding of IL-6, IL-21, and/or STAT3 to its receptor. For example, inhibition of the activity of any of IL-6, IL-21, and STAT3 may be accomplished by binding the corresponding receptor with a therapeutically effective amount of a receptor antagonist such as a blocking antibody. Functions of Tfh cells which may be reduced by interrupting a IL-6, IL-21 , and STAT3 signaling pathway include, for example, expansion of Tn, cells and production of factors produced by Tf11 cells.
[0039] Methods of modulating Ta cell mediated immune response through inhibition of
IL-6, IL-21, and/or STAT3 may be useful for the treatment of disorders or a wide variety of conditions where decreased Ta cell mediated immune response is useful. Disorders or conditions advantageously treated by these methods include autoimmune diseases, which involve Ta cell-mediated immune response. Examples of disorders or conditions which may be treated by decreasing Ta cell-mediated immune response include, antibody-mediated auto immune diseases such as lupus.
[0040] Further disclosed herein are methods of treating infection or cancer by inducing
Ta cell -mediated immune response through induction of IL-6, IL-21, and/or STAT3 activity and/or expression. These methods include administering a therapeutically effective amount of IL-6, IL-21, and/or STAT3 or an agonist for IL-6, IL-21, and/or STAT3 to an individual in need thereof. Here, the IL-6, IL-21, and/or STAT3, or agonist thereof, increases the generation, differentiation and function of T^ cells in the individual, thereby increasing T^ cell-mediated response to infection or, in some cases, tumors. Examples of the types of infections or tumors that may benefit from increased Ta, cell mediated immune response include immunity against excellular bacteria, fungus, viruses, and tumors including melanoma. IL-6, IL-21, and/or STAT3 agonists or similar include mimicry fragments, small molecules, and molecules and proteins of similar function.
[0041] The phrase "therapeutically effective" is intended to qualify the amount of active ingredients used in the treatment of a disease or disorder. This amount will achieve the goal of reducing or eliminating the said disease or disorder.
[0042] As used herein, reference to "treatment" of a patient is intended to include prophylaxis. The term "patient" means all mammals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, pigs, and rabbits. Preferably, the patient is a human.
[0043] While it may be possible for the molecules which inhibit or induce IL-6, IL-21, and/or STAT3 activity to be administered as the raw chemical, it is also possible to present them as a pharmaceutical formulation. Accordingly, the pharmaceutical formulation may include the molecule or a pharmaceutically acceptable salt, ester, prodrug or solvate thereof, where appropriate, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Proper formulation is dependent upon the route of administration chosen. Any
of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences.
[0044] The formulations of use molecules include those suitable for oral, parenteral
(including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. All methods may include the step of bringing into association the molecule or a pharmaceutically acceptable salt, ester, prodrug or solvate thereof ("active ingredient") with the carrier which constitutes one or more accessory ingredients.
[0045] In certain instances, it may be appropriate to administer at least one molecule (or a pharmaceutically acceptable salt, ester, or prodrug thereof) in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving one molecule is hypertension, then it may be appropriate to administer an antihypertensive agent in combination with the initial therapeutic agent. Or, by way of example only, the therapeutic effectiveness of one molecule may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). Or, by way of example only, the benefit of experienced by a patient may be increased by administering one molecule as described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
[0046] Multiple therapeutic agents may be administered in any order or even simultaneously. If simultaneously, the multiple therapeutic agents may be provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). One of the therapeutic agents may be given in multiple doses, or both may be given as multiple doses. If not simultaneous, the timing between the multiple doses may be any duration of time ranging from a few minutes to four weeks.
[0047] Thus, a subset of T cells, termed T follicular helper cells ("TFH", "Tfh", Tf]1", and the like), provides a helper function to B cells and contributes to pathogenesis of certain antibody-mediated autoimmune diseases and immunodeficiencies. CXCR5+ TFH cells are important in humoral immunity. The targeting of factors regulating TFH generation and function is a novel method of treating disease. It is shown that TFH cells have a distinct gene expression
profile from other effector T cells and develop in vivo independent of the THl or TH2 lineages. TFH cell generation is regulated by B7h expressed on B cells and, similar to TH 17 cell development, is dependent on IL-21, IL-6 and STAT3. However, differentiation of TFH cells, unlike THl 7 cells, does not require TGFβ signaling or TH17-specific orphan nuclear receptors RORα and RORγ in vivo. Further, naϊve T cells activated in vitro in the presence of IL-21 but not TGFβ signaling preferentially acquire TFH gene expression and function to promote germinal center reactions in vivo. TFH is thus demonstrated as a distinct lineage of effector TH differentiation, and IL-6, IL-21, and STAT3 are shown to be important regulators of these cells. Antagonists of these factors can be used in the treatment of autoimmune diseases, while agonists of these factors can be used to promote generation and expansion of TFH cells, which can help antibody production against infections and cancers.
METHODS
[0048] Mice. IL-4-, IFNγ- and IL-6-defϊcient mice on C57BL/6 background and STAT6 and STAT4-deficient mice on BALB/c background were purchased from Jackson Laboratories and C57BL/6, B6.SJL (CD45.1) and BALB/c mice were used as controls. Rordt/st, Rordl/sl Rorγ'~ and wild-type bone marrow chimeras were generated as described by Yang et al. Yang, X.O., et al., T Helper 17 Lineage Differentiation is Programmed by Orphan Nuclear Receptors RORfalphaJ and RORfgammaJ, Immunity, 2008b, 28:29-39. Stat3 fl/fl mice were bred with CD4-Cre mice provided by Dr. Christopher Wilson. Takeda, K., et al., Enhanced ThI Activity and Development of Chronic Enterocolitis in Mice Devoid of Stat3 in Macrophages and Neutrophils, Immunity, 1999, 10:39-49; Lee, P.P., et al., A Critical Role for Dnmtl and DNA Methylation in T Cell Development, Function, and Survival, Immunity, 2001, 15:763-774. IL-21 deficient mice on 129xC57BL/6 Fl mixed background were obtained from NIH Mutant Mouse Regional Resource Centers (MMRRC). Nurieva, R., et al., Essential Autocrine Regulation by IL-21 in the Generation of Inflammatory T Cells, Nature, 2007, 448:480-483. ICOSL-BKO mice were created by breeding ICOSL flox mice with CD19-cre mice. Nurieva, R.I., et al., B7h is Required for T Cell Activation, Differentiation, and Effector Function, Proc Natl Acad Sci U S A, 2003, 100:14163-14168; Rickert, R.C., et al., Impairment of T-cell-dependent B-cell Responses and B-I Cell Development in CD 19 -deficient Mice, Nature, 1995, 376:352-355. IL- 17- and IL-17F- deficient mice were recently generated in the lab. Yang, X., et al., Regulation of Inflammatory Responses by 1L-17F, J Exp Med In press, 2008. Mice were housed in the SPF animal facility at M. D. Anderson Cancer Center and the animal experiments were performed at
the age of 6-10 weeks using protocols approved by Institutional Animal Care and Use Committee.
