WO2007070856A2 - Method for treating immune dysfunction by regulation of cd40 ligand expression - Google Patents
Method for treating immune dysfunction by regulation of cd40 ligand expression Download PDFInfo
- Publication number
- WO2007070856A2 WO2007070856A2 PCT/US2006/062111 US2006062111W WO2007070856A2 WO 2007070856 A2 WO2007070856 A2 WO 2007070856A2 US 2006062111 W US2006062111 W US 2006062111W WO 2007070856 A2 WO2007070856 A2 WO 2007070856A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- tfeb
- tfe3
- cells
- tdn
- expression
- Prior art date
Links
- 230000014509 gene expression Effects 0.000 title claims abstract description 142
- 238000000034 method Methods 0.000 title claims abstract description 44
- 208000026278 immune system disease Diseases 0.000 title claims abstract description 21
- 230000033228 biological regulation Effects 0.000 title abstract description 14
- 239000003446 ligand Substances 0.000 title description 2
- 102100028502 Transcription factor EB Human genes 0.000 claims abstract description 232
- 101000837845 Homo sapiens Transcription factor E3 Proteins 0.000 claims abstract description 213
- 102100028507 Transcription factor E3 Human genes 0.000 claims abstract description 209
- 102100032937 CD40 ligand Human genes 0.000 claims abstract description 169
- 108010029697 CD40 Ligand Proteins 0.000 claims abstract description 159
- 239000003112 inhibitor Substances 0.000 claims abstract description 29
- 101000837841 Homo sapiens Transcription factor EB Proteins 0.000 claims abstract 35
- 108090000623 proteins and genes Proteins 0.000 claims description 84
- 102000004169 proteins and genes Human genes 0.000 claims description 56
- 230000027455 binding Effects 0.000 claims description 35
- 238000009739 binding Methods 0.000 claims description 34
- 230000000903 blocking effect Effects 0.000 claims description 30
- 238000013518 transcription Methods 0.000 claims description 20
- 230000035897 transcription Effects 0.000 claims description 20
- 108020004414 DNA Proteins 0.000 claims description 16
- 230000015572 biosynthetic process Effects 0.000 claims description 14
- 230000002452 interceptive effect Effects 0.000 claims description 13
- 239000000539 dimer Substances 0.000 claims description 12
- 230000002103 transcriptional effect Effects 0.000 claims description 12
- 208000027866 inflammatory disease Diseases 0.000 claims description 10
- 230000003993 interaction Effects 0.000 claims description 10
- 238000003786 synthesis reaction Methods 0.000 claims description 9
- 208000023275 Autoimmune disease Diseases 0.000 claims description 5
- 230000000415 inactivating effect Effects 0.000 claims description 5
- 206010061218 Inflammation Diseases 0.000 claims description 4
- 230000004054 inflammatory process Effects 0.000 claims description 4
- 230000002401 inhibitory effect Effects 0.000 claims description 4
- 230000026731 phosphorylation Effects 0.000 claims description 4
- 238000006366 phosphorylation reaction Methods 0.000 claims description 4
- 210000001744 T-lymphocyte Anatomy 0.000 description 213
- 101710162524 Transcription factor EB Proteins 0.000 description 197
- 210000004027 cell Anatomy 0.000 description 88
- 101000716102 Homo sapiens T-cell surface glycoprotein CD4 Proteins 0.000 description 81
- 102100036011 T-cell surface glycoprotein CD4 Human genes 0.000 description 81
- 210000003719 b-lymphocyte Anatomy 0.000 description 67
- 241000699670 Mus sp. Species 0.000 description 58
- 230000000694 effects Effects 0.000 description 55
- 238000011830 transgenic mouse model Methods 0.000 description 51
- 101150093750 CD40LG gene Proteins 0.000 description 42
- 241000282414 Homo sapiens Species 0.000 description 34
- 241000699666 Mus <mouse, genus> Species 0.000 description 34
- 230000000638 stimulation Effects 0.000 description 33
- 108091008874 T cell receptors Proteins 0.000 description 31
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 31
- 238000000684 flow cytometry Methods 0.000 description 30
- 230000001419 dependent effect Effects 0.000 description 29
- 230000006698 induction Effects 0.000 description 25
- 238000003119 immunoblot Methods 0.000 description 23
- 230000004044 response Effects 0.000 description 23
- 230000003393 splenic effect Effects 0.000 description 23
- 239000000284 extract Substances 0.000 description 22
- 230000004913 activation Effects 0.000 description 21
- 101150087532 mitF gene Proteins 0.000 description 21
- 101150013553 CD40 gene Proteins 0.000 description 20
- 102100040245 Tumor necrosis factor receptor superfamily member 5 Human genes 0.000 description 20
- 239000000427 antigen Substances 0.000 description 20
- 108091007433 antigens Proteins 0.000 description 20
- 102000036639 antigens Human genes 0.000 description 20
- 230000009261 transgenic effect Effects 0.000 description 20
- 238000004458 analytical method Methods 0.000 description 19
- 238000002487 chromatin immunoprecipitation Methods 0.000 description 19
- 239000013612 plasmid Substances 0.000 description 19
- 102000040945 Transcription factor Human genes 0.000 description 18
- 108091023040 Transcription factor Proteins 0.000 description 18
- 210000000952 spleen Anatomy 0.000 description 18
- 102000017420 CD3 protein, epsilon/gamma/delta subunit Human genes 0.000 description 17
- 108050005493 CD3 protein, epsilon/gamma/delta subunit Proteins 0.000 description 17
- 230000007812 deficiency Effects 0.000 description 17
- 238000002474 experimental method Methods 0.000 description 17
- 230000002950 deficient Effects 0.000 description 16
- 229920002477 rna polymer Polymers 0.000 description 16
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 description 15
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 description 15
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 description 15
- 102100027268 Interferon-stimulated gene 20 kDa protein Human genes 0.000 description 15
- 108091034117 Oligonucleotide Proteins 0.000 description 15
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 description 15
- 238000002337 electrophoretic mobility shift assay Methods 0.000 description 15
- 239000012634 fragment Substances 0.000 description 15
- 230000001771 impaired effect Effects 0.000 description 15
- 102000053602 DNA Human genes 0.000 description 14
- 241000699660 Mus musculus Species 0.000 description 14
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 14
- PHEDXBVPIONUQT-RGYGYFBISA-N phorbol 13-acetate 12-myristate Chemical compound C([C@]1(O)C(=O)C(C)=C[C@H]1[C@@]1(O)[C@H](C)[C@H]2OC(=O)CCCCCCCCCCCCC)C(CO)=C[C@H]1[C@H]1[C@]2(OC(C)=O)C1(C)C PHEDXBVPIONUQT-RGYGYFBISA-N 0.000 description 14
- 101000868215 Homo sapiens CD40 ligand Proteins 0.000 description 13
- 230000006870 function Effects 0.000 description 13
- 229940027941 immunoglobulin g Drugs 0.000 description 13
- 108091035710 E-box Proteins 0.000 description 11
- 230000002159 abnormal effect Effects 0.000 description 11
- 230000005875 antibody response Effects 0.000 description 11
- 230000011712 cell development Effects 0.000 description 11
- 239000003623 enhancer Substances 0.000 description 11
- 238000001727 in vivo Methods 0.000 description 11
- 210000004698 lymphocyte Anatomy 0.000 description 11
- 102000004388 Interleukin-4 Human genes 0.000 description 10
- 108090000978 Interleukin-4 Proteins 0.000 description 10
- 108010018525 NFATC Transcription Factors Proteins 0.000 description 10
- 102000002673 NFATC Transcription Factors Human genes 0.000 description 10
- 201000010099 disease Diseases 0.000 description 10
- 108020004999 messenger RNA Proteins 0.000 description 10
- 230000001105 regulatory effect Effects 0.000 description 10
- 230000004568 DNA-binding Effects 0.000 description 9
- 108010002350 Interleukin-2 Proteins 0.000 description 9
- 102000000588 Interleukin-2 Human genes 0.000 description 9
- 239000005089 Luciferase Substances 0.000 description 9
- 108700019146 Transgenes Proteins 0.000 description 9
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 9
- 238000003556 assay Methods 0.000 description 9
- 230000007547 defect Effects 0.000 description 9
- 244000052769 pathogen Species 0.000 description 9
- 230000037361 pathway Effects 0.000 description 9
- 210000004988 splenocyte Anatomy 0.000 description 9
- 238000010186 staining Methods 0.000 description 9
- 239000013598 vector Substances 0.000 description 9
- 108060001084 Luciferase Proteins 0.000 description 8
- 210000001280 germinal center Anatomy 0.000 description 8
- 239000000185 hemagglutinin Substances 0.000 description 8
- 229940028885 interleukin-4 Drugs 0.000 description 8
- 230000035772 mutation Effects 0.000 description 8
- 206010039073 rheumatoid arthritis Diseases 0.000 description 8
- 210000001541 thymus gland Anatomy 0.000 description 8
- 101710154606 Hemagglutinin Proteins 0.000 description 7
- 241000713666 Lentivirus Species 0.000 description 7
- 101710093908 Outer capsid protein VP4 Proteins 0.000 description 7
- 101710135467 Outer capsid protein sigma-1 Proteins 0.000 description 7
- 108010046016 Peanut Agglutinin Proteins 0.000 description 7
- 101710176177 Protein A56 Proteins 0.000 description 7
- 210000001185 bone marrow Anatomy 0.000 description 7
- 238000009826 distribution Methods 0.000 description 7
- 239000012636 effector Substances 0.000 description 7
- 230000003053 immunization Effects 0.000 description 7
- 238000002649 immunization Methods 0.000 description 7
- 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 6
- 108020004635 Complementary DNA Proteins 0.000 description 6
- 102100034343 Integrase Human genes 0.000 description 6
- 102000043136 MAP kinase family Human genes 0.000 description 6
- 108091054455 MAP kinase family Proteins 0.000 description 6
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 6
- 239000012190 activator Substances 0.000 description 6
- 238000010804 cDNA synthesis Methods 0.000 description 6
- 239000002299 complementary DNA Substances 0.000 description 6
- 239000005090 green fluorescent protein Substances 0.000 description 6
- 230000002779 inactivation Effects 0.000 description 6
- 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 6
- 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 6
- 230000001404 mediated effect Effects 0.000 description 6
- 230000002829 reductive effect Effects 0.000 description 6
- 230000000284 resting effect Effects 0.000 description 6
- 239000000523 sample Substances 0.000 description 6
- 230000028327 secretion Effects 0.000 description 6
- 210000002966 serum Anatomy 0.000 description 6
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 6
- 238000011144 upstream manufacturing Methods 0.000 description 6
- 108700031361 Brachyury Proteins 0.000 description 5
- 241000282412 Homo Species 0.000 description 5
- 241000700159 Rattus Species 0.000 description 5
- 241000242739 Renilla Species 0.000 description 5
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 5
- 239000011324 bead Substances 0.000 description 5
- 230000003915 cell function Effects 0.000 description 5
- 230000001276 controlling effect Effects 0.000 description 5
- 238000011161 development Methods 0.000 description 5
- 230000018109 developmental process Effects 0.000 description 5
- 230000009368 gene silencing by RNA Effects 0.000 description 5
- 230000012178 germinal center formation Effects 0.000 description 5
- 210000000987 immune system Anatomy 0.000 description 5
- 230000005764 inhibitory process Effects 0.000 description 5
- 230000007246 mechanism Effects 0.000 description 5
- OXNIZHLAWKMVMX-UHFFFAOYSA-N picric acid Chemical compound OC1=C([N+]([O-])=O)C=C([N+]([O-])=O)C=C1[N+]([O-])=O OXNIZHLAWKMVMX-UHFFFAOYSA-N 0.000 description 5
- 102000005962 receptors Human genes 0.000 description 5
- 108020003175 receptors Proteins 0.000 description 5
- 208000011580 syndromic disease Diseases 0.000 description 5
- 229950002929 trinitrophenol Drugs 0.000 description 5
- 229920001917 Ficoll Polymers 0.000 description 4
- 108090000331 Firefly luciferases Proteins 0.000 description 4
- 101100059511 Homo sapiens CD40LG gene Proteins 0.000 description 4
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 4
- 108700018351 Major Histocompatibility Complex Proteins 0.000 description 4
- 101100059514 Mus musculus Cd40lg gene Proteins 0.000 description 4
- 101100536761 Mus musculus Tfe3 gene Proteins 0.000 description 4
- 101100536763 Mus musculus Tfeb gene Proteins 0.000 description 4
- 108700008625 Reporter Genes Proteins 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 230000006044 T cell activation Effects 0.000 description 4
- 108010018242 Transcription Factor AP-1 Proteins 0.000 description 4
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 4
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 4
- 230000001413 cellular effect Effects 0.000 description 4
- 230000006735 deficit Effects 0.000 description 4
- 208000035475 disorder Diseases 0.000 description 4
- 210000004602 germ cell Anatomy 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 238000011534 incubation Methods 0.000 description 4
- 238000011068 loading method Methods 0.000 description 4
- 206010025135 lupus erythematosus Diseases 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 239000002245 particle Substances 0.000 description 4
- 230000001717 pathogenic effect Effects 0.000 description 4
- 102000013415 peroxidase activity proteins Human genes 0.000 description 4
- 108040007629 peroxidase activity proteins Proteins 0.000 description 4
- 210000004180 plasmocyte Anatomy 0.000 description 4
- 230000004043 responsiveness Effects 0.000 description 4
- 230000004936 stimulating effect Effects 0.000 description 4
- 230000020382 suppression by virus of host antigen processing and presentation of peptide antigen via MHC class I Effects 0.000 description 4