[0049] T cell differentiation. Differentiation of OT-II cells as represented in Figures
IA-B was performed as previously described. Chung, Y., et al., Expression and Regulation of IL-22 in the IL- 17 -Producing CD4+ T Lymphocytes, Cell Res, 2006, 16:902-907. In Figure 7A, the cytokine stimuli for Thl7 differentiation were 100 ng/ml of IL-21, 5 ng/ml of TGFβ, and 10 μg/ml of anti-IL-4 and 10 μg/ml of anti-IFNγ, and for generation of Tfh cells were 50 ng/ml of IL-21, 10 μg/ml anti-IFNγ, 10 μg/ml anti-IL-4 and 20 μg/ml TGF-β (lDl l) neutralizing antibodies. IL-4, IL-6, IL- 12 and TGFβ were purchased from Peprotech. IL-21, IL-23 and TGF- β (IDl 1) neutralizing antibodies were purchased from R&D. To characterize the in vitro- differentiated CD4+ T cells under these conditions, these cells were restimulated with plate- bound anti-CD3 (5 μg/ml) for 4 hours for real-time PCR analysis, or for 24 hours for cytokine measurement by ELISA.
[0050] Keyhole Limpet Hemocyanin (KLH) Immunization. Various strains of mice
(6-8 wk old; three per group) were immunized with KLH (0.5 mg/ml) emulsified in CFA (0.5 mg/ml) at the base of the tail (100 μl each mouse). In Figure 6 for local blockade of TGF-β, 100 μg anti-TGFβ (IDl 1) was included in the emulsion; the control group received 100 μg isotype control antibodies. Seven days after immunization, these mice were sacrificed and analyzed individually. The germinal center B cells were determined by staining with FITC-labeled PNA (Pharmingen) and PerCP-labeled anti-B220 mAb (Pharmingen). The Tf1, cell induction was determined by staining with PerCP-labeled anti-CD4 mAb (Pharmingen) and biotinylated anti- CXCR5 mAb (Pharmingen), followed by APC-labeled streptavidin (Jackson ImmunoResearch Laboratories, Inc.). In some experiments, sera from immunized mice were collected, and antigen- specific IgM, and IgG antibodies were measured by using ELISA. Briefly, serum samples were added in a 3 -fold serial dilution onto plates precoated with 10 μg/ml KLH or Ova protein. Antigen-specific antibodies were detected with biotinylated goat antimouse IgM or rat anti- mouse IgG antibodies (Southern Biotechnology Associates). To analyze the role of B7h in regulation of T cell responses in vivo (Fig. 3A and C), spleen cells from KLH-immunized mice were stimulated in 96-well plates as triplicates with or without KLH. Effector cytokines (IFN-γ and IL-21) were analyzed 4 days later by ELISA (Pharmingen). In Figure 6 B, spleen cells from immunized mice were restimulated with 50 μg KLH for 24 h. In the final 5 hours, Golgi-stop (BD Bioscience) was added and IL-17- and IFNγ-producing cells were analyzed using a BD CytoFix/CytoPerm intracellular staining kit (BD Bioscience).
[0051] Statistical analysis of microarray data. The DNA microarray analysis was carried out at the Institute for Systems Biology microarray core facility using Affymetrix Mouse 430 2.0 chips. The total RNA samples were labeled according to manufacturer's instruction using One-Cycle Target Labeling method, which consists of oligo-dT primed cDNA synthesis followed by in vitro transcription that incorporates biotinylated nucleotides. The microarray data were normalized using GCRMA. Zhijin, W., et al., A Model-based Background Adjustment for Oligonucleotide Expression Arrays, Journal of the American Statistical Association, 2004, 99:909-917. We then selected the genes whose expression was changed across the ThI, Th2, ThI 7, and Tf1, cells using a False Discovery Rate (FDR) estimation method: 1) one-way ANOVA was performed to compute unadjusted p-value for the four classes of cells; 2) the number of non- differentially expressed probe-sets (m0) was estimated as 2 x the number of the probe-sets with p-value>0.5 (mθ= 25088); 3) the expected number of false positives under the complement null hypothesis E(Vo) was estimated for a given ANOVA F-statistic value by performing 500 times of resampling; and 4) FDR was finally estimated as mo/m x E(V0)/R where m is the total number of the probe-sets (m=45037) and R is the number of the genes being selected using the given ANOVA F-statistic value (FDR estimated for various F-statistic value is shown in Figure 9B). Storey, J.D., et al., Estimating the Positive False Discovery Rate Under Dependence, With Applications to DNA Microarrays, (Department of Statistics, Stanford University), 2001, VoI 2001. Then, the expression of 8350 probe-sets showing differential expressions among the four types of cells (FDR = 0.1, corresponding to an unadjusted p-value = 0.033 from one-way ANOVA) was used for hierarchical clustering (Euclidian distance metric and Ward Minimum Variance Linkage) and PCA. The microarray data have been deposited into NCBI GEO with accession GSEl 1924.
[0052] Adoptive transfer study. CD4+ T cells from OT-II mice (CD45.2) were intravenously transferred into C57BL/6 (CD45.1+) mice (3x106 cells/mouse) (3 groups; 3 mice per group). 2 groups of recipient mice were immunized subcutaneously with 100 μg Ova protein emulsified in CFA and treated with a 300 μg of control rat Ig or anti-IFNγ and anti-IL-4 mAbs at the time of immunization (day 0) and on days 2 and 4. Seven days after the immunization, experimental mice were sacrificed and splenic CD45.1+ and CD45.2+ CD4 cells were stained with biotinylated anti-CXCR5 mAb, followed by APC-labeled streptavidin. In Figure 7C-E, FACS-sorted naϊve (CD4+CD62LhiCD44~CD25~) T cells from CD45.1+ OT-II mice were activated under THO or Tn1 condition, washed and intravenously transferred into C57BL/6 (CD45.2+) mice (4x106 cells/mouse) and the recipient mice were subcutaneously immunized
with 100 μg OVA protein emulsified in IFA. A group of C57BL/6 mice that did not receive T cells was used as a control (No transfer). In Fig. 7G, CXCR5+CD44hl cells were sorted from B6.SJL (CD45.1) mice immunized with KLH. These cells were transferred into C57BL/6 (CD45.2+) mice (5x106 cells/mouse) (3 mice per group). Second group did not receive cells. All mice were immunized subcutaneously with 1000 μg KLH. Seven days after immunzation, lymphoid cells from the draining lymph nodes of the recipient mice were isolated and stained with FITC-labeled anti-CD45.1 mAb and PerCP-labeled anti-CD4 mAb plus biotinylated anti- CXCR5, followed by APC-labeled streptavidin, or stained with FITC-labeled PNA and PerCP- labeled anti-B220.