- 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 4
- 230000003612 virological effect Effects 0.000 description 4
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 4
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 3
- 201000007155 CD40 ligand deficiency Diseases 0.000 description 3
- 108700011215 E-Box Elements Proteins 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 206010020464 Humoral immune defect Diseases 0.000 description 3
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 3
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 3
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 3
- 108091054438 MHC class II family Proteins 0.000 description 3
- 102000043131 MHC class II family Human genes 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 102100030608 Mothers against decapentaplegic homolog 7 Human genes 0.000 description 3
- 241001529936 Murinae Species 0.000 description 3
- 108010014608 Proto-Oncogene Proteins c-kit Proteins 0.000 description 3
- 102000016971 Proto-Oncogene Proteins c-kit Human genes 0.000 description 3
- 108091030071 RNAI Proteins 0.000 description 3
- 101700026522 SMAD7 Proteins 0.000 description 3
- -1 TL-2 Proteins 0.000 description 3
- 102100023132 Transcription factor Jun Human genes 0.000 description 3
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 3
- 102100040247 Tumor necrosis factor Human genes 0.000 description 3
- 108700005077 Viral Genes Proteins 0.000 description 3
- 241000700605 Viruses Species 0.000 description 3
- 201000001696 X-linked hyper IgM syndrome Diseases 0.000 description 3
- 239000000556 agonist Substances 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- 229960002685 biotin Drugs 0.000 description 3
- 235000020958 biotin Nutrition 0.000 description 3
- 239000011616 biotin Substances 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 210000001772 blood platelet Anatomy 0.000 description 3
- 210000004970 cd4 cell Anatomy 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- 230000008859 change Effects 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 210000002889 endothelial cell Anatomy 0.000 description 3
- 239000013604 expression vector Substances 0.000 description 3
- 239000012091 fetal bovine serum Substances 0.000 description 3
- 238000001415 gene therapy Methods 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 239000001963 growth medium Substances 0.000 description 3
- 238000003306 harvesting Methods 0.000 description 3
- 210000002443 helper t lymphocyte Anatomy 0.000 description 3
- 230000008348 humoral response Effects 0.000 description 3
- 208000026095 hyper-IgM syndrome type 1 Diseases 0.000 description 3
- 230000028993 immune response Effects 0.000 description 3
- 238000003670 luciferase enzyme activity assay Methods 0.000 description 3
- 238000003752 polymerase chain reaction Methods 0.000 description 3
- 230000001124 posttranscriptional effect Effects 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 230000035755 proliferation Effects 0.000 description 3
- 238000003753 real-time PCR Methods 0.000 description 3
- 238000012755 real-time RT-PCR analysis Methods 0.000 description 3
- 241000894007 species Species 0.000 description 3
- 230000009885 systemic effect Effects 0.000 description 3
- 210000001519 tissue Anatomy 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- 229920000936 Agarose Polymers 0.000 description 2
- 102100039341 Atrial natriuretic peptide receptor 2 Human genes 0.000 description 2
- 101710102159 Atrial natriuretic peptide receptor 2 Proteins 0.000 description 2
- 108090001008 Avidin Proteins 0.000 description 2
- 230000003844 B-cell-activation Effects 0.000 description 2
- 102000000905 Cadherin Human genes 0.000 description 2
- 108050007957 Cadherin Proteins 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 102000004127 Cytokines Human genes 0.000 description 2
- 108090000695 Cytokines Proteins 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- 102100025137 Early activation antigen CD69 Human genes 0.000 description 2
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 2
- 108700039691 Genetic Promoter Regions Proteins 0.000 description 2
- 101000934374 Homo sapiens Early activation antigen CD69 Proteins 0.000 description 2
- 101000928479 Homo sapiens Microtubule organization protein AKNA Proteins 0.000 description 2
- 208000003352 Hyper-IgM Immunodeficiency Syndrome Diseases 0.000 description 2
- 108060003951 Immunoglobulin Proteins 0.000 description 2
- 102100020985 Immunoglobulin mu heavy chain Human genes 0.000 description 2
- 101710099741 Immunoglobulin mu heavy chain Proteins 0.000 description 2
- 102000015696 Interleukins Human genes 0.000 description 2
- 108010063738 Interleukins Proteins 0.000 description 2
- 102000004058 Leukemia inhibitory factor Human genes 0.000 description 2
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 2
- 108010064548 Lymphocyte Function-Associated Antigen-1 Proteins 0.000 description 2
- 208000030289 Lymphoproliferative disease Diseases 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 102100025748 Mothers against decapentaplegic homolog 3 Human genes 0.000 description 2
- 101710143111 Mothers against decapentaplegic homolog 3 Proteins 0.000 description 2
- 101100444898 Mus musculus Egr1 gene Proteins 0.000 description 2
- 101001043827 Mus musculus Interleukin-2 Proteins 0.000 description 2
- 102100038895 Myc proto-oncogene protein Human genes 0.000 description 2
- 101710135898 Myc proto-oncogene protein Proteins 0.000 description 2
- 206010028980 Neoplasm Diseases 0.000 description 2
- 102100034400 Nuclear factor of activated T-cells, cytoplasmic 2 Human genes 0.000 description 2
- 101710151538 Nuclear factor of activated T-cells, cytoplasmic 2 Proteins 0.000 description 2
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 2
- 108010029485 Protein Isoforms Proteins 0.000 description 2
- 102000001708 Protein Isoforms Human genes 0.000 description 2
- 102000001253 Protein Kinase Human genes 0.000 description 2
- 206010038389 Renal cancer Diseases 0.000 description 2
- 208000006265 Renal cell carcinoma Diseases 0.000 description 2
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N Sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 2
- 102000015215 Stem Cell Factor Human genes 0.000 description 2
- 108010039445 Stem Cell Factor Proteins 0.000 description 2
- 101710150448 Transcriptional regulator Myc Proteins 0.000 description 2
- 239000007983 Tris buffer Substances 0.000 description 2
- 208000006391 Type 1 Hyper-IgM Immunodeficiency Syndrome Diseases 0.000 description 2
- 230000005856 abnormality Effects 0.000 description 2
- 230000033289 adaptive immune response Effects 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 230000001363 autoimmune Effects 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 230000036755 cellular response Effects 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 238000010276 construction Methods 0.000 description 2
- 230000010250 cytokine signaling pathway Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 238000006471 dimerization reaction Methods 0.000 description 2
- 230000003828 downregulation Effects 0.000 description 2
- 231100001129 embryonic lethality Toxicity 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 239000012894 fetal calf serum Substances 0.000 description 2
- 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 2
- 238000001502 gel electrophoresis Methods 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 230000023597 hemostasis Effects 0.000 description 2
- 210000003630 histaminocyte Anatomy 0.000 description 2
- 206010066130 hyper-IgM syndrome Diseases 0.000 description 2
- 210000002865 immune cell Anatomy 0.000 description 2
- 230000001900 immune effect Effects 0.000 description 2
- 230000003832 immune regulation Effects 0.000 description 2
- 230000036039 immunity Effects 0.000 description 2
- 102000018358 immunoglobulin Human genes 0.000 description 2
- 239000012133 immunoprecipitate Substances 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 230000015788 innate immune response Effects 0.000 description 2
- 229940047122 interleukins Drugs 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 238000003468 luciferase reporter gene assay Methods 0.000 description 2
- 210000005210 lymphoid organ Anatomy 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 210000002752 melanocyte Anatomy 0.000 description 2
- 239000003226 mitogen Substances 0.000 description 2
- 210000001616 monocyte Anatomy 0.000 description 2
- 238000002703 mutagenesis Methods 0.000 description 2
- 231100000350 mutagenesis Toxicity 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 2
- 210000001539 phagocyte Anatomy 0.000 description 2
- 210000004986 primary T-cell Anatomy 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 238000002331 protein detection Methods 0.000 description 2
- 108060006633 protein kinase Proteins 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 201000010174 renal carcinoma Diseases 0.000 description 2
- 238000003571 reporter gene assay Methods 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 238000004448 titration Methods 0.000 description 2
- 238000003146 transient transfection Methods 0.000 description 2
- 230000014616 translation Effects 0.000 description 2
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- BHNQPLPANNDEGL-UHFFFAOYSA-N 2-(4-octylphenoxy)ethanol Chemical compound CCCCCCCCC1=CC=C(OCCO)C=C1 BHNQPLPANNDEGL-UHFFFAOYSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- XMTQQYYKAHVGBJ-UHFFFAOYSA-N 3-(3,4-DICHLOROPHENYL)-1,1-DIMETHYLUREA Chemical compound CN(C)C(=O)NC1=CC=C(Cl)C(Cl)=C1 XMTQQYYKAHVGBJ-UHFFFAOYSA-N 0.000 description 1
- YRNWIFYIFSBPAU-UHFFFAOYSA-N 4-[4-(dimethylamino)phenyl]-n,n-dimethylaniline Chemical compound C1=CC(N(C)C)=CC=C1C1=CC=C(N(C)C)C=C1 YRNWIFYIFSBPAU-UHFFFAOYSA-N 0.000 description 1
- HRPVXLWXLXDGHG-UHFFFAOYSA-N Acrylamide Chemical compound NC(=O)C=C HRPVXLWXLXDGHG-UHFFFAOYSA-N 0.000 description 1
- 108700028369 Alleles Proteins 0.000 description 1
- 102100021631 B-cell lymphoma 6 protein Human genes 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 238000011740 C57BL/6 mouse Methods 0.000 description 1
- 101100297347 Caenorhabditis elegans pgl-3 gene Proteins 0.000 description 1
- 102100035933 Calcium-responsive transcription factor Human genes 0.000 description 1
- 101710116997 Calcium-responsive transcription factor Proteins 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 108091035707 Consensus sequence Proteins 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- 101710088194 Dehydrogenase Proteins 0.000 description 1
- 102000007260 Deoxyribonuclease I Human genes 0.000 description 1
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 1
- 238000003718 Dual-Luciferase Reporter Assay System Methods 0.000 description 1
- 102100040068 E3 ubiquitin-protein ligase TRIM37 Human genes 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 101000759376 Escherichia phage Mu Tail sheath protein Proteins 0.000 description 1
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 1
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 1
- 108010087819 Fc receptors Proteins 0.000 description 1
- 102000009109 Fc receptors Human genes 0.000 description 1
- 108091006027 G proteins Proteins 0.000 description 1
- 102000030782 GTP binding Human genes 0.000 description 1
- 108091000058 GTP-Binding Proteins 0.000 description 1
- 101001066288 Gallus gallus GATA-binding factor 3 Proteins 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 229920002527 Glycogen Polymers 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 108010017213 Granulocyte-Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 102100039620 Granulocyte-macrophage colony-stimulating factor Human genes 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 1
- HVLSXIKZNLPZJJ-TXZCQADKSA-N HA peptide Chemical compound C([C@@H](C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](C(C)C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](C)C(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=C(O)C=C1 HVLSXIKZNLPZJJ-TXZCQADKSA-N 0.000 description 1
- 101000971234 Homo sapiens B-cell lymphoma 6 protein Proteins 0.000 description 1
- 101100005713 Homo sapiens CD4 gene Proteins 0.000 description 1
- 101000610400 Homo sapiens E3 ubiquitin-protein ligase TRIM37 Proteins 0.000 description 1
- 101000608935 Homo sapiens Leukosialin Proteins 0.000 description 1
- 101000597273 Homo sapiens PHD finger protein 11 Proteins 0.000 description 1
- 101000935642 Homo sapiens Phosphoinositide 3-kinase adapter protein 1 Proteins 0.000 description 1
- 101000934346 Homo sapiens T-cell surface antigen CD2 Proteins 0.000 description 1
- 101000946860 Homo sapiens T-cell surface glycoprotein CD3 epsilon chain Proteins 0.000 description 1
- 101000946843 Homo sapiens T-cell surface glycoprotein CD8 alpha chain Proteins 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 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
- 102000004556 Interleukin-15 Receptors Human genes 0.000 description 1
- 108010017535 Interleukin-15 Receptors Proteins 0.000 description 1
- 241000254158 Lampyridae Species 0.000 description 1
- 102100039564 Leukosialin Human genes 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 1
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 1
- 108010058398 Macrophage Colony-Stimulating Factor Receptor Proteins 0.000 description 1
- 102100027754 Mast/stem cell growth factor receptor Kit Human genes 0.000 description 1
- 208000009795 Microphthalmos Diseases 0.000 description 1
- 102100036470 Microtubule organization protein AKNA Human genes 0.000 description 1
- 101000868216 Mus musculus CD40 ligand Proteins 0.000 description 1
- 101001002703 Mus musculus Interleukin-4 Proteins 0.000 description 1
- 108010057466 NF-kappa B Proteins 0.000 description 1
- 102000003945 NF-kappa B Human genes 0.000 description 1
- 108700019961 Neoplasm Genes Proteins 0.000 description 1
- 102000048850 Neoplasm Genes Human genes 0.000 description 1
- 108010077850 Nuclear Localization Signals Proteins 0.000 description 1
- 108091028043 Nucleic acid sequence Proteins 0.000 description 1
- 208000001388 Opportunistic Infections Diseases 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 102100035126 PHD finger protein 11 Human genes 0.000 description 1
- 101710084414 POU domain, class 2, transcription factor 1 Proteins 0.000 description 1
- 102100035593 POU domain, class 2, transcription factor 1 Human genes 0.000 description 1
- 239000002033 PVDF binder Substances 0.000 description 1
- 241000237988 Patellidae Species 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 101710150344 Protein Rev Proteins 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 1
- 238000010240 RT-PCR analysis Methods 0.000 description 1
- 102000004278 Receptor Protein-Tyrosine Kinases Human genes 0.000 description 1
- 108090000873 Receptor Protein-Tyrosine Kinases Proteins 0.000 description 1
- 108010052090 Renilla Luciferases Proteins 0.000 description 1
- 108091027981 Response element Proteins 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 102000001712 STAT5 Transcription Factor Human genes 0.000 description 1
- 108010029477 STAT5 Transcription Factor Proteins 0.000 description 1
- 238000002105 Southern blotting Methods 0.000 description 1
- 230000006052 T cell proliferation Effects 0.000 description 1
- 230000005867 T cell response Effects 0.000 description 1
- 102100035794 T-cell surface glycoprotein CD3 epsilon chain Human genes 0.000 description 1
- 102100034922 T-cell surface glycoprotein CD8 alpha chain Human genes 0.000 description 1
- 210000000662 T-lymphocyte subset Anatomy 0.000 description 1
- 239000012163 TRI reagent Substances 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- QPMSXSBEVQLBIL-CZRHPSIPSA-N ac1mix0p Chemical compound C1=CC=C2N(C[C@H](C)CN(C)C)C3=CC(OC)=CC=C3SC2=C1.O([C@H]1[C@]2(OC)C=CC34C[C@@H]2[C@](C)(O)CCC)C2=C5[C@]41CCN(C)[C@@H]3CC5=CC=C2O QPMSXSBEVQLBIL-CZRHPSIPSA-N 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000008186 active pharmaceutical agent Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- 150000001413 amino acids Chemical group 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 208000006673 asthma Diseases 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000003190 augmentative effect Effects 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 230000024245 cell differentiation Effects 0.000 description 1
- 230000010261 cell growth Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 208000032852 chronic lymphocytic leukemia Diseases 0.000 description 1
- 210000004081 cilia Anatomy 0.000 description 1
- 230000004186 co-expression Effects 0.000 description 1
- 238000000749 co-immunoprecipitation Methods 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 108091008034 costimulatory receptors Proteins 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 210000001151 cytotoxic T lymphocyte Anatomy 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 230000007423 decrease Effects 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 239000003599 detergent Substances 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- 230000008034 disappearance Effects 0.000 description 1
- VHJLVAABSRFDPM-QWWZWVQMSA-N dithiothreitol Chemical compound SC[C@@H](O)[C@H](O)CS VHJLVAABSRFDPM-QWWZWVQMSA-N 0.000 description 1
- 238000001378 electrochemiluminescence detection Methods 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 230000008011 embryonic death Effects 0.000 description 1
- 210000002257 embryonic structure Anatomy 0.000 description 1
- 210000003743 erythrocyte Anatomy 0.000 description 1
- DEFVIWRASFVYLL-UHFFFAOYSA-N ethylene glycol bis(2-aminoethyl)tetraacetic acid Chemical compound OC(=O)CN(CC(O)=O)CCOCCOCCN(CC(O)=O)CC(O)=O DEFVIWRASFVYLL-UHFFFAOYSA-N 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 239000013613 expression plasmid Substances 0.000 description 1
- 210000003722 extracellular fluid Anatomy 0.000 description 1
- 210000002744 extracellular matrix Anatomy 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 230000009395 genetic defect Effects 0.000 description 1
- 210000001102 germinal center b cell Anatomy 0.000 description 1
- 229940096919 glycogen Drugs 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 108060003552 hemocyanin Proteins 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 239000000833 heterodimer Substances 0.000 description 1
- 238000005734 heterodimerization reaction Methods 0.000 description 1
- 230000013632 homeostatic process Effects 0.000 description 1
- 239000000710 homodimer Substances 0.000 description 1
- 230000004727 humoral immunity Effects 0.000 description 1
- 230000005934 immune activation Effects 0.000 description 1
- 208000003669 immune deficiency disease Diseases 0.000 description 1
- 230000036737 immune function Effects 0.000 description 1
- 230000008102 immune modulation Effects 0.000 description 1
- 238000003018 immunoassay Methods 0.000 description 1
- 238000010324 immunological assay Methods 0.000 description 1
- 238000001114 immunoprecipitation Methods 0.000 description 1
- 230000004957 immunoregulator effect Effects 0.000 description 1
- 239000003547 immunosorbent Substances 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 230000002147 killing effect Effects 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000004807 localization Effects 0.000 description 1
- 210000004324 lymphatic system Anatomy 0.000 description 1
- 210000003563 lymphoid tissue Anatomy 0.000 description 1
- 230000012976 mRNA stabilization Effects 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 230000036210 malignancy Effects 0.000 description 1
- 210000001161 mammalian embryo Anatomy 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 210000004379 membrane Anatomy 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- 201000010478 microphthalmia Diseases 0.000 description 1
- 230000002297 mitogenic effect Effects 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 210000004400 mucous membrane Anatomy 0.000 description 1
- 210000000066 myeloid cell Anatomy 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 210000004967 non-hematopoietic stem cell Anatomy 0.000 description 1
- 230000009871 nonspecific binding Effects 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 230000009437 off-target effect Effects 0.000 description 1
- 210000000287 oocyte Anatomy 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 210000002997 osteoclast Anatomy 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 230000002085 persistent effect Effects 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 230000007180 physiological regulation Effects 0.000 description 1
- 239000000049 pigment Substances 0.000 description 1
- 210000004694 pigment cell Anatomy 0.000 description 1
- 230000003169 placental effect Effects 0.000 description 1
- 230000010118 platelet activation Effects 0.000 description 1
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- 230000029279 positive regulation of transcription, DNA-dependent Effects 0.000 description 1
- 238000012809 post-inoculation Methods 0.000 description 1
- 230000002516 postimmunization Effects 0.000 description 1
- 230000035935 pregnancy Effects 0.000 description 1
- 230000009696 proliferative response Effects 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 230000004952 protein activity Effects 0.000 description 1
- 230000029983 protein stabilization Effects 0.000 description 1
- 238000001243 protein synthesis Methods 0.000 description 1
- 230000007115 recruitment Effects 0.000 description 1
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 1
- 230000000754 repressing effect Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 210000003296 saliva Anatomy 0.000 description 1
- 239000012723 sample buffer Substances 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 210000001082 somatic cell Anatomy 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000000758 substrate Substances 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 230000002195 synergetic effect Effects 0.000 description 1
- 238000007910 systemic administration Methods 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 210000001138 tear Anatomy 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 230000002992 thymic effect Effects 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- IHIXIJGXTJIKRB-UHFFFAOYSA-N trisodium vanadate Chemical compound [Na+].[Na+].[Na+].[O-][V]([O-])([O-])=O IHIXIJGXTJIKRB-UHFFFAOYSA-N 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000011592 zinc chloride Substances 0.000 description 1
- JIAARYAFYJHUJI-UHFFFAOYSA-L zinc dichloride Chemical compound [Cl-].[Cl-].[Zn+2] JIAARYAFYJHUJI-UHFFFAOYSA-L 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2875—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
Definitions
- the present invention relates to immunology and, more particularly, to the treatment of immune dysfunction by the regulation of CD40 ligand (CD40L) expression.
- CD40L CD40 ligand
- the immune system is a responsive protection system comprised of cells in bone marrow, the thymus, and the lymphatic system of ducts and nodes, spleen, and blood. Aspects of the immune system include the innate immune response and the adaptive immune response.
- An innate immune response is nonspecific, and includes, for example, skin, cilia in mucous membranes, tears, saliva, nasal secretions, and phagocytic cells that migrate to infected areas and engulf pathogens.
- An adaptive immune response is a specific response against an individual antigen, and includes humoral and cellular systems.
- the humoral system produces antibodies to eliminate pathogens and their products.
- the cellular system eliminates pathogens that have invaded cells and regulates the body's entire immune response.
- B cells are specialized white blood cells produced in the bone marrow. Every B cell contains multiple copies of one kind of antibody as a surface receptor for antigen. When the antibody on the surface of a B cell binds to an antigen, the cell can be stimulated to undergo a process called clonal selection. This process includes proliferation and differentiation, where the cells produced make the same antibody, but become memory cells and plasma cells. Memory cells insure that subsequent infections by that particular pathogen receive a quicker and more efficient response. Plasma cells secrete large quantities of the antigen-specific antibody. The antigen-specific antibody forms complexes with free pathogens and their harmful products, inactivating pathogens and stimulating other innate systems including phagocytic cells and complement to eliminate the danger from extracellular fluids.
- the cellular system responds to cells containing pathogen display antigen fragments on their cell surfaces.
- Receptors on the surface of CD8 cells cytotoxic T cells
- CD8 cells must interact with CD4 cells (helper T cells) to regulate destruction of infected cells.
- CD4 cells regulate other ceils of the immune system through secretion of molecules called cytokines.
- CD4 cells are generally required for the clonal selection of B cells, as described above.
- CD40 ligand CD40L, also referred to as CD154, and gp39 (glycoprotein of 39 kiloDaltons) is a critical effector molecule expressed mainly by activated CD4+ T cells and is essential for thymus (T)-dependent immunity.
- Surface CD40L expressed by activated T cells is required to activate B cells and monocytes via CD40 molecules expressed on those cells.
- IgM hyper immunoglobulin M
- CD40L expression by T cells is associated with multiple human immunological and inflammatory diseases such as, but not limited to, systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA).
- CD40L is also expressed by other cells, such as, for example, endothelial cells and platelets, where it is thought to have a role in inflammation and hemostasis.
- CD40L Given the relationship of CD40L to human disease and the importance of its controlled expression, knowledge of the transcription factors that regulate CD40L expression in vivo is critically important for understanding normal immune regulation. Accordingly, there exists a need for techniques to manipulate CD40L expression as a way of treating human disorders in which such CD40L expression is abnormal, and yet does not suffer from one or more of the problems exhibited by conventional treatment methodologies.
- Principles of the present invention provide techniques for treating immune dysfunction by regulation of CD40 Ligand expression.
- a technique treating at least one of an immunological disease and an inflammatory disease in a patient includes the step of suppressing at least one of transcription factor binding to ⁇ enhancer site 3 (TFE3) and transcription factor EB (TFEB) in the patient to thereby suppress CD40L expression.
- TFE3 and TFEB may be suppressed, in accordance with a preferred embodiment of the invention, by blocking the synthesis of at least one of TFE3 and TFEB, and/or by blocking an ability of the TFE3 and TFEB molecules to interact with each other and/or interact (for example, bond) with deoxyribonucleic acid (DNA).
- TFE3/TFEB inhibitors Shown in the drawings that support the present invention are the effects of two types of TFE3/TFEB inhibitors on CD40L expression, a trans-dominant negative protein (TDN) and stem-loop RNA mediated interference (slRNA).