[0053] Quantitative real-time PCR. Total RNA was prepared from T cells using TriZol regent (Invitrogen). cDNA were synthesized using Superscript reverse transcriptase and oligo(dT) primers (Invitrogen) and gene expression was examined with a Bio-Rad iCycler Optical System using iQ™ SYBR green real-time PCR kit (Bio-Rad Laboratories, Inc.). The data were normalized to β-actin reference. The following primer pair for Bcl-6, IL-6R, IL-6st, and CXCR5 were used: Bcl6 forward: CACACCCGTCCATCATTGAA, reverse: TGTCCTCACGGTGCCTTTTT; IL6R forward: GGTGGCCCAGTACCAATGC, reverse: GGACCTGGACCACGTGCT; CXCR5 forward: ACTCCTTACCACAGTGC ACCTT, reverse: GGAAACGGGAGGTGAACCA; IL-6st forward: ATT TGT GTG CTG AAG GAG GC, reverse: AAA GGA CAG GAT GTT GCA GG. The primers for IL-21, IL-17, IL- 17F, IL-4, IFNγ, RORγ, RORα, T-bet, GATA3, and β-actin were previously described. Yang, X.O., et al., STAT3 Regulates Cytokine-mediated Generation of Inflammatory Helper T Cells, J Biol Chem, 2007, 282:9358-9363.
[0054] Immunohistochemical analysis. Fresh mouse spleen tissues were embedded in
OCT and frozen with isopentane in Histobath. Tissue blocks were sliced 6 μm with cryotome. Slides were fixed with acetone cold. Purified anti-mouse CD4 or biotin labeled anti-mouse PNA were applied as primary antibody ("Ab") following with biotinylated anti-rat secondary antibody and avidin-peroxidase complex reagent. Novared was used as substrate. For double staining, biotin-labeled B220 was applied and following with avidin-aklinphosphotase complex reagent. Vector blue was used as substrate. CD4 and B220 are from BD Pharmagen. PNA and other reagents were from Vector Laboratary. Slides for PNA staining were count stained with Hematoxlin.
EXAMPLE 1 Distinct Gene Expression Profile of Tfh Cells
[0055] As a first step toward understanding Tβ, cell regulation, we compared the gene expression profiles of ThI, Th2 and ThI 7 cells differentiated in vitro with in- vivo generated Tfh cells. Taking advantage of the co-expression of B and T lymphocyte attenuator (BTLA) by CXCR5+ Ta cells (Figure 8), CD4+CD44hiCXCR5+BTLA+ were FACS sorted from splenocytes of C57BL/6 mice seven days after immunization with keyhole limpet hemocyanin (KLH). These cells as well as ThI, Th2 and ThI 7 cells were restimulated with anti-CD3 for 4 hours and subject to gene profiling analysis in duplicates using Affimetrix gene chips. The microarray data were normalized using GCRMA and the genes whose expression was changed across the ThI, Th2, Th 17 and Tfh cells were then selected using a False Discovery Rate (FDR) estimation method. Then, the expression levels of 8350 probe-sets showing differential expressions among the four types of cells were used for hierarchical clustering, which revealed that Tfh cells have a very distinct gene expression profile (Figure IA, Figure 9A).
[0056] To confirm the above results, we performed real-time RT-PCR analysis on multiple subset-specific genes. The data indicate that Tf1, cells did not express the typical markers for ThI (IFNγ and T-bet) or Th2 (IL-4 and GATA3) cells (Figure IB), consistent with a previous report on human Tf]1 cells. Chtanova, T., et al., T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help or B Cells, J Immunol, 2004, 173:68-78. Although Tf11 cells shared IL-21 expression with ThI 7 cells, they did not express IL-17, IL-17F, IL-22 or RORγt (Figure IB). Instead, similar to their human counterparts, mouse Ta cells express mRNAs for CXCR5 as well as Bcl-6 (Figure IB). Chtanova, T., et al., T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78; Kim, CH. , et al., Unique Gene Expression Program of Human Germinal Center T Helper Cells, Blood, 2004, 104:1952-1960. In addition, Tf11 cells preferentially expressed mRNAs for IL-6R and IL-6st (gpl30) and also upregulated the expression of IL-21R (Figure IB), suggesting possible regulation Of Tf11 cells by IL-6 and IL-21. Moreover, consistent with human Tf11 cells and a report by Haynes et al. indicating that Tf11 cells express programmed death- 1 (PD-I) protein, PD-I mRNA was highly upregulated in Tf11 cells compared to other Th subsets (data not shown). Chtanova, T., et al., T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Helpor B Cells, J Immunol, 2004, 173:68-78; Haynes, N.M., et al., Role of CXCR5 and CCRl in
Follicular Th Cell Positioning and Appearance of a Programmed Cell Death Gene-lHigh Germinal Center-associated Subpopulation, J Immunol, 2007, 179:5099-5108.
[0057] To substantiate the above results, we also measured cytokine secretion of purified
Ta cells after they were activated ex vivo with anti-CD3 and anti-CD28 for 24 hours. High expressions of IL-21 but not IL-4, IL-10, IFNγ or IL- 17 were observed in Tfh cells (Figure 1C). In addition, purified Ta cells were activated with KLH and irradiated splenic APC. Consistent with above results, Ta cells preferentially produced IL-21, but not ThI, Th2 and Th 17 cytokines (Figure 10). Furthermore, intracellular analysis on CXCR5+ and CXCR5" cells following PMA and ionomycin restimulation also revealed that Tfh cells did not express IFNγ or IL- 17 (Figure ID). These results indicate that Tf1, cells are distinct from ThI, Th2 and ThI 7 cells in their gene expression and cytokine production.