- TDN trans-dominant negative protein
- slRNA stem-loop RNA mediated interference
- FIG. Ia depicts an illustrative immunoblot of TFE3 and TFEB in primary CD4+ mouse splenocytes and human Jurkat T cells, according to an embodiment of the present invention
- Ib depicts an illustrative time course of TFE3 and TFEB protein expression in mouse CD4 T cells after TCR stimulation, according to an embodiment of the present invention
- FIG. Ic depicts an illustrative analysis of Mitf protein expression in mouse CD4 and human Jurkat T cells before and after stimulation and in mouse thymocytes, according to an embodiment of the present invention
- FIG. Id depicts an illustrative semi-quantitative reverse transcriptase dependent- polymerase chain reaction (RT-PCR) assay to measure relative abundance of Tcfe3, Tcfeb, and Mitf RHSTA transcripts in resting and stimulated CD4+ T cells over a time course, according to an embodiment of the present invention
- FIG. 1 e is an illustrative schematic view depicting the E ⁇ P ⁇ -TDN ("E ⁇ " and "P ⁇ ” refer to the immunoglobulin ⁇ heavy chain intronic enhancer and promoter, respectively) transgene vector used in one or more embodiments of the present invention
- FIG. If depicts an illustrative immunoblot of TDN protein in extracts from total bone marrow (BM), total spleen (Spleen) and total thymocytes (Thymus) from trans-dominant negative-transgenic (TDN-Tg) mice (+) but not in the same types of cells isolated from non-transgenic (non-Tg) littermates (-), according to an embodiment of the present invention;
- FIG. Ig depicts illustrative flow cytometry histograms of permeabilized lymphocytes from non-Tg and the T cell-specific TDN-Tg mice for intensity of staining with an anti-hemagglutinin (-HA) antibody that detects the TDN protein, according to an embodiment of the present invention
- FIG. Ih depicts an illustrative immunoblot of TDN protein present in extracts of purified CD4+ T cells, but not splenic B cells, from TDN-transgenic mice, according to an embodiment of the present invention
- FIGS. 2a and 2b are illustrative graphical views of cell counts and flow cytometry depicting how numbers and a distribution of major B and T cell populations is normal in TDN-transgenic mice, according to an embodiment of the present invention
- FIGS. 3a and 3b are illustrative graphical views using imrnunohistochemistry and flow cytometry to depict impaired germinal center formation in TDN-transgenic mice, according to an embodiment of the present invention
- FIGS. 4a through 4d are illustrative graphical views of ELISA assays depicting impaired T-dependent, but normal T-independent, humoral responses in TDN-transgenic mice, according to an embodiment of the present invention
- FIG. 5 a depicts an illustrative flow cytometry demonstrating impaired surface expression of CD40L by TDN-transgenic (TDN) as compared to non-Tg control (wild type (WT)) CD4+ T splenocytes stimulated with monoclonal antibodies (niAb) to CD3 and analyzed at 8 hours, according to an embodiment of the present invention
- TDN TDN-transgenic
- WT wild type
- niAb monoclonal antibodies
- FIG. 5b depicts an illustrative quantification of flow cytometry histogram data for percent CD4+ T cells positive for CD40L (left) and CD25 (right) after S hours of stimulation with various amounts of mAb to CD3 (Anti-CD3), according to an embodiment of the invention
- FIGS. 5c and 5d depict illustrative flow cytometry demonstrating comparable levels of ICOS (Inducible Co-stimulator of T cells) and CD28, respectively, by control (wild type (wt)) and TDN-transgenic (TDN) CD4+ splenocytes, according to an embodiment of the present invention
- FIG. 5e depicts an illustrative graphical representation of ELISA (enzyme linked immunoadsorbance assay) analyses showing comparable levels of interleukin-4 (IL-4) secretion by control (wild type (WT)) and TDN-transgenic (TDN) CD4+ splenocytes, according to an embodiment of the present invention
- IL-4 interleukin-4
- FIG. 5 f depicts an illustrative bar graph showing normal proliferation of TDN-Tg T cells in response to CD3/CD28 stimulation, according to an embodiment of the present invention
- FIG. 5g depicts a representative semi-quantitative RT-PCR analysis showing that Smad 7 expression is normal in TDN-Tg splenic T cells, according to an embodiment of the invention
- FIG. 5h depicts an illustration of representative flow cytometry histograms depicting impaired GD40L but normal CD25 expression by TFE3 -deficient (TcfeS-/-) CD4 T cells infected with a lentivirus that expresses an interfering stem-loop RNA against TFEB, according to an embodiment of the present invention
- FIG. 5i depicts an illustrative representation of an immunoblot showing the expected down-regulation of TFEB protein in CD4+ T cells infected with a retrovirus that expresses the interfering stem-loop TFEB RNA in support of the data in FIG. 5h ⁇ according to an embodiment of the present invention
- FIG. 5j depicts an illustrative real-time RT-PCR analysis of ribonucleic acid (RNA) from CD4+ T cells showing reduced abundance of CDAQIg transcripts in CD4+ T cells from TDN-transgenic mice relative to non-transgenic littermates at various times after incubation with mAb to CD3 (horizontal axis), according to an embodiment of the invention
- FIG. 6a depicts an illustrative flow cytometry histogram demonstrating indistinguishable CD40 expression in B cells from non-transgenic and TDN-transgenic mice, according to an embodiment of the present invention
- FIGS. 6b and 6c depict illustrative flow cytometry histograms, demonstrating equivalent expressions of CD86 and major histocompatibility complex (MHC) class II, respectively, after stimulation of B cells from non-transgenic and TDN-transgenic mice with mAb to CD40, according to an embodiment of the present invention
- MHC major histocompatibility complex
- FIG. ⁇ d depicts an illustrative ELISA measuring immunoglobulin G (IgG) responses to TNP-KLH (Trinitrophenol-keyhole limpet hemocyanin) which shows equivalent responses in non-transgenic and TDN-transgenic mice treated with an agonist mAb to CD40 during immunization, according to an embodiment of the present invention
- FIG. 7a depicts an illustrative schematic alignment of promoters for the gene encoding
- CD40L from humans, mice and rats, showing MiT (the MitfTTFE family) sites (a subset of E-boxes; "E” refers to “enhancer” boxes from the IgH enhancer) where TFE3 and TFEB bind to and activate the CD40lg and CD40LG promoters, according to an embodiment of the present invention
- FIG. 7b depicts an illustrative Chromatin immunoprecipitation (ChIP) of a CD40lg promoter fragment by anti-TFE3 and anti-TFEB antibodies, as well as the illustrative controls for the ChIP assays, according to an embodiment of the present invention
- FIG. 7c depicts illustrative electrophoretic mobility-shift assays (EMSA) demonstrating the binding of native TFE3 and TFEB proteins present in CD4 T cell extracts to individual MiT sites from the Cd40lg promoter, according to an embodiment of the present invention
- FIG. 7d depicts illustrative electrophoretic mobility shift assays (EMSAs) of nuclear extracts of human embryonic kidney (HEK293) cells transfected with control (-), TFE3, or TFEB pEBB expression vectors showing TEE3 and TFEB binding to the MiT sites from the Cd40lg promoter, according to an embodiment of the present invention
- FIG. 8a is a bar graph depicting illustrative luciferase assays to measure Cd40lg promoter activity in CD4 T cells from non-Tg and TDN-Tg mice, showing that a CD40lg fragment containing the MiT sites depends on endogenous TFE3 and TFEB to enhance promoter activity, according to an embodiment of the present invention
- FIG. 8b is a bar graph depicting illustrative luciferase assays showing the contributions of individual and combinations of MiT sites to Cd40lg promoter activity in non-Tg, TDN-Tg, and Tcfei-f- CD4 T cells, according to an embodiment of the present invention
- FlG. 8c depicts immunological assays that show that native TFE3 and TFEB in CD4 T cells can form heterodimers, an interaction that is blocked by the TDN protein, according to an embodiment of the present invention
- FIG. 9a is a bar graph depicting illustrative luciferase assay showing the importance of MiT sites and endogenous TFE3 and TFEB for the human CD40LG and mouse CD40lg promoter activity in human Jurkat T cells, according to an embodiment of the present invention
- FIG. 9b is an illustrative graphical view of a flow cytometry histogram depicting how inhibition of TFE3 and TFEB activity by expression of the TDN protein blocks endogenous CD40L induction after activation of human Jurkat T cells but that CD25 induction is not affected, according to an embodiment of the present invention
- FIG. 10a depicts a table illustrating oligonucleotides for EMSA in FIGS. 7, including SEQ. ID NOs: 1-16, according to an embodiment of the present invention
- FIG. 10b depicts a table illustrating oligonucleotides for cloning Cd40lg and CD40LG promoters, including SEQ. ID NOs: 17-22, according to an embodiment of the present invention
- FIG. 10c depicts a table illustrating oligonucleotides for E-box mutagenesis, including
- FIG. 11a depicts an immunoblot showing that TFE3 and TFEB proteins are expressed in B cells, according to an embodiment of the present invention
- FIG. lib depicts a representative immunoblot showing TDN protein in B cell containing lymphoid organs from B-cell-specific TDN-Tg mice, according to an embodiment of the present invention
- FIG. lie depicts representative flow cytometry histograms showing that the TDN protein is expressed only in B, but not T cells in "B-cell-specific TDN-Tg" mice, according to an embodiment of the present invention
- FIG. 1 Id depicts representative flow cytometry dot plots showing that major B and T cell populations in these "B cell-specific TDN-Tg" mice are normal according to an embodiment of the present invention
- FlG. 12 is a flow chart illustrating a technique for treating at least one of an immunological disease and an inflammatory disease in a patient, according to an embodiment of the present invention.
- One or more embodiments of the present invention meet the above-noted need by providing methods of treating an immune disease in which CD40L expression is abnormal or otherwise drives pathology.
- the invention in an illustrative embodiment thereof, beneficially exploits an advantageous role of transcription factor binding to immunoglobulin heavy chain ⁇ (IGHM) enhancer ( ⁇ E) site 3 (TFE3) and transcription factor EB (TFEB) in regulating CD40L expression.
- IGHM immunoglobulin heavy chain ⁇
- ⁇ E ⁇ E site 3
- TFEB transcription factor EB
- One or more embodiments of the present invention teach, for example, that TFE3 and TFEB directly activate CD40L at the transcriptional level in vivo without significantly affecting the synthesis of other activation and effector molecules, Biactivation of TFE3 and TFEB attenuates the induction of CD40L expression on T cells.
- TFE3 and TFEB are broadly expressed transcription factors related to the transcription factor Mitf. Although they have been linked to cytokine signaling pathways in non-lymphoid cells, their function in T cells is unknown. TFE3 -deficient mice are phenotypically normal, whereas TFEB deficiency causes early embryonic death.
- One or more embodiments of the present invention illustrate that combined inactivity of TFE3 and TFEB in T cells result in a hyper-immunoglobulin M (hyper-IgM) syndrome due to impaired expression of the CD40 ligand gene (Cd40lg) by CD4+ T cells.
- hyper-IgM hyper-immunoglobulin M
- CD40lg CD40 ligand gene
- CD40L expression by T cells is associated with multiple human immunological and inflammatory diseases such as, but not limited to, systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA).
- CD40L is also expressed by other cells, such as, for example, endothelial cells and platelets, where it is thought to have a role in inflammation and hemostasis.
- CD40L refers to the protein
- Cd40lg refers to the name of the mouse gene encoding the CD40L protein
- CD40LG refers to the human gene.
- TFE3 and TFEB refer to the proteins
- Tcfe3 and Tcfeb refer to the mouse genes. Native TFE3 and TFEB bound to multiple cognate sites in the promoter of the gene encoding CD40 ligand, and maximum Cd40lg promoter activity and gene expression required TFE3 or TFEB.
- TFE3 and TFEB are direct, physiological and mutually redundant activators of Cd40lg expression in activated CD4+ T cells critical for T cell-dependent antibody responses.
- the teachings of one or more embodiments of the present invention relate generally to the regulation of CD40L expression. Control of CD40L expression by T cells is complex and highly regulated, befitting its central role in immune activation and modulation.
- T cells and B cells it is contemplated that the techniques of the invention may be used for controlling CD40L expression by immune cells in general.
- immune cells as used herein is intended to include (but is not limited to) hematopoietic and endothelial cells.
- CD40L is rapidly and transiently induced upon stimulation of naive T cells via the T cell receptor (TCR), with surface levels peaking by 6-8 hours and then declining.
- TCR T cell receptor
- This profile of TCR-dependent CD40L induction primarily reflects transcriptional activity.
- CD40L expression can be prolonged and augmented by additional stimulatory input from co-stimulatory and/or accessory molecules such as, but not limited to, CD28, CD2, LFA-I (lymphocyte-function associated antigen-1), and CD43, and interleukins, such as TL-2, IL-12, and IL-15, via mechanisms that involve enhanced transcription, increased CD40L messenger-RNA (mRNA) stability, or both. Consequently, the overall dynamics of CD40L expression is dependent on TCR stimulatory conditions, activation and/or effector status, developmental stage of the T cell, and age.
- mRNA messenger-RNA
- mice engineered to express CD40L via heterologous promoters in T cells have been shown to develop lymphoproliferative disorders and tumors, highlighting the importance of regulated CD40L transcription (see, for example, Brown, M.P. et al., "Thymic lymphoproliferative disease after successful correction of CD40 ligand deficiency by gene transfer in mice," Nat Med 4, pp. 1253-1260 (1998), and Sacco, M.G. et al., "Lymphoid abnormalities in CD40 ligand transgenic mice suggest the need for tight regulation in gene therapy approaches to hyper immunoglobulin M (IgM) syndrome," Cancer Gene Ther. 7, pp. 1299-1306 (2000), the disclosures of which are incorporated by reference herein).
- IgM hyper immunoglobulin M
- CD40L induction is dependent on subunits of nuclear factors of activated T-cells (NFAT), a Ca 2+ -dependent transcription factor complex.
- Additional regulators include subunits of AP-I (Activator Protein- 1) and NFkB (Nuclear Factor that binds to the kappa enhancer B site) and Egr-1 (early growth response-1), which were also shared by other TCR and/or co-stimulator responsive genes, and AKNA.
- Binding sites for other transcription factors such as, for example, STAT5, GATA-3, Oct-1 subunits, have also been identified (see, for example, Crow, M.KL & Kirou, K.A, "Regulation of CD40 ligand expression in systemic lupus erythematosus,” Curr Opin Rheumatol 13, pp. 361-369 (2001), and Cron, R. Q, "CD154 transcriptional regulation in primary human CD4 T cells,” Immunol Res 27, pp. 185-202 (2003), which are incorporated by reference herein), but their physiological contribution to CD40L transcription is not yet known.
- other transcription factors such as, for example, STAT5, GATA-3, Oct-1 subunits
- CD40L transcription in T cells may involve as yet unknown factors/ or elements.
- relatively less is known about how CD40L transcription is regulated in non-lymphoid cells.
- CD40L Given the relationship of CD40L to human disease and the importance of its controlled expression, knowledge of the transcription factors that regulate CD40L expression in vivo is critically important for understanding normal immune regulation and for designing strategies to manipulate CD40L expression as a way to treat the multiple human disorders in which CD40L expression is abnormal or undesirable.
- the transcription factors TFE3 and TFEB are demonstrated to be physiological transcription regulators of CD40L expression in T cells.
- TFE3 is a member of the helix-loop-helix family of transcription factors that binds to the mu-E3 ( ⁇ E3) motif of the immunoglobulin heavy-chain enhancer and is expressed in many cell types.
- TFEB is a transcription factor with a basic region-DNA binding domain, a helix-loop-helix and leucine zipper dimerization domains, and a nuclear localization signal, thought to be located adjacent to the helix-loop-helix domain. TFEB is ubiquitously expressed.
- TFE3 and TFEB are closely related members of a functionally interactive DNA binding family known as Mitf/TFE (MiT), that includes the microphthalmia transcription factor Mitf and TFEC (Transcription factor E-box C).
- MiT proteins bind to so called ⁇ E3 sites, a subset of E-boxes that match a general CANNTG consensus sequence, with those binding to TFE3 in vitro first identified and characterized in immunoglobulin heavy-chain and T cell receptor (TCR) enhancers.
- TCR immunoglobulin heavy-chain and T cell receptor
- BRs basic regions
- FILH conserved helix-loop-helix
- LZ leucine zipper
- MiT proteins share similar structures and are often expressed together, yet genetic studies have demonstrated both overlapping and non-overlapping functions for MiT proteins in different cell-types.
- Mitf a well-characterized family member, is expressed mainly in pigment and myeloid cells, where it is involved in melanocyte and mast cell development as a transcription mediator of the c-kit pathway,- and is a negative regulator of B cell activation and terminal differentiation.
- TFE3 and Mitf serve redundant roles in osteoclast development as transcriptional mediators of the macrophage colony-stimulating factor (m-csf) pathway. These functions of Mitf correspond to its cell-type and/or lineage restricted expression pattern.
- TFEC expression is restricted mainly to the myeloid lineage, but TFEC-deficient mice are phenotypically normal, even though TFEOdeficient macrophages have lower expression of a subset of interleukin 4 (IL-4) -responsive transcripts, including the transcript for granulocyte-macrophage colony-stimulating factor.
- IL-4 interleukin 4
- TFE3 and TFEB are more broadly expressed, and relatively less is understood about their individual biological functions, primarily because germline TFE3-deficient animals have no reported defects.
- TFE3 facilitates the activation of a subset of genes dependent on transcription factor Smad3 (Mothers against decapentaplegic homolog 3), in response to transforming growth factor- ⁇ (TGF- ⁇ ) including those encoding components of the extracellular matrix and Smad7, a TGF- ⁇ pathway inhibitor.
- Smad3 transcription factor- ⁇
- TGF- ⁇ transforming growth factor- ⁇
- Ectopic over-expression of TFE3 in hepatocytes in mice promotes glycogen synthesis.
- mice deficient in TFE3 are phenotypically normal, with no defects noted in development, reproduction or the immune response.
- TFEB deficient embryos die early in gestation because of defects in placental vascularization. The function of TFEB in adults is not known.
- the present invention exploits a central role of TFE3 and TFEB in the immune system via their control of CD40L expression, and thereby provides a beneficial methodology for treating immune dysfunction.
- immunodeficiency as used herein is intended to refer to any syndrome in which activity of imrnunocytes is undesirable, including, but not limited to, leukemia, lymphoma, lupus and rheumatoid arthritis, transplant rejection, asthma and other allergic diseases, autoimmune diseases, etc.
- TFE3 and TFEB are critical for T cell function and humoral immunity through their direct control of CD40L expression. This was demonstrated by expression of the TDN protein in mouse T cells in vivo or in human T cells in culture, which blocked existing TFE3 and TFEB proteins expressed therein, and by de novo inhibition of TFEB protein synthesis via expression of an interfering RNA against TFEB in Tcfe3-/ ⁇ T cells. TDN expression in mouse T cells in vivo via the E ⁇ P ⁇ transgene resulted in hyper-IgM syndrome caused by defective CD40L expression. As mentioned previously, like the induction of many effector and activation molecules by
- T cell stimulation such as IL-2 and TNF family members
- CD40L induction is dependent on subunits of NFAT, a calcium-responsive transcription factor complex.
- Additional regulators include the AT-Hook ("AT-Hook" refers to the AT-rich sequences this type of transcription factor binds to and the shape of the protein itself) transcription factor AKNA and subunits of transcription factors AP-I, NF-kB and Egr-1, which are also shared by other TCR and/or co-stirnulator-responsive genes.
- AT-Hook refers to the AT-rich sequences this type of transcription factor binds to and the shape of the protein itself
- transcription factor AKNA and subunits of transcription factors AP-I, NF-kB and Egr-1, which are also shared by other TCR and/or co-stirnulator-responsive genes.
- Cd40lg is the only one identified that requires TFE3 or TFEB. That requirement may contribute to the unique expression profile of Cd40lg
- TFE3 and TFEB were mutually redundant in controlling Cd40lg transcription, as a combined deficiency in TFE3 and TFEB inhibited TCR-dependent CD40L induction in the primary T cell culture systems, whereas individual deficiencies had a minor or no effect.
- TFE3 and TFEB have demonstrated physiological functional redundancy between TFE3 and TFEB in vivo, which may be a more general property of these proteins in other cell types.
- TFE3 and TFEB each have a unique contribution to the activation of Cd40lg. For example, only TFEB protein was induced after TCR stimulation of primary cells, most likely through post-transcriptional mechanisms, and TFEB bound to the Cd40lg promoter independently of TFE3.
- Tcfe3 refers to the gene encoding TFE3 T cells. Consequently, there may be physiological contexts in which TFEB or TFE3 may be more advantageous, such as, for example, for NFAT subunits in T cell subsets or in other cell types expressing CD40L.
- CD40L expression in activated T cells lacking TFE3 and TFEB activity is consistent with the limited degree of germinal center formation in TDN-transgenic mice, in contrast to that of mice with genetic Cd40tg deficiency, in which no germinal centers are found, We believe that the remaining CD40L expression was not due to incomplete mactivation of TFE3 and TFEB by the TDN protein, given that TDN protein expression inhibited the binding of endogenous TFE3 or TFEB to the Cd40lg promoter to basal binding amounts, as evaluated by electrophoretic mobility-shift assay and chromatin immunoprecipitation.
- ⁇ E3 also referred to as "MiT” site-binding transcription factors not in the MiT family may bind to the promoter in their absence.
- These may include upstream stimulating factor (USF-I) and c-Myc, which are from distinct and non-interactive helix-loop-helix families.
- USF-I upstream stimulating factor
- c-Myc can also activate the Cd40lg promoter, but its effect is apparently indirect.
- NF-B subunits which can radically affect T cell development, effector function or homeostasis.
- TFE3 has been directly and indirectly linked to several cytokine signaling pathways that control cell growth and differentiation in non-lymphoid cells, including c-K ⁇ t (c-kit is a tyrosine kinase receptor that is mutated in malignancies and is activated by "steel factor,” and is also referred to as the c-kit ligand.
- c-kit is a tyrosine kinase receptor that is mutated in malignancies and is activated by "steel factor,” and is also referred to as the c-kit ligand.
- CDl 17 Another name for c-kit is CDl 17.
- C-kit is critical for the development of mast cells and melanocytes
- transforming growth factor- ⁇ , cell type-specific and gene-specific, regulation by each MiT protein is an established characteristic of this family. Pathway involvement established in one cell type cannot be directly extrapolated to other cell types and must be determined experimentally.
- CD40L expression is often constitutively increased on T cells from patients with systemic lupus erthythematosus and rheumatoid arthritis. In systemic lupus erthythematosus, this occurs without abnormal expression of other markers of T cell activation. Transcriptional and post-transcriptional mechanisms that normally enhance CD40L expression are all thought to contribute to that phenomenon. It has been shown that the Ras GTPase-mitogen-activated protein kinase (MAPK) pathway is necessary for the maintenance of abnormal CD40L expression by T cells from patients with systemic lupus erthythematosus.
- MPK Ras GTPase-mitogen-activated protein kinase
- TFE3 and TFEB can each be regulated by the Ras-mitogen-activated protein kinase (-MAPK) pathway, for example, in response to activation of the m-csf receptor, a key issue will be whether the activity of TFE3 and TFEB can be regulated by MAPK activation in T cells (for example, in response to the TCR and IL-15 receptor) and if there are differences in their activity in normal versus autoimmune T cells.
- MAPK activation of TFE3 and TFEB can be defined, at least in part, as phosphorylation which enables TEF3 and TEFB to interact with other molecules critical for transcription.
- Ia depicts an illustrative imrmmoblot of TFE3 and TFEB protein in un-stiimilated and stimulated primary CD4+ mouse splenocytes (via CD3 mAb) and human Jurkat T cells (stimulated via phorbol myristate acetate [PMA] plus ionomycin), according to an embodiment of the present invention.
- the GAPDH immunoblot is also shown as a loading control.
- FIG. Ib depicts an illustrative time course of TFE3 and TFEB protein expression after TCR stimulation, according to an embodiment of the present invention.
- CD4+ T cells were stimulated with the CD3 mAb and aliquots taken at each time point for analysis by immunoblot. These data are extensions of the data in FIG. Ia.
- FIG. Ic depicts an illustrative analysis of Mi tf protein expression, according to an embodiment of the present invention. Extracts from resting and LPS stimulated CD 19+ B cells, resting and CD3-stimulated (8 hours) CD4+ T cells, un-stim ⁇ lated and pharmacologically stimulated Jurkat T cells, and resting thymocytes, were probed by immunoblot with a Mitf mAb. The cross-reactive band corresponding in size to the A isoform of Mitf is indicated with the line extending from "Mitf.” This designation is also based on the disappearance of this band in stimulated B cells. The GAPDH immunoblot is also shown as a loading control.
- FIG. Id depicts an illustrative semi-quantitative reverse transcriptase (RT) dependent-polymerase chain reaction (PCR) to measure relative abundance of Tcfe3, Tcfeb, (the mouse genes encoding TFE3 and TFEB proteins, respectively) and Mitf mRNA transcripts in resting and stimulated CD4+T cells over a time course, according to an embodiment of the present invention.
- Reverse transcriptase in this context, is required to convert the mRNA into a DNA copy (cDNA) that is then used as a template for amplification in the PCR reaction. The identities of the bands are indicated.
- the titrations illustrate the relative band intensities of PCRs programmed with the same amount of 0 hour complementary DNA (cDNA) used for the PCR series (1/1), one-third the amount (1/3), or three-fold more (3/1).
- Immunoblot (FIGS. Ia, Ib, and Ic) and RT-PCR (FIG. Id) analyses were used to assess the MiT protein and mRNA expression profile, respectively, in T cells.
- TFE3 and TFEB proteins were detected in CD4+ splenic mouse T cells and in the human transformed T cell line Jurkat, as shown in FIG. Ia 5 which depicts an exemplary immunoblot of TFE3 and TFEB in primary CD4+ mouse splenocytes and Jurkat T cells.
- CD4+ splenic T cells were stimulated by incubation with an anti-CD3 mAb for 8 hrs.
- Jurkat T cells were stimulated with phorbol myristate acetate (PMA)/ionomycin (Iono) for 20 hours.
- PMA phorbol myristate acetate
- Iono ionomycin
- Glyseraldehyde-3- ⁇ hosphate dehydrogenase (GAPDH) protein was the loading control.
- the TFEB and GAPDH antibodies recognized epitopes on the corresponding proteins from mouse and human and were used for probing both extracts.
- TFE3 proteins from mouse and human extracts were revealed with species restricted anti-TFE3 antibodies. Consequently, band intensities cannot be directly compared between mouse and human.
- the exemplary images shown in FIGS. la-Id are representative of at least three independent experiments.
- TFEB protein levels increased (five-fold) in response to TCR-engagement in parallel with CD40L induction in primary mouse T cells, whereas TFEB levels were already elevated in un-stimulated Jurkat cells and did not change in response to stimulation.
- TFE3 levels remained relatively constant in both cases, as apparent from FIG. Ia.
- the increase in TFEB was at the post-transcriptional level, as relative amounts of steady-state Tcfeb (the mouse gene encoding TFEB protein) mRNA were unchanged (FIG. Id).
- a band was detected that was interpreted to be the A isoform of Mitf in un-stimulated CD4+ T cells, but we did not detect it in TCR-stimulated cells (FIG. Ic). This corresponds to a decrease in Miff mRNA (FIG. Id).
- One or more embodiments of the present invention illustrate that T-cel ⁇ -specific inactivation of endogenous TFE3 and TFEB in vivo via transgenesis results in a phenotype resembling X-linked hyper-IgM syndrome *
- FIG. Ie is an illustrative schematic view depicting an E ⁇ P ⁇ -TDN ("E ⁇ " and "P ⁇ ” refer to the immunoglobulin ⁇ heavy chain intronic enhancer and promoter, respectively) transgene vector used to create the TDN-transgenic (Tg) mice, according to an embodiment of the present invention.
- E ⁇ and P ⁇ refer to the immunoglobulin ⁇ heavy chain intronic enhancer and promoter, respectively
- Tg TDN-transgenic mice
- the construction of the TDN protein has been described (see, for example, Huan, C, Sashital, D., Hailemariam, T., Kelly, M.L. & Roman, C.A.J., Renal carcinoma associated transcription factors TFE3 and TFEB are leukemia-inhibitory factor-responsive transcription activators of E-cadherin," J Biol Chem 13, p. 13 (2005), which is incorporated by reference herein).
- HLH-Z region from TFE3 but lacks the DNA binding BR and transcriptional activation domains.
- a SV40-nuclear localization signal was added to compensate for the loss of that activity by deletion of the BR, and a HA-tag was added for immunodetection.
- FIG. If depicts an illustrative immunoblot of extracts from total bone marrow (BM), total spleen (Spleen) and total thymocytes (Thymus) from TDN-transgenic mice (+) and non-transgenic littermates (-), according to an embodiment of the present invention.
- Non-transgenic mice are control littermates that did not receive the chromosome containing the transgene from the transgenic parent.
- FIG. If depicts, in part, TDN expression in total extracts of HEKL293 cells transfected with a plasmid expressing the TDN cDNA (lane labeled HEK TDN) for comparison.
- the anti-HA antibody detects the cognate hemagglutinin (HA) epitope built into the TDN protein.
- FIG. 1 f shows that the TDN protein is only detected in lymphoid organs that contain T cells from TDN-Tg mice and not in non-Tg mice, which established, in part, that TDN expression in this transgenic line was T cell specific.
- FIG. Ig depicts illustrative flow cytometry histograms of permeabilized lymphocytes from non-Tg and T cell-specific TDN-Tg mice for staining with an anti-HA antibody that detects the TDN protein, according to an embodiment of the present invention. In the exemplary histograms in FIG.
- Ig filled curves are HA staining profiles of non-Tg (control) cells
- line overlays are profiles of cells from TDN-Tg mice from bone marrow, spleen, and thymocytes.
- B and T lymphocyte populations are delineated using lineage-specific markers.
- FIG. Ih depicts an illustrative immunoblot of extracts of purified splenic B cells and CD4+ T cells from TDN-transgenic mice (-), showing that the TDN protein is expressed only by splenic T cells but not in B cells from TDN-Tg mice, according to an embodiment of the present invention.