EXAMPLE 2 Naϊve T cell differentiation into Ta cells is independent of ThI and Th2 lineages
[0058] Previous and our current gene expression analysis on human and mouse Tfh cells, respectively, suggested that they are distinct from ThI and Th2 cells. Chtanova, T., et al., T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78. To determine whether generation of Ta cells is distinct from ThI or Th2 differentiation, we transferred FACS sorted naϊve CD4+ T cells (CXCR5 ) from OT-II TcR transgenic mice (CD45.2+, Figure 2A) into C57BL/6 recipients (CD45.1+) followed by immunization with Ova protein in CFA. Seven days later, we examined splenic CD45.1+ and CD45.2+ CD4+ T cells, and found a substantial increase in the frequency of CXCR5+CD4+ T cells (Figure 2A). Treatment of recipient mice with antibodies to IL-4 and IFNγ did not reduce the proportion of CXCR5+CD4+ T cells (Figure 2A), suggesting that Tf1, cell development is independent of ThI or Th2 development. In addition, donor CD44hiCXCR5+ and CD44hiCXCR5" OT-II cells from immunized mice were purified and real-time RT-PCR analysis of Tf11 specific genes was performed. CXCR5+ T cells were generated in the presence of blocking antibodies to IL-4 and IFNγ exhibited gene expression patterns comparable to those from control group (Figure 2A), supporting that these CXCR5+ cells were indeed Tf1, cells.
[0059] To substantiate the above finding, we also immunized mice deficient in IL-4,
IFNγ, STAT6 or STAT4 and their appropriate controls with KLH. Seven days after immunization, we did not detect any defect in Tfh cells or PNA+ germinal center B cells in the IL-4, IFNγ, STAT6 or STAT4 deficient animals compared to wild-type controls (Figure 2B-D,
Figures 1 IA-11C). To confirm the above results, we also measured cytokine secretion of purified CXCR5+ cells from IL-4- and IFNγ-deficient mice and their controls after they were activated ex vivo with anti-CD3 and anti-CD28 for 24 hours. Ta cells from IL-4- or IFNγ-deficient mice produced similar amount of IL-21, but did not express IL-4, IL-10, IFNγ or IL-17 (Supplementary Figure 4D), supporting that they were Ta cells. Thus, we conclude that CXCR5+CD4+ Ta cells develop independent of the ThI and Th2 lineages.
EXAMPLE 3 ICOSL expressed on B cells regulates the generation of Tfh cells
[0060] Inducible costimulator (ICOS) is the third member of the CD28 family with an important role in regulation of T-dependent antibody responses and germinal center reactions. Dong, C, et al., Regulation of Immune and Autoimmune Responses by ICOS, J Autoimmun, 2003, 21 :255-260. ICOS was previously shown to be expressed at high levels on human tonsillular CXCR5+ T cells within the light zone of germinal centers and efficiently supported the immunoglobulin production. Breitfeld, D., et al., Follicular B Helper T Cells Express CXC Chemokine Receptor 5, Localize to B Cell Follicles, and Support Immunoglobulin Production, J Exp Med, 2000, 192:1545-1552; Schaerli, P., et al., CXC Chemokine Receptor 5 Expression Defines Follicular Homing T Cells With B Cell Helper Function, J Exp Med, 2000, 192:1553-1562. In addition, ICOS deficiency in human and mouse resulted in substantially reduced numbers of Ta cells, indicating an essential role of ICOS in the differentiation of CXCR5+ CD4 T cells. Akiba, H., et al., The Role of ICOS in the CXCR5+ Follicular B Helper T Cell Maintenance In Vivo, J Immunol, 2005, 175:2340-2348; Bossaller, L., et al., ICOS Deficiency is Associated With a Severe Reduction of CXCR5+CD4 Germinal Center Th Cells, J Immunol, 2006, 177:4927-4932. Consistently, we also observed reduced percentages of CXCR5+CD4+ cells and decreased expression of IL-21 but not IFNγ in our ICOSL germline deficient animals following KLH immunization (Figure 3A). Nurieva, R.I., et al., B7h is Required for T Cell Activation, Differentiation, and Effector Function, Proc Natl Acad Sci U S A, 2003, 100:14163-14168. Tfh cells are regarded as regulators of the germinal center reaction by providing help to activated B cells that also express CXCR5. Because B cells constitutively express ICOSL, we asked whether the generation of Ta cells may require B cell help via engagement of ICOS receptor on T cells. We thus bred mice carrying the ICOSL conditional flox (f) allele with CD19-cre mice. Nurieva, R. I., et al., B7h is Required for T Cell Activation, Differentiation, and Effector Function, Proc Natl Acad Sci U S A, 2003, 100:14163-14168; Rickert, R. C, et al., Impairment of T-cell-dependent B-cell Responses and B-I Cell Development
in CD19-deficient Mice, Nature, 1995, 376:352-355. In ICOSL f/f mice carrying the CD19-cre (ICOSL-BKO), in comparison to those without ere expression, there was efficient deletion of the ICOSL gene in B cells, similar to those in ICOSL germline deficient mice (Figure 3B). We then immunized ICOSL BKO mice with KLH and found that absence of ICOSL in B cells led to a greatly reduced frequency of CXCR5+CD4+ cells (Figure 3C). Similarly, when CD45.1+ OT-II cells were transferred into B cell specific ICOSL deficient mice, the numbers of CD45.1+ Tfh cells were also greatly reduced following Ova peptide immunization (data not shown). In addition to CXCR5 expression, we found that the expression of IL-21 was also greatly reduced in ICOSL BKO mice, whereas IFNγ expression was elevated in these mice (Figure 3C). Moreover, PNA+ germinal center B cells were greatly reduced in these animals (Figure 3 C and E). KLH-specific IgG production was also reduced (Figure 3D). Overall, these data indicate that B cell expression of ICOSL is necessary for IL-21 production and for the generation of Tfh cells and appropriate antibody responses, indicating that Tf11 cell differentiation is regulated by B cells.
EXAMPLE 4 IL-21 and IL-6 are required for generation of Tfh cells, which is dependent on STAT3
[0061] IL-21 has been recently shown to be induced by IL-6 and to autoregulate its own expression during ThI 7 differentiation. Nurieva, R., et al., Essential Autocrine Regulation by IL-21 in the Generation of Inflammatory T Cells, Nature, 2007, 448:480-483. In addition, TQ1 T cells produced a greater amount of IL-21 compared to ThI and Th2 subsets, and induced the differentiation of autologous B cells into Ig-secreted plasma cells through IL-21. Chtanova, T., et al. , T Follicular Helper Cells Express a Distinctive Transcriptional Profile, Reflecting Their Role as Non-Thl/Th2 Effector Cells That Provide Help for B Cells, J Immunol, 2004, 173:68-78; Bryant, V.L., et al., Cytokine-mediated Regulation of Human B Cell Differentiation Into Ig-secreting Cells: Predominant Role of IL-21 Produced by CXCR5+ T Follicular Helper Cells, J Immunol, 2007, 179:8180-8190. Becouse IL-21 is also expressed in Ta cells, we assessed if IL-21 is important for Tf1, cell generation. I121+/+' +/~ and ~'~ mice were immunized with KLH and splenic Tf11 cells were analyzed in these mice. Il21+/~ mice exhibited reduced number of Tfh cells, which was further reduced in I121'1' mice (Figure 4A). In addition, PNA+ germinal center B cells were also greatly reduced in 1121~'~ mice (Figure 4B-C). In contrast, CD4 T cells from 112 f'~ and ~'~ mice showed normal proliferation and IFNγ expression after re-stimulation with KLH ex vivo. (Figure 5D). Thus, these results indicate that IL-21 is necessary for Tf11 cell development.
[0062] Because IL-6 induces IL-21 expression, we also tested Tn1 cell generation in mice lacking IL-6. It has been previously that IL-6 deficient mice showed reduced germinal centers
and antigen specific Ig-production (IgG). Kopf, M., et al., Interleukin 6 Influences Germinal Center Development and Antibody Production Via a Contribution of C3 Complement Component, J Exp Med, 1998, 188:1895-1906. Compared to the wild-type controls, I16~'~ mice exhibited greatly reduced numbers of Tf1, cells and germinal center B cells (Figure 5A), indicating that IL-6 is also necessary for Tf1, cell generation.
[0063] Because Tf]1 cells expressed high amounts of IL-21R, IL-6R and IL-6st (Figure
IB), we also examined if IL-6 and IL-21 signaling regulates Tf1, cells. C57BL/6 mice were immunized with KLH and CD4+CD44hi T cells with or without CXCR5 expression were FACS sorted and restimulated with anti-CD3 and anti-CD28 in the absence or presence of IL-21 or IL- 6. Compared to non-Tfh cells, CXCR5+ T cells showed a significant reduction in proliferation when activated with anti-CD3 and anti-CD28. Treatment of CXCR5+ T cells with IL-6 or IL-21 significantly enhanced their proliferation (p<0.005) (Figure 5B), suggesting that IL-6 and IL-21 preferentially regulate Tf1, cells. Furthermore, we stained restimulated CXCR5+ and CXCR5" cells with Annexin V. We found that a substantial portion of CXCR5+ cells underwent apoptosis compare to CXCR5- (Figure 5B). However, IL-6 or IL-21 treatment reduced apoptosis of Tf1, cells.
[0064] Because both IL-6 and IL-21 signal through STAT3, we analyzed Tf1, cell generation in STAT3 flox/flox mice bred with CD4-cre mice. Takeda, K., et al., Enhanced ThI Activity and Development of Chronic Enterocolitis in Mice Devoid ofStat3 in Macrophages and Neutrophils, Immunity, 1999, 10:39-49; Lee, P.P., et al., A Critical Role for Dnmtl and DNA Methylation in T Cell Development, Function, and Survival, Immunity, 2001, 15:763-774. The deletion of STAT3 gene in CD4+ thymocytes was found to be complete (data not shown). When we immunized these mice as well as their controls with KLH, the numbers of CXCR5+ Tf1, cells were found to be greatly reduced in the absence of STAT3 (Figure 5C). Moreover, STAT3 deficiency in T cells also led to defective germinal center B cell generation (Figure 5C). KLH- specific IgG and IgM production was also reduced in the absence of STAT3 in T cells (Figure 12). Overall, these data indicate that Tf1, cell generation is dependent on IL-21, IL-6 and STAT3.
EXAMPLE 5 Tfh cell generation is independent of ThI 7 differentiation or function
[0065] The above results indicate that the IL-6-IL-21 axis and STAT3 transcription factor are necessary for Tf1, cell generation, which is similar to the regulation of ThI 7 differentiation. To test whether the development of Tf1, cells is dependent or independent upon the ThI 7 lineage, we utilized Ragl'1' mice reconstituted with bone marrow cells from wild-type,
Rordt/st or RordtlstRorγ'~ mice. We previously reported that the latter mice were completely impaired in ThI 7 differentiation in vitro and in vivo. Yang, X.O., et al., T Helper 17 Lineage Differentiation is Programmed by Orphan Nuclear Receptors RORfalphaJ and RORfgammaJ, Immunity, 2008, 28:29-39. However, upon KLH immunization, these mice deficient in both RORα and RORγ in lymphocytes developed more CXCR5+ cells in spleen (Figure 6A), indicating that Tf1, cells can be generated in the absence of ThI 7 development. Moreover, PNA+ germinal center B cells were not affected in these animals (Figure 13B).
[0066] Since ThI 7 differentiation requires also TGFβ in addition to IL-6 or IL-21, we examined whether TGFβ signaling is required for Tf11 cell generation. C57BL/6 mice were immunized with KLH in the absence or presence of TGFβ blocking antibody as previously described. Veldhoen, M., et al., Signals Mediated by Transforming Growth Factor-beta Initiate Autoimmune Encephalomyelitis, but Chronic Inflammation is Needed to Sustain Disease, Nat Immunol, 2006, 7:1151-1156. While IL-17 expression was substantially decreased in anti- TGFβ-treated mice, CXCR5 expression was not (Figure 6B), indicating that TGFβ signaling is not essential for Tn, cell generation. Moreover, PNA expression on B cells was not affected either (Figure 6B).
[0067] To understand if Tf1, cell generation requires Th 17 function, we also immunized mice deficient in IL-17 or IL- 17F. Lack of IL-17 or IL- 17F did not significantly reduce the number of Tf11 cells in spleen or the number of germinal center B cells (Figure 6C, Figure 13B). IL-17 or IL- 17F is thus not essential in the generation of Tf11 cells in vivo.