- FIG. Ih includes, in part, an immunoblot for the hemagglutinin (HA) epitope built into the TDN protein, and GAPDH, a loading control. Data are representative of at least three independent experiments.
- HA hemagglutinin
- FIGS. 2a and 2b are illustrative graphical views depicting how the numbers and a distribution of major B and T cell populations is normal in TDN-transgenic mice, according to an embodiment of the present invention.
- TDN-transgenic mice exhibited humoral immune defects that were consistent with a hyper IgM syndrome caused by a deficit in CD40L expression.
- An E ⁇ P ⁇ transgene as described, for example, in Tepper, R.I. et al, "IL-4 induces allergic-like inflammatory disease and alters T cell development in transgenic mice," Cell 62, pp.457-467 (1990), which is incorporated by reference herein, was used to direct expression of the TDN protein.
- transgenic mouse lines were analyzed in which the TDN was expressed exclusively in T cells, which was shown in FIGS. If-Ih. TDN expression was found to be highest in thymocytes and lower in mature single positive splenic T cells.
- FIGS. If-Ih No TDN protein was detected in B cells or no ⁇ i-lymphoid cells and tissues.
- FIGS. If-Ih Analysis of lymphoid compartments in TDN-transgenic mice by cell counting and flow cytometry revealed that B and T cell development proceeded, with normal to nearly normal numbers and distributions of major splenic B and T cell populations (Spleen) and thymocytes (Thymus) at 5 weeks of age. This is shown in FIGS. 2a and 2b. Li hyper-IgM syndrome due to CD40L deficiency, antigen-independent lymphocyte development is normal. Rather, the hallmark phenotype includes defects in germinal center (GC) formation and antibody responses to T-dependent antigens.
- GC germinal center
- FIGS. 3a and 3b are illustrative graphical views depicting impaired germinal center formation in TDN-transgenic mice immunized with SRBCs, according to an embodiment of the present invention.
- FIG. 3a PNA and PNA plus B220 stains; PNA (peanut agglutinin) and B220 (also referred to as B cell antigen of 220 kiloDalton molecular weight) identify GC B cells).
- PNA and PNA plus B220 stains PNA (peanut agglutinin) and B220 (also referred to as B cell antigen of 220 kiloDalton molecular weight) identify GC B cells.
- PNA peanut agglutinin
- B220 also referred to as B cell antigen of 220 kiloDalton molecular weight
- FIGS. 3a and 3b illustrate that TDN-transgenic T cells are present in the normal, expected locations in the spleens of immunized mice, adjacent to B cells. This is another property of T cells that could have been affected and caused the immune deficiency, but FIGS. 3a and 3b illustrate that it was not such a cause. That is, if T cells cannot make it to the proper place in the spleen, they cannot interact with B cells even if they had expressed CD40L.
- FIGS. 3a and 3b depict that T cells can make it to the proper place in the spleen, thereby illustrating that this property is normal and therefore does not account for the GC defect.
- FIGS. 4a through 4d are illustrative graphical views of ELISAs depicting impaired T-dependent, but normal T-independent, humoral (antibody) responses in TDN-transgenic mice, according to an embodiment of the present invention.
- Impaired GC formation is indicative of a lapse in T ⁇ -cell (helper T cells, primarily CD4+ T cells) dependent B cell activation and is predictive of impaired T-dependent antibody responses.
- serum levels of total IgG and IgA isotypes in naive TDN-transgenic mice were reduced compared to controls, whereas total IgM was normal if not slightly elevated, as shown in FIG.4a.
- This prediction was then tested directly by evaluating the humoral responses of mice that were challenged with the T-independent and T-dependent antigens TNP-Ficoll and TNP-KLH, respectively.
- TNP-specif ⁇ c antibody responses to TNP-Ficoll were comparable in TDN-transgenic and control mice.
- TDN-transgenic mice failed to mount a characteristic humoral response to the T-dependent antigen TNP-KLH compared to normal- as evidenced by dramatically reduced TNP -specific IgG antibodies at 14 and 21 days post immunization compared to controls, as illustrated in FIG. 4c.
- TNP-KLH T-dependent antigen
- IgG antibodies TNP -specific IgG antibodies
- per capita SRBC-specific plasma cell formation was also proportionally reduced, as shown in FIG. 4d.
- TFE3 and TFEB inactivation in T cells mediated by transgenic TDN expression led to poor thymus dependent B cell responses to model antigens in a manner consistent with a CD40L deficiency.
- T cell-specific TDN-transgenic mice showed the hallmark phenotypes of hyper-lgM syndrome, in which antigen-independent lymphocyte development and antibody responses to T cell-independent antigens are intact, but germinal center formation and IgG responses to T cell-dependent antigens are defective.
- TDN-transgenic mice The normal distribution and locations of major T cell populations in unimmunized TDN transgenic mice suggests that any other possible target genes of TFE3 and/or TFEB in T cells were not required for T cell development per se beyond the double positive (DP) stage when the TDN protein was first clearly detected (see, for example, FIGS. 2 and 3a). Also unaffected were antibody responses to the thymus independent antigen TNP-Ficoll (FIG. 4), which suggests that any T cell functions that influence this response were largely intact in the absence of TFE3 and TFEB activity. As detailed below, one or more embodiments of the invention illustrate that defective CD40L expression by TDN-transgenic T cells underlies the humoral immune defect in TDN-transgenic mice.
- FIG. 5a depicts an illustrative flow cytometry of surface expression of CD40L by control (wild-type (WT)) and TDN-transgenic (TDN) CD4+ splenocytes stimulated, by way of example, with the anti-CD3 mAb and analyzed by flow cytometry for CD40L marker expression, according to an embodiment of the present invention.
- Filled curves represent CD40L staining of un-stimulated cells, and grey lines represent CD3 -stimulated cells. Numbers above bracketed lines indicate CD40L+ cells. Data are representative of more than three experiments.
- FIG. 5a depicts how surface expression of CD40L is impaired in TDN-transgenic T cells.
- CD40L was much lower on the surface of TDN-transgenic CD4+ T cells than that of non-transgenic (WT) cells after TCR stimulation.
- Representative data is of T cells from one TDN-Tg line of mice, but is similar in the three lines studied.
- FIG. 5b depicts an illustrative quantification in bar graph form of histogram data as in FIG. 5a for percent CD4+ T cells positive for CD40L (left) and CD25 (right) after S hours of stimulation with various amounts of mAb to CD3 (Anti-CD3), according to an embodiment of the invention.
- FIGS. 5c and 5d depict illustrative flow cytometry histograms of expressions of ICOS and CD28, respectively, showing that their expression is unaffected in TDN-Tg T cells compared to non-Tg (WT) 5 according to an embodiment of the present invention.
- Tn FIGS. 5c and 5d, filled curves represent the relative intensity of ICOS or CD28 on freshly isolated CD4+ T cells.
- Green lines depicted in FIG. 5d represent the intensity on cells left un-stimulated in culture for 2 days.
- FIG. 5e depicts an illustrative graphical representation of ELISA measurements of
- IL-4 secretion according to an embodiment of the present invention.
- T cells from control and transgenic mice were cultured with plate-bound anti-CD3 mAb for 2 days, followed by IL-2 for 7 days, and then re-stimulated with anti-CD3 without IL-2 for 2 days.
- FIG. 5f depicts an illustrative bar graph showing that the proliferative response of TDN-Tg
- CD4 T cells in vitro to mitogenic stimulation by CD3/CD28 beads is indistinguishable from control non-Tg CD4 T cells.
- T cell proliferation in response to CD3 and CD28 co-stimulation is indistinguishable between non-Tg and TDN-Tg T cells.
- FIG. 5g depicts an illustrative RT-PCR showing that Srnad7 expression is unaffected in
- TDN-Tg and T ⁇ 3-/ ⁇ splenic CD4 T cells are analyzed here because it is regulated by TFE3 and TFEB in non-hematopoietic cells.
- FIG. 5 of the present invention teaches us that inactivation of TFE3 and TFEB had no effect on TCR-dependent CD25, CD69, or CD28 expression, IL-4 secretion, IL-2 responsiveness or CD3/CD28 responsiveness of CD4+ T cells, or Smad7 expression.
- the data therefore suggest that the effect of TFE3 and/or TFEB inactivation was relatively restricted to
- CD40L did not globally affect T cell responses to stimulation, in contrast to deficiencies in some NFAT and NFkB subunits.
- One or more embodiments of the present invention illustrate that CD40L expression by CD4 T cells depends on endogenous TFE3 or TFEB. That is, they are mutually redundant activators of CD40L expression in CD4 T cells.
- FIG. 5h depicts illustrative flow cytometry histograms showing CD40L and CD25 staining profiles of un-stimulated (filled curve) and TCR-stimulated (line overlay) green fluorescent protein (GFP+) cells from normal and TcfeS-/- mice (for three independent experiments), in which GFP shows cells that express either a control virus or a virus that expresses an inhibitory (interfering) stem-loop RNA that blocks TFEB synthesis, according to an embodiment of the present invention.
- Primary CD4+ splenic T cells were isolated from normal and Tcfe3-/- mice infected with the indicated stem-loop-interfering RNA/GFP lentivirus and incubated with IL-2 for about 4-6 days. Cells were then stimulated with plate-bound anti-CD3 mAb for about 8 hours and analyzed for CD40L or CD25 surface expression by flow cytometry.
- Germline Tcfe3-/- mice have no reported defects in immune function, and the very early embryonic lethality caused by germline TFEB-deficicncy has precluded the study of Tcfeb-/- T cells. Therefore, the remaining models consistent with the biochemical, promoter, and genetic data prior to the experiment in FIG.5h were that TFEB itself is important for CD40L expression, or there exists a functional redundancy between TFE3 and TFEB with respect to CD40L expression. However, the TDN, by blocking both TFE3 and TFEB, does not allow discrimination between these models. To address this issue, in FTG.
- FIG. 5i depicts an illustrative representation of an immunoblot confirming down-regulation of TFEB in CD4+ T cells infected with a retrovirus that expresses the inhibitory stem-loop TFEB RNA as in FIG. 5h, according to an embodiment of the present invention.
- FIG. 5i depicts an immunoblot of extracts from sorted GFP+ cells infected with a control (left) or TFEB-slRNA lentivirus, for TFEB and GAPDH. The immunoblot is representative of two independent experiments.
- induction of CD40L by TCR-stimulation with an anti-CD3 mAb was comparable between wt T cells infected with the control or TFEB slRNA lentiviruses and Tcfe3-/- T cells infected with the control lentivirus, suggesting TFEB itself was not critical if TFE3 was present.
- CD40L induction on Tcfe3-/- T cells infected with the TFEB slRNA vector was significantly impaired compared to control infected and wt cells.
- Induction of CD25 was unaffected by the TFEB slRNA in all cases.
- These one or more embodiments of the present invention therefore represent the first known demonstration of a physiological functional redundancy between TFE3 and TFEB and the first known demonstration of their role in controlling T cell function via their joint regulation of CD40L (see, for example, FIGS. 5, 7, 8, and 9). It remains possible that under certain conditions o each has a unique contribution to the activation of CD40L based on observations that only TFEB was induced after TCR stimulation of primary cells (FIG. Ia and Ib) and that TFEB could bind independently from TFE3 to the CD40L promoter (FIG. 7).
- the present invention teaches us that TFE3 and TFEB control the expression of CD40L by directly controlling transcription of the gene encoding CD40L (Cd40lg and 5 CD40LG).
- FIG. 5j depicts an illustrative real-time RT-PCR analysis of RNA isolated from CD4+ T cells from wild-type or TDN-transgenic littermate mice at various times after incubation with mAb to CD3 (horizontal axis), according to an embodiment of the invention.
- the illustrative bar graph shows the relative amounts of mRNA encoding 0 CD40lg from CD4+ T cells from wild type (wt) (black bars) or TDN-transgenic littermate mice (white bars) at the indicated times after incubation with the anti-CD3 mAb.
- wild type wild type
- TDN-transgenic littermate mice white bars
- real-time RT-PCR analysis of CD40lg RNA isolated from TDN-transgenic and control CD4+ splenic T cells at various times after TCR-stimulation showed a reduction of Cd40lg transcripts in TDN-Tg T cells at the induction peak to one-third that of 5 non-Tg (control) cells, with unchanged overall kinetics (FIG. 5j).
- one or more embodiments of the present invention teach a fundamental 0 relationship between the transcription factors TFE3 and TFEB and T cell function via their direct regulation of CD40L expression.
- the data are most consistent with a mutually redundant role of these molecules with respect to controlling CD40L transcription, as a combined TFE3/TFEB deficiency inhibited TCR-dependent CD40L induction in the primary T cell culture systems, whereas individual deficiencies had either a minor or no measurable effect (see, for example, FIG. 5h).
- a degree of functional redundancy has also been observed with NFAT components, as a combined deficiency of NFATcI and NFATc2 resulted in the greatest impairment in CD40L expression compared to individual deficiencies.
- the transgenic approach to inactivate endogenous TFE3 and TFEB via T cell specific expression of the TDN protein was therefore a key and advantageous strategy to address the redundancy and also served to circumvent the embryonic lethality of germline TFEB deficiency by directing expression to somatic cells.
- One or more embodiments of the present invention teaches us that B cells from the T-cell specific TDN-Tg mice (FIGS. 1 e-lh) are not themselves defective and are capable of responding normally to antigen and CD40 signaling (FIG. 6). Consistent with that, CD 19+ B cells from TDN-transgenic mice and non-transgenic mice had similar expression of CD40 and responded similarly to in vitro stimulation, including the CD40-dependent induction of CD86 and major histo-compatibility complex (MHC) class II expression.