EXAMPLE 6 IL-21 in the absence of TGFβ initiates Tf1, cell differentiation
[0068] Our results thus far suggest that although IL-6 and IL-21 are required for both Tfh and Th 17 differentiation, these two subsets appear to have distinct genetic program and differ in their dependency on TGFβ signaling. We next assessed whether IL-21 is sufficient to drive Tfh cell development in vitro in the absence of TGFβ signaling. Naϊve OT-II cells were activated by Ova peptide and splenic APC in the absence (neutral condition) or presence of IL-21, TGFβ and antibodies to IL-4 and IFNγ (Th 17 condition) or IL-21 plus antibodies to IL-4, IFNγ and TGFβ. 5 days later, the activated T cells were extensively washed and restimulated with anti-CD3 for 4 hours and their gene expression was assessed by real-time RT-PCR. As expected, cells cultured under Th 17 condition highly expressed TH17-specific genes, including genes encoding IL-17, IL-17F, IL-22, RORα and RORγt (Figure 7A). In contrast, T cells treated with IL-21 in the
absence of TGFβ signaling upregulated genes that are specifically expressed in Tβ, cells, including those encoding CXCR5, Bcl-6, IL-6R and IL-6st (Figure 7A). They also upregualted IL-21R expression but did not express Th 17 genes (Figure 7A). The gene encoding PD-I was also highly expressed in these cells (data not shown). To confirm this result, cell supernatants were measured 24 hours after restimulation for cytokine secretion by ELISA. IL-21 in the absence of TGFβ signaling did not support IL- 17 expression, but the resulting cells expressed high levels of IL-21 (Figure 7B), supporting that IL-21 expression is independent of TGFβ signaling.
[0069] To test the function of these Tfh-like cells in vivo, we transferred OT-II cells
(CD45.1+) activated under neutral conditions or with IL-21 plus blocking antibodies to IFNγ, IL- 4 and TGFβ into recipient mice (CD45.2+) followed by immunization with Ova protein. Compared to mice receiving no cells or T cells activated under neutral condition, the recipients of IL-21 -treated cells exhibited greatly increased CD45.1"1" Tf1, cells (Figure 7C). Interestingly, host T cells in these mice also had approximately 4 times Tfh cells compared to those receiving T cells activated under neutral condition (Figure 7C). The transferred cells also promoted Ova- specific antibody production and germinal center reactions (Figure 7D-F). As a control for the above experiment, we sorted CD45.1+ CXC5+CD44hl cells from KLH immunized mice and transferred them into recipient mice (CD45.2+) followed by immunization with KLH. Similar to in vitro generated Tfh cells, in vivo generated CXCR5+ cells also promoted a substantial increase in PNA+ germinal center B cells (Figure 7G). These results indicate that IL-21, in the absence of ThI, Th2 and Th 17 differentiation, drives Ta cell development.
[0070] As shown in Figure 1, C57BL/6 mice were immunized with KLH in CFA. Seven days later CD4+CD44hiCXCR5+ BTLA+ (Tn,) cells were sorted and restimulated with anti-CD3 for 4 hours for gene profiling analysis (A) and for real-time PCR analysis (B). OT-II T cells differentiated under various conditions were restimulated with anti-CD3 for 4 hours and were analyzed together with Tfh cells. (A). A distinct gene expression profile of Tfh cells from ThI, Th2, and Th 17 cells. Hierarchical clustering and PCA were applied to the expression levels of 8350 probe-sets showing differential expressions among the four types of cells (FDR = 0.1, corresponding to an unadjusted p-value = 0.033 from one-way ANOVA). (B). Real-time RT- PCR analysis of Th specific genes. (C-D). CD4+CD44hiCXCR5+ BTLA+ and CD4+CD44hiCXCR5' cells were restimulated with anti-CD3 plus anti-CD28 for 48 hours for cytokine measurement by ELISA (C) and with PMA and Ionomicin for 6 hours for detection of
IL-17 and IFNγ expression by intracellular cytokine staining (D). The data represent one of two independent experiments with similar results.
[0071] As shown in Figure 2, (A) CD4+ T cells from OT-II mice (CD45.2) were transferred into C57BL/6 (CD45.1) mice which were subsequently divided into 2 groups (3 mice per group). Mice were immunized subcutaneously with Ova protein emulsified in CFA and treated with a 300 μg of control rat Ig or anti-IFNγ and anti-IL-4 mAbs. Seven days after the immunization, experimental mice were sacrificed and splenic CD45.1+ and CD45.2+ CD4 cells were stained with biotinylated CXCR5 mAb, followed by APC-labeled streptavidin. Numbers in dot plot quadrants represent the percentages. CD44hiCXCR5+ and CD44MCXCR5" cells from immunized mice were purified and real-time RT-PCR analysis of Ta specific genes were performed. (B-C). IU''', Ifnγ1' (B), Stat6~'~ (C) and Stat4~A (D) and their appropriate controls (WT, 3 mice per group) were immunized with KLH emulsified in CFA. Seven days after the immunization, experimental mice were sacrificed and the germinal center B cells were determined by staining with FITC-labeled PNA and PerCP-labeled B220 mAb. The Tft cells were analyzed by staining with PerCP-labeled CD4 mAb and biotinylated CXCR5 mAb, followed by APC-labeled streptavidin. Numbers in dot plot quadrants represent the percentages. The experiments were repeated three times with consistent results.
[0072] As shown in Figure 3, (A) ICOSL germline deficient mice (JcosT') and their controls (WT, 3 mice per group) were immunized with KLH in CFA. Seven days after the immunization, experimental mice were sacrificed and spleen cells from immunized mice were stimulated in 96-well plates as triplicates with the indicated concentration of KLH. Effector cytokines (IFN -γ and IL-21) were measured after 4 days of treatment. The germinal center B cells and Tf]1 cells were analyzed. P values were calculated with the t-test by comparing the CXCR5+ cells and B220+PNA+ cells between wild-type and ICOSL deficient mice and are indicated as followed: * P < 0.005; #, P < 0.001. Numbers in dot plot quadrants represent the percentages. (B). Splenic B220+ B cells from ICOSL germline deficient mice (Icosl'''), B cell specific ICOSL deficient mice (ICOSL-BKO, cre+) and the ere- controls were analyzed for ICOSL expression. (C-E). Wild-type (WT) and B cell specific ICOSL deficient mice (3 mice per group) were immunized with KLH in CFA. Seven days after the immunization, experimental mice were sacrificed and analyzed as in A . P values were calculated with the t-test by comparing the CXCR5+ cells and B220+PNA+ cells between wild-type and B cell specific B7h deficient mice and are indicated as followed: ** P < 0.001 ; ##, P < 0.001. (C). (D). KLH specific antibodies (IgM and IgG) were measured in the sera by ELISA. The sera from WT and B cell
specific ICOSL deficient mice were subject to a 3-fold serial dilution, and the concentrations of KLH-specific IgM and IgG were analyzed by ELISA and averaged for each group. (E). GC in the spleens of KLH -immunized WT and B cell specific ICOSL deficient mice were identified by PNA staining (brown). T and B cells were identified by staining with anti-CD4 (red) and anti- B220 (blue). The data represent at least three independent experiments with consistent results.
[0073] As shown in Figure 4, I121+/+, Il21+/~ and 1121'1' mice (3 mice per group) were immunized subcutaneously with KLH emulsified in CFA. Seven days after the immunization, experimental mice were sacrificed and Ta cells (A) and the germinal center B cells (B) were analyzed. Numbers in dot plot quadrants represent the percentages. Germinal centers were determined by immunohistochemical analysis (C). Spleen cells from immunized mice were stimulated in 96-well plates as triplicates with the indicated concentration of KLH peptide. Proliferation was assayed after 3 days of treatment by adding [3H]thymidine to the culture for the last 8 h. IFN-γ was measured after 4 days of treatment. The experiments were repeated twice with consistent results.
[0074] As shown in Figure 5, I16'A (A) or T-cell specific Stat3'A mice (C) and their appropriate controls (WT, 3 mice per group) were immunized subcutaneously with KLH emulsified in CFA. Seven days after the immunization, experimental mice were sacrificed and Ta cells and the germinal center B cells were analyzed. Numbers in dot plot quadrants represent the percentages. (B) C57BL/6 mice were immunized with KLH in CFA. Seven days later, CD4+CD44hiCXCR5+ and CD4+CD44hiCXCR5" cells were sorted and restimulated with anti- CD3 and anti-CD28 with or without IL-6 or IL-21 for 48 hours. Proliferation was assayed by adding [3H]-thymidine to the culture for the last 8 h. P values were calculated with the t-test by comparing the CXCR5+ cells proliferation in the absence and in the presence of IL6 (*p<0.005) or in the absence and in the presence of IL21 (*p<0.005). CD4+CD44hiCXCR5+ and CD4+CD44hiCXCR5" cells were restimulated with anti-CD3 and anti-CD28 for 24 hours and stained with Annexin V-FITC. The experiments were performed two times with consistent results.
[0075] As shown in Figure 6, Ragl'1' reconstituted with WT and Rordtl$t/Rorγ ~'~ bone marrow cells (A) or IUT'' and Ill7f ~ mice and their controls (C) (3 mice per group) were immunized subcutaneously with KLH in CFA. Seven days after the immunization, experimental mice were sacrificed and the Tn1 cells were determined. Numbers in dot plot quadrants represent the percentages. (B). C57BL/6 mice were immunized with KLH emulsified in CFA with 100 μg of isotype control antibodies or TGFβ blocking antibodies (3 mice per group). Seven days later,
experimental mice were sacrificed and splenic Ta cells and germinal center B cells were analyzed. Splenocytes were restimulated with KLH for overnight and the production of IL- 17 and IFNγ was analyzed in CD4+ gate by intracellular cytokine staining. The results represent one of three individuals with similar results.
[0076] As shown in Figure 7, A-F, FACS-sorted CD62hiCD44IoCD25negCD4+ T cells from CD45.1+ OT-II mice were cultured with irradiated splenic APC plus OVA323-339 peptide under ThO, Tfh (IL-21 plus antibodies to IL-4, IFNγ and TGFβ) or TH 17 condition for 5 days. After 5 days, CD4+ T cells were restimulated with anti-CD3 for 4 hours for real-time PCR analysis (A) or for 24 hours for cytokine measurement by ELISA (B). (C-F). Five days after in vitro differentiation, cells were adoptively transferred into CD45.2+ congenic mice (n=3-4) before the recipient mice were subcutaneously immunized with OVA in IFA. A group of mice that did not receive T cells was used as a control (No transfer). Seven days after immunzation, lymphoid cells from the draining lymph nodes of recipient mice were isolated and Tg1 cells and germinal center B cells were analyzed (D). Numbers in the boxes represent the percentages. (E). The sera from the recipient mice were subject to a 3-fold serial dilution, and the concentrations of OVA-specific IgM and IgG were analyzed by ELISA and averaged for each group. (F). Germinal center in the spleens of the recipient mice were identified by PNA staining (brown). The results are a representative of multiple mice of two independent experiments with similar results. (G). CXCR5+CD44hl cells were sorted from CD45.1 mice on day 7 after immunization with KLH and transferred to CD45.2 (n=3) recipient mice following immunization with KLH in CFA. A group of mice that did not receive T cells was used as a control (No transfer). Seven days after immunization, germinal center in the spleens of the recipient mice were identified by PNA staining (brown).
[0077] As shown in Figure 8, follicular helper T cells co-express CXCR5 and BTLA.
C57BL/6 mice were immunized with KLH in CFA. Seven days after the immunization, experimental mice were sacrificed and the Tf1, cell induction was determined by staining CD4 cells with PE-labeled anti-BTLA mAb and biotinylated anti-CXCR5 mAb, followed by APC-labeled streptavidin.
[0078] As shown in Figure 9, FDR estimated from ANOVA tests using resampled samples.
[0079] As shown in Figure 10, antigen-specific cytokine production by Tf]1 cells.
C57BL/6 mice were immunized with KLH in CFA. Seven days later CD4+CD44h'CXCR5+ (Tft)
and CD4+CD44hiCXCR5~ (non-Ta) cells were sorted and restimulated with KLH and irradiated APC for 48 hours for cytokine measurement by ELISA.
[0080] As shown in Figure 11, Tfh cell generation is independent of THl and TH2 cells.
(A-C). Basal levels of CXCR5 expression in non-immunized IL-4 KO, IFNγ KO, STAT6 KO and STAT4 KO mice were analyzed by staining with PerCP-labeled anti-CD4 mAb and biotinylated anti-CXCR5 mAb, followed by APC-labeled streptavidin. Numbers in dot plot quadrants represent the percentages. (D). IL-4 KO, IFNγ KO and their appropriate controls (WT, 3 mice per group) were immunized with KLH emulsified in CFA. Seven days after the immunization, CD4+CD44hlCXCR5+ (Tft) and CD4+CD44hiCXCR5" (non-Ta) cells were sorted and restimulated with anti-CD3 for 4 hours for real-time RT-PCR analysis of TH subset-specific genes.