- MHC major histo-compatibility complex
- an agonist monoclonal antibody (mAb) to CD40 administered during immunization with TNP-KLH, enhanced day-7 TNP-specific IgG titers of both non-transgenic and TDN-transgenic mice, but most notably rendered the IgG responses indistinguishable from each other. Ih contrast, IgG antibody titers in mice treated with the isotype-matched control mAb were lower, and TDN-transgenic mice also had lower IgG responses than those of non-transgenic mice.
- FIG. 6a depicts an illustrative flow cytometry histogram showing equivalent CD40 expression in B cells from non-transgenic and TDN-transgenic mice, according to an embodiment of the present invention. Filled curves represent unstained cells, and grey lines represent cells stained with mAb to CD40.
- FIGS. 6b and 6c depict illustrative flow cytometry histograms showing that expressions of CD86 and MHC class II respectively, by B cells from TDN-Tg and non-Tg mice are equivalent, after stimulation of B cells with mAb to CD40, according to an embodiment of the present invention.
- filled curves represent CD86 or MHC class II staining on un-stimulated cells, and grey lines on stimulated cells.
- TNP-KLH is an experimental antigen to which an antibody response requires T-cell derived CD40L to B cells.
- Control mice (filled circles) and TDN-transgenic mice (open circles) were immunized intraperitoneally once with 100 micrograms of TNP-KLH plus 100 micrograms of either isotype-matched control antibody (control) or mAb to CD40 (3/23), delivered intravenously. The same amount of each mAb was administered once a day over 7 days. Data represent TNP-specific IgG isotypes from serum samples obtained on days 0 and 7.
- One or more embodiments of the present invention illustrate that TFE3 and TFEB bind to and activate the CD40L promoter.
- FIG. 7a depicts an illustrative schematic alignment of CD40L promoters from humans, mice and rats, showing where TFE3 and TFEB bind to and activate the CD40L promoter, according to an embodiment of the present invention.
- FIG. 7a depicts an illustrative schematic alignment of CD40L promoters from humans, mice and rats, showing where TFE3 and TFEB bind to and activate the CD40L promoter.
- E-boxes are categorized as either ones that match known and optimal MiT transcription factor binding sites and that are present in all promoters (gray boxes with letters) or as non-conserved and non-optimal (dotted open boxes).
- the optimal MiT sites are further subcategorized based on the precise sequence of the site, as represented by letters A through E. Nucleotide positions of boxes are numbered, with A of the ATG translation start site equal to +1 , as will be understood by those skilled in the art.
- FIG. 7b upper panel depicts an illustrative Chromatin immunoprecipitation (ChIP) of a Cd40lg promoter fragment by anti-TFE3 and anfi-TFEB antibodies, according to an embodiment of the present invention.
- the lower panel depicts illustrative controls for ChIP assays in the upper panel of FIG. 7b, according to an embodiment of the present invention. Titration of input DNA confirms amplification conditions in the ChIP assay are in the semi-quantitative, linear range.
- Input material (l/900th of total) from the upper panel of FIG. 7b was serially diluted as indicated above each lane and amplified in parallel with the experimental samples from FIG, 7b.
- Chromatin immunoprecipitation (ChIP) assays were used to determine whether endogenous TFE3 and TFEB could bind to the CD40L promoter in vivo. ChIP analysis of un-stimulated and TCR-stimulated primary CD4+ splenic T cells from wild-type (wt) mice showed that TFE3 and TFEB each bound to a fragment of the murine Cd40lg promoter under both resting and stimulated conditions.
- FIG. Tb depicts exemplary Chromatin immunoprecipitation (ChIP) of a Cd40lg promoter fragment by anti-TFE3 and anti-TFEB antibodies.
- FIG. 7b Shown is a semi-quantitative PCR analysis of starting material (Input, lanes 10-13) and immunoprecipitated material (ChIP, lanes 1-9) from the indicated sources and with the indicated Abs. ChIP using an anti-GAPDH antibody (lane 9) served as an additional negative control for background band intensity.
- the exemplary data shown in FIG. 7b is representative of three independent experiments.
- FIG. 7c also illustrates how TDN expression in T cells in vivo via transgenesis blocks TFE3 and TFEB binding to the CD40L promoter using a different methodology.
- FIG. 7c depicts illustrative electrophoretic mobility-shift assays (EMSAs) to evaluate the binding of native TFE3 and TFEB proteins to the individual MiT sites in the Cd40lg promoter, according to an embodiment of the present invention.
- Nuclear extracts (NE) from stimulated wild-type and Tcfe3-/- splenic CD4+ T cells were incubated with radio-labeled oligonucleotides spanning a single MiT E-box (sites 1-8) and complexes were resolved by native gel electrophoresis.
- the top two panels show relative E-box-binding activity in extracts made from WT (left) or Tcfe3-/- (right) CD4 T cells. The markers on the left point out shifted complexes and free probe.
- the bottom EMSAs represent cold oligonucleotide competition and antibody- interference assays to determine the identity and specificity of E-box-binding complexes in cell extracts.
- Each of the panels in the lower sets of EMSAs shows binding activity to one particular E-box probe from WT (left, first six lanes), TDN-Tg (left, last lane) or Tcfe3-/- (right panels) CD4 T cells.
- WT wild-type
- Tut point mutations
- anti-TFE3 or anti-TFEB were included in some binding reactions (+; above lanes).
- the exposures for the bottom panels were optimized for each probe. Data are representative of at least three independent experiments.
- FIG. 7d depicts illustrative electrophoretic mobility shift assays (EMSAs) of nuclear extracts of human embryonic kidney (HEK293) cells transfected with control (-), TFE3, or TFEB pEBB (pEBB is a plasmid used by many labs to express genes in mammalian cells; the "E” in EBB stands for El alpha, the name of the mammalian gene from where the promoter was obtained) expression vectors showing TFE3 and TFEB binding to the MiT sites from the Cd40lg promoter, according to an embodiment of the present invention. As shown in FIG.
- EBB electrophoretic mobility shift assays
- EMSAs with nuclear extracts from primary CD4+ T cells established that native TFE3 and TFEB bound to the multiple MiT consensus E-boxes in the Cd40lg promoter, although to differing relative degrees and sometimes with preferential binding of one or both subunits to individual sites. Similar results were obtained here (FIG. 7d) by analyzing DNA-binding activities in extracts of HEK293 cells over-expressing TFE3 or TFEB. We demonstrated the specificity of the protein-DNA interactions by including TFE3- or TFEB-speciftc antibodies that interfered with DNA binding.
- FIG. 8a depicts a bar graph plotting the relative activities of Cd40lg promoter fragments containing (full-length) or lacking (truncated) the MiT E-boxes and in the presence or absence of the TDN protein, as measured by (firefly) luciferase reporter gene assays, and shows that Cd40lg promoter activity depends on endogenous TFE3, TFEB and MiT boxes to enhance promoter activity, according to an embodiment of the present invention.
- mice Primary CD4+ splenic T cells from wild-type and TDN-transgenic mice were transfected with mouse Cd40lg promoter-luciferase constructs, either full-length (from the ATG to -1535 base pairs upstream) or truncated (to -382) lacking the MiT sites (rectangles).
- n three independent transfections using cells prepared from three separate sets of wild-type and TDN-transgenic mice.
- FTG. 8b depicts illustrative contributions of individual and combinations of MiT E-box sites to promoter activity, according to an embodiment of the present invention.
- Point mutations abrogating binding of TFE3 and/or TFEB binding 1 X 1 were introduced into E-box sites (numbered 1-8; left margin) in the context of the full-length mouse Cd40lg promoter.
- Primary splenic CD4+ T cells from wild-type and TcfeS-A mice were transfected, stimulated and analyzed for luciferase activity as in. described in FIG. 8a.
- Point mutations that abrogated the binding of TFE3 and/or TFEB to individual sites also attenuated Cd40lg promoter activity in primary wild-type and TFE3-deficient CD4+ T cells (FIG. 8b), although to differing degrees.
- a full-length construct containing mutations of all eight sites had the greatest effect on promoter activity, reducing it to the activity of the truncated promoter.
- Individual mutation of sites three through seven had a less severe but measurably attenuating effects in both wild-type and TFE3 -deficient T cells.
- mutation of site one or two had no effect in wild-type cells but had a substantial effect in TFE3-deficient T cells. That result was consistent with the preferential binding of TFEB from wild-type extracts to site one and the greater relative binding of TFEB to site two than to all other sites in the absence of TFE3.
- TFE3 -deficiency versus wild-type cells may indicate that the TFE3 and TFEB redundancy was not complete or that sites had differential responsiveness to TFE3 or TFEB protein abundance. These results indicated that all sites in some context could act in concert for full TFE3- and/or TFEB-dependent enhancement of Cd40lg promoter activity. Thus, we infer that TFE3 or TFEB binding is critical for achieving the physiological regulation of CD40L expression necessary for T cell-dependent antibody responses.
- FIG. 8c depicts that native TFE3 and TFEB can form heterodimers and that the TDN protein blocks this interaction, (Le., that heterodimerization, by way of example, is an interaction that is blocked by the TDN protein), according to an embodiment of the present invention
- the graphical depiction on the left represents an immunoblot of stimulated CD4+ splenic T cell extracts from WT, Tcfe3-/- and TDN-Tg mice for TFE3, TFEB, TDN (HA), and GAPDH protein expression.
- the graphical depiction on the right represents extracts from the same sources that were incubated with anti-TFE3, immune complexes that were precipitated, and immuno-precipitates resolved on SDS-PAGE and blotted with anti-TFEB.
- Ja wild-type CD4+ T cells, native TFE3 and TFEB were immuno-precipitated together (FIG. 8c lane 1), suggesting that, in addition to TFE3 homodimers and TFEB homodimers, that TFE3/TFEB heterodimers can form and may represent a distinct DNA binding species. That interaction was blocked in TDN-transgenic T cells (FIG 8c lane 3).
- maximal Cd40lg and CD40LG promoter activity required an upstream fragment that contains conserved MiT boxes and endogenous TFE3 and/or TFEB.
- the firefly luciferase reporter gene assay was used.
- murine Cd40lg and human CD40LG promoter fragments were linked to the luciferase gene and transiently transfected into the human T cell line Jurkat.
- Jurkat T cells were transfected either with full-length (-1.5 kB upstream: human -1562, mouse to -1535 relative to the ATG) or truncated (-382, mouse; -944, human) CD40L promoter-luciferase constructs with control or TDN-expressing plasmid and with Renilla as an internal control.
- TDN trans-dominant-negative
- the TDN protein contains the TFE3 HLH/Zip (helix-loop-helix/leucine zipper) dimerization domains but lacks the DNA binding BR and transcription activation domains and thus forms heterodimers with MiT proteins that are incapable of binding DNA (FIG. 8c).
- transient transfection of the reporters into Jurkat T cells showed that the activity of the full-length mouse Cd40lg and human CD40LG promoters exhibited about 3-fold higher activity at the induction peak following PMA/iono stimulation than the corresponding truncated promoters that lacked the consensus MiT binding sites.
- simultaneous expression of the TDN protein reduced the activity of the full-length promoters to the levels of the truncated promoter fragments, whereas the activities of the truncated promoters were unaffected by TDN expression.
- FIG. 9b is an illustrative graphical view depicting how inhibition of native TFE3 and TFEB activity by expression of trans-dominant negative protein (TDN) blocks endogenous CD40L induction after activation of Jurkat T cells, according to an embodiment of the present invention.
- TDN trans-dominant negative protein
- FIG. 9b illustrates that inhibition of TFE3 and TFEB activity by expression of the TDN via the pEBB plasmid blocks CD40L induction after activation of human Jurkat T cells.
- T cells were transiently transfected with the indicated pEBB-GFP-expression construct (control or TDN-expressing) and two days later stimulated with PMA/iono. About 12 hours later, cells were analyzed, for example, by flow cytometry, or alternative analysis methods, for surface expression of CD40L (left histogram) and CD25 (right histogram).
- CD40L and CD25 expression profiles of GFP+ (plasmid transfected) cells that were un-stimulated (filled profiles), cells that received the control plasmid and were stimulated (red line-overlays), and cells that received the TDN plasmid (green line overlay). Data shown is representative of at least three independent experiments.
- one or more embodiments of the present invention teaches us that endogenous TFE3 and/or TFEB are important for maximum induction of Cd40lg and CD40LG promoter activity and endogenous gene expression in mouse and human T cells and that they have evolutionarily conserved functions in that capacity (FIGS. 9a and 9b). In other words, what was defined in mouse CD4 T cells is also operational in human T cells (even though they are transformed).
- FIG. 10a depicts a table illustrating oligonucleotides for EMSA in FIG. 7, according to an embodiment of the present invention.
- the oligonucleotides are very short sequences of DNA that are the same MiT DNA sequences as they exist in the Cd40lg promoter. This is why and how we used them to show that TFE3 and TFEB proteins interact with the MiT sites in the Cd40lg promoter.
- FlG. 10a is merely an exemplary embodiment, and one or more embodiments of the present invention are not limited to the MiT sites used therein.
- One or more embodiments of the present invention any implement other MiT sites to bind to TFE3 and TFEB that do not come from the Cd40lg promoter.
- FIG. 1 Ob depicts an exemplary table illustrating oligonucleotides for cloning Cd40lg and
- CD40LG promoters according to an embodiment of the present invention.
- FIG. 10c depicts an exemplary table illustrating oligonucleotides for E-box mutagenesis for the analyses in FIG. 8b, according to an embodiment of the present invention.
- FIGS. 1 Ia-I Ic show that TDN expression in the B cell lineage does not block B cell development.
- one or more embodiments of the invention include experiments that are from different lines of transgenic mice in which the TDN inhibitor is expressed in B cells, which make antibodies, and T cells in these mice are normal and do not express the TDN inhibitor. These lines are called "B-cell-specif ⁇ c TDN-Tg mice.”