[0081] As shown in Figure 12, regulation of antibody responses by STAT3. T cell-specific STAT3 KO and their controls (WT, 3 mice per group) were immunized with KLH in CFA. Seven days after the immunization, Anti-KLH antibodies (IgM and IgG) were measured in the sera by ELISA. The sera from WT and KO mice were subject to a 3-fold serial dilution, and the concentrations of KLH-specific IgM and IgG were analyzed by ELISA and averaged for each group.
[0082] As shown in Figure 13, normal germinal center reaction in IL-17 KO, IL-17F KO and RORα/γ KO mice. IL-17 KO, IL-17F KO (A), RORα/γ KO (B) mice and their controls (WT, 3 mice per group) were immunized with KLH in CFA. Seven days after the immunization, germinal centers in the spleens of KLH-immunized mice were identified by PNA staining (brown).
[0083] As shown in Figure 14, schematic demonstration that development of Tfh cells, independent of TH 1 , TH2 or TH 17 lineages, is regulated by IL-6 and IL-21.
Claims
1. A method of treating a disease or condition associated with TFH cell-mediated immune response in a subject in need thereof comprising administering to the subject an agent that modulates the IL-6, IL-21, and/or STAT3 signaling pathway, wherein the agent is administered in an amount effective to modulate the differentiation of TFH cells.
2. The method of claim 1 wherein the agent is an antagonist of one or more of IL-6, IL-21, and STAT3.
3. The method of claim 1 wherein the agent is an agonist of one or more of IL-6, IL- 21, and STAT3.
4. A method of producing a Tfh cell comprising the steps of: providing an activated T cell; and contacting said cell with IL-2, IL-21 or STAT3, wherein said T cell differentiates into one or more Ta cells.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US7782108P | 2008-07-02 | 2008-07-02 | |
US61/077,821 | 2008-07-02 |
Publications (3)
Publication Number | Publication Date |
---|---|
WO2010003002A2 WO2010003002A2 (en) | 2010-01-07 |
WO2010003002A3 WO2010003002A3 (en) | 2010-04-22 |
WO2010003002A9 true WO2010003002A9 (en) | 2010-05-20 |
Family
ID=41466595
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2009/049421 WO2010003002A2 (en) | 2008-07-02 | 2009-07-01 | Modulation of follicular helper t cells |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2010003002A2 (en) |
Families Citing this family (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012063875A1 (en) | 2010-11-11 | 2012-05-18 | シスメックス株式会社 | Markers for detecting human follicular helper t cells and method for detecting human follicular helper t cells |
WO2013063019A1 (en) | 2011-10-28 | 2013-05-02 | The Wistar Institute Of Anatomy And Biology | Methods and compositions for enhancing the therapeutic effect of anti-tumor t cells |
US20150118257A1 (en) * | 2012-04-20 | 2015-04-30 | The Wistar Institute Of Anatomy And Biology | Methods and Compositions for Manipulating the Immune System |
WO2014074852A1 (en) * | 2012-11-09 | 2014-05-15 | President And Fellows Of Harvard College | Compositions and methods for modulating an immune response |
AU2013204922B2 (en) | 2012-12-20 | 2015-05-14 | Celgene Corporation | Chimeric antigen receptors |
EP2970372B1 (en) | 2013-03-15 | 2020-09-30 | Celgene Corporation | Modified t lymphocytes |
CN106795497A (en) * | 2014-08-12 | 2017-05-31 | 人类起源公司 | It is engineered to go back to the nest to the CAR T lymphocytes of lymph node B cells area, skin or intestines and stomach |
WO2017091729A1 (en) * | 2015-11-23 | 2017-06-01 | President And Fellows Of Harvard College | Compositions and methods for modulating an immune response |
-
2009
- 2009-07-01 WO PCT/US2009/049421 patent/WO2010003002A2/en active Application Filing
Also Published As
Publication number | Publication date |
---|---|
WO2010003002A2 (en) | 2010-01-07 |
WO2010003002A3 (en) | 2010-04-22 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Nurieva et al. | Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages | |
WO2010003002A9 (en) | Modulation of follicular helper t cells | |
Yamazaki et al. | CCR6 regulates the migration of inflammatory and regulatory T cells | |
Linton et al. | A critical role for B cells in the development of memory CD4 cells | |
Ito et al. | TLR9 regulates the mycobacteria-elicited pulmonary granulomatous immune response in mice through DC-derived Notch ligand delta-like 4 | |
Salam et al. | T cell ageing: effects of age on development, survival & function | |
Boniface et al. | From interleukin‐23 to T‐helper 17 cells: human T‐helper cell differentiation revisited | |
Staudt et al. | Interferon-regulatory factor 4 is essential for the developmental program of T helper 9 cells | |
Vieira et al. | IL-10-secreting regulatory T cells do not express Foxp3 but have comparable regulatory function to naturally occurring CD4+ CD25+ regulatory T cells | |
Linton et al. | Intrinsic versus environmental influences on T‐cell responses in aging | |
Yong et al. | Role and mechanisms of CD4+ CD25+ regulatory T cells in the induction and maintenance of transplantation tolerance | |
WO2006007486A2 (en) | COMPOSITIONS AND METHODS FOR MODULATION OF RORϜt | |
Reinhard et al. | c‐Rel promotes type 1 and type 17 immune responses during Leishmania major infection | |
Maiti et al. | Immunosuppression of rat myasthenia gravis by oral administration of a syngeneic acetylcholine receptor fragment | |
Frasca et al. | Aging impairs murine B cell differentiation and function in primary and secondary lymphoid tissues | |
JP6258922B2 (en) | Production and use of regulatory B cells | |
US20100247547A1 (en) | Modulation of the th-17 cell mediated immune responses | |
Zhivaki et al. | Correction of age-associated defects in dendritic cells enables CD4+ T cells to eradicate tumors | |
US20110044939A1 (en) | Regulatory t cells in adipose tissue | |
Olalekan et al. | Tissue specific CD4+ T cell priming determines the requirement for interleukin-23 in experimental arthritis | |
JP2004208548A (en) | Antigen-specific suppression of immunoreaction | |
Tatsumi et al. | Rapid activation of IL-2 receptor signaling by CD301b+ DC-derived IL-2 dictates the outcome of helper T cell differentiation | |
Mathieu et al. | Inflammation enhances the vaccination potential of CD40‐activated B cells in mice | |
Bailey | Role of the Akt signalling pathway in CD8 T cell immune response | |
Hansson | Unravelling the mechanisms for mucosal tolerance induction using the CTA1R7K-X-DD immunomodulating fusion protein |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 09774452 Country of ref document: EP Kind code of ref document: A2 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 09774452 Country of ref document: EP Kind code of ref document: A2 |