- FIG. 11a depicts a representative immunoblot showing that TFE3 and TFEB proteins are expressed in B cells, according to an embodiment of the present invention.
- FIG. 1 Ib depicts representative immunoblots probing for TDN expression in lymphoid tissues from a different line of E ⁇ P ⁇ transgenic mice that are shown here to express the TDN in B cells but not T cells (so called "B cell-specific TDN-Tg mice”), according to an embodiment of the present invention.
- FTG. 11c depicts representative flow cytometry, also showing that these mice express the TDN protein only in B, but not T cells, according to an embodiment of the present invention.
- FIG. 1 Id depicts representative flow cytometry analyses showing that major B and T cell populations in these "B cell-specific TDN-Tg" mice are normal, according to an embodiment of the present invention.
- FIG. 1 Ib and 1 Ic The immunoblot and flow cytometry shown in FIG. 1 Ib and 1 Ic is proof of this principle showing that the TDN inhibitor is made only in B cells in these lines, and that TFE3 and TFEB proteins are expressed in B cells.
- the flow cytometry analysis of lymphocytes in these mice shows that, remarkably, B cell development is normal (FIG. 11 c). This is important because it suggests that systemic administration of a hypothetical TFE3 and/or TFEB inhibitor will not effect the production of B cells.
- FIG. 12 is a flow chart illustrating a technique for treating at least one of an immunological disease and an inflammatory disease in a patient, according to an embodiment of the present invention.
- the technique for treating an immune disease in a patient includes step 1202, suppressing at least one of TFE3 and TFEB in a patient to thereby suppress CD40L expression in the patient.
- CD40L expression is typically elevated on T cells from SLE and RA patients.
- CD40L Inappropriate or undesirable expression of CD40L by T cells (or B cells and possibly other cell types) can cause autoantibody production by B cells and/or directly or indirectly (for example, via autoantibody complexes) trigger inflammatory responses mediated by the cellular immune system that destroy tissues and organs.
- CD40L is also necessary for B cells to become transformed by the Epstein Barr Virus. Interfering with CD40L-CD40 interactions - either genetically (in mice) or pharmacologically (for example, via CsA or interfering CD40L mAb) has had proven efficacy in mouse models and some efficacy in the clinic for ameliorating symptoms of and thus treating many autoimmune/inflammatory diseases.
- TFE3 and TFEB control the expression of CD40L in CD4 T cells and thus provides a new way to control abnormal or undesirable CD40L expression in disease states.
- TFE3 and TFEB can each be regulated by the MAPK pathway, which can be activated by stimulation of multiple receptors including the TCR and the 1L-15 receptor, and IL-15 is elevated in RA.
- MAPK pathway which can be activated by stimulation of multiple receptors including the TCR and the 1L-15 receptor, and IL-15 is elevated in RA.
- CD40L expression in other cell types may similarly depend on TFE3 and TFEB and the effects of disease state on their expression, in these cells.
- B cell abnormalities in some autoimmune diseases may be due to homotypic stimulation via co-expression of CD40 and CD40L on abnormal B cells.
- the step of suppressing at least one of TFE3 and TFEB may include selectively inactivating TFE3 and/or TFEB, respectively.
- the step of suppressing at least one of TFE3 and TFEB may alternatively include the step of blocking an ability of at least one of TFE3 and TFEB to interact with DNA.
- the step of suppressing at least one of TFE3 and TFEB includes the step of blocking an ability of at least one of TFE3 and TFEB synthesis.
- Suppressing TFE3 and/or TFEB may be achieved, in accordance with an aspect of the invention, by blocking TFE3 and/or TFEB synthesis, respectively, via an interfering RNA (which includes slRNA, as, by way of example only, in an embodiment of the invention). Blocking TFE3 and/or TFEB synthesis would leave other MiT proteins untouched and would be predicted to have the least invasive, least collateral off-target effects.
- the step of suppressing at least one of TFE3 and TFEB includes at least one of blocking an interaction between TFE3 and TFEB, blocking TFE3 and TFEB from forming dimers, and blocking TFE3 and TFEB from binding with DNA.
- Blocking TFE3 and TFEB from forming dimers may comprise blocking TFE3 and TFEB from forming dimers with each other, blocking TFE3 from forming dimers with itself, and/or blocking TFEB from forming dimers with itself.
- the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient may include the use of the suppressing TDN protein.
- Blocking TFE3 and/or TFEB protein activity via a TDN-like molecule or oligonucleotide MiT binding site DNA binding blockers may also affect Mitf and TFEC.
- molecular inhibitors of this type may have great efficacy if used locally, for example, at the site of a transplant or in bead form and released locally.
- TDN can experimentally block Mitf and TFEC as illustrated, for example, by the depiction herein that Mitf is expressed in thymocytes and un-stimulated T cells. Additionally, Mitf and TFEC may be capable of binding to the MiT sites in the CD40L gene promoters in other cell types that express these molecules. Ih one or more embodiments of the present invention the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient may include some type of interfering RNA (this includes the interfering stem-loop RNA, as was used here, and double stranded oligonucleotide RNAi).
- interfering RNA this includes the interfering stem-loop RNA, as was used here, and double stranded oligonucleotide RNAi).
- the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient may include some type of agent that would block the activation of TFE3 and/or TFEB by MAPK phosphorylation or prevent interactions between TFE3 and TFEB and upstream or downstream effectors that depend on this phosphorylation.
- tiie technique for treating at least one of an immunological disease and an inflammatory disease in a patient may comprise gene therapy, which may also benefit by including TFE3 and TFEB binding sites in the vector to promote appropriate expression of genes within the gene therapy vector in the patient.
- the immune disease includes at least one of an autoimmune disease and an inflammatory disease.
- the technique for treating an immune disease in a patient may also include step 1204, administering at least one of a TFE3 inhibitor and a TFEB inhibitor for suppressing at least one of TFE3 and TFEB, respectively, in the patient.
- At least one of the TFE3 inhibitor and the TFEB inhibitor is administered systemically. Also, in one or more embodiments of the present invention, at least one of the TFE3 inhibitor and the TFEB inhibitor is administered as an inhalant. Furthermore, in one or more embodiments of the present invention, at least one of the
- TFE3 inhibitor and the TFEB inhibitor is administered subcutaneously.
- the TFE3 inhibitor and the TFEB inhibitor may be administered at site of inflammation and/or rejection.
- CD4+ T cells were purified from spleens of 5 week old mice with a mouse CD4 negative isolation kit (Dynal Biotech) to greater than or equal to about 96% purity as determined by flow cytometry.
- T cells were cultured in RPMI supplemented with 10% (volume/volume) heat-inactivated FCS (fetal calf serum) (Gibco), and 50 ⁇ M ⁇ -mercaptoethanol (Sigma).
- FCS fetal calf serum
- Gibco fetal calf serum
- CD28 expression the cells were incubated with or without 1 O ⁇ g/ml anti-CD3 for 4Shr after initiation of the culture.
- T cells were incubated for 48 hours with 10 ⁇ g/ml anti-CD3 following the manufacturer's instructions (BD Pharmingen).
- BD Pharmingen the manufacturer's instructions
- For the real-time PCR freshly isolated and purified spleen CD4+ T cells were incubated in 10 ⁇ g/ml anti-CD3 fixed plates in RPMI/FBS/BME for 0 hour (hr), 3hr, 6hr, and 9hr before harvesting for RNA extraction and reverse transcription. Approximately 2xlO 7 cells were used per time point.
- CD4+ T cells were removed from anti-CD3 stimulation after 48hr, cultured for 7 days in Roswell Park Memorial Institute medium (RPMI)/ Fetal bovine serum (FBSybeta-mercaptoethanol (BME) containing 100 units/ml mouse IL-2 (Roche), then re-stimulated at 1O 5 cells/well in 96-welI plate bound with 5 ⁇ g/ml anti-CD3 without IL-2 for 48 hours. Supernatants were collected for quantitative enzyme-linked immunosorbent assay (ELISA).
- splenic B cells were purified with a mouse B cell Negative Isolation kit (Dynal Biotech) such that 94% or more of the cells were CD 19+. Purified B cells were then analyzed directly for CD40 expression or were stimulated for 48 hours with mAb to CD40 (3/23; BD Pharmingen) and then were analyzed for CD86 and MHC class II surface expression by flow cytometry.
- the 3' hemagglutin (HA) epitope-tagged TDN cDNA was subcloned into an immunoglobulin heavy chain gene enhancer and promoter based transgene cassette (E ⁇ P ⁇ ) that can direct expression in T cells, B cells or both, depending on the line. Fragments that contained the transgene cassette were excised from the plasmid backbone, purified and microinjected into fertilized FVB oocytes as described in the SUNY-Downstate Transgenic Facility (see, e.g., Hogan, B., Beddington, R., Costantini, F. & Lacy, E, "Manipulating the Mouse Embryo: A Laboratory Manual," Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1994, which is incoipoiated by reference herein).
- E ⁇ P ⁇ immunoglobulin heavy chain gene enhancer and promoter based transgene cassette
- mice were identified by Southern blotting and PCR of genomic DNA from tails. Transgenic mice analyzed were the progeny of transgenic animals that had been back-crossed at least four generations onto the C57BL/6 strain. Five independent lines were derived that were confirmed to express TDN transgene only in T cells. Three lines were evaluated for recapitulation of impaired induction of CD40L in primary CD4+ T cells. All showed the same defect, and results from one line is shown. All transgenic mice used were 5-6 weeks old and hemizygous for the transgene, and non-transgenic control mice were sex-matched Iittermates of transgenic mice. The mice were housed under SPF (specific pathogen free) conditions according to guidelines approved by the Division of Laboratory Animal Resources.
- SPF specific pathogen free
- control and TFEB slRNA lentiviruses are described in Huan et al. and viral particles produced according to published protocols (see, for example, Rubinson, D.A. et al., "A lenti virus-based system to functionally silence genes in primary mammalian cells, stem cells and transgenic mice by RNA interference;," Nat Genet 33, pp. 401-406 (2003), which is incorporated by reference herein).
- pMDLg/pRRE packaging vectors
- p refers to "plasmid”
- MDLg and RRE are conventional names for viral genes that encode a function necessary to build viral particles, so called “helper” functions
- CMV-VSVG CVM refers to transcription activation sequences from the cytomegalo virus that are built into plasmids to express other genes, in this case the G protein, from VSV; and VSVG is a name for viral genes that encode a function necessary to build viral particles
- RSV-Rev RSV refers to transcription activation sequences from the Rous sarcoma viruses that are built into plasmids to express other genes, in this case the Rev protein from HIV; and Rev is a name for viral genes that encode a function necessary to build viral particles).
- the viruses in culture media were concentrated by centrifugation, and used for infecting purified mouse spleen CD4+ T cells.
- the infected T cells were cultured with 20 units/ml IL-2 for 4-5 days before evaluating infection efficiency and anti-CD3 stimulation.
- Fc receptor block 2-4G2, BD Pharmingen
- Fc receptor block 2-4G2, BD Pharmingen
- FITC 5 PE or Cy-Chrome (BD Pharmingen) or with fluorescein isothiocyanate-conjugated anti-MHC class II (Caltag).
- TDN protein for detection of the TDN protein in transgenic lymphocytes, cells were treated with Fix & Perm Kit (Caltag), before being stained with FITC conjugated anti-HA mAb (3F10, Roche).
- ChIP assay was done as described (see, for example, Huan, C, Sashital, D., Hailemariam, T., Kelly, M.L. & Roman, C.A.J., "Renal carcinoma associated transcription factors TFE3 and TFEB are leukemia-inhibitory factor-responsive transcription activators of E-cadherin," J Biol Chem 13, p.
- RNA from purified CD4+ splenic T cells was isolated with TRI reagent (TR-118, MAR) and contaminant DNA was removed by DNase I (AMP-Dl, Sigma) according to the manufacturer s instructions.
- Total RNA (5 ⁇ g) was reverse-transcribed and PCR amplified with QuantiTect SYBR Green RT-PCR kit (Q ⁇ agen) in an Opticon Continuous Fluorescence Detector (MJ Research).
- CD40L and GAPDH inRNAs were determined by real-time RT-PCR using following primers, for CD40L, 5' AAAATGGG AAAC AGCTGACG 3', and 5' GGTATTTGCCGCCTTGAGTA 3 ⁇ for GAPDH, 5' TCACCACCATGGAGAAGGC 3% and 5' GCTAAGCAGTTGGTGGTGCA 3'.
- CD40L mRNA expression was normalized to GAPDH mRNA.
- TMB tetramethylbenzidine
- KPL peroxidase substrate system
- mice were immunized intraperitoneally with 100 ⁇ g TNP-KLH (Biosearch Technologies) in complete Freund's adjuvant (Difco) and were injected intravenously each day for seven days after immunization with 100 ⁇ g anti-CD40 (mAb 3/23; BD Pharmingen) or the isotype control rat IgG2a K (mAb R35-95; BD Pharmingen), and then blood was collected to obtain £erum.
- Serum Igs specific for TNP Trinitrophenol
- TNP-BSA Trinitrophenol-bovine serum albumin conjugate
- ELISA was performed with EL405 auto plate washer (BIO-TEK), the plates were read at 450nm with ⁇ Quant microplate spectrophotometer (BIO-TEK), and data was analyzed with
- KCjunior microplate data analysis software BIO-TEK.
- T cells culture supematants were assayed for cytokine content by Mouse IL-4 Immunoassay Elisa kit
- mice were immunized intraperitoneally with 7.5 ⁇ l SRBC (Colorado Serum) in 200 ⁇ l PBS. The spleens were harvested S days after immunization and fixed in 10% formalin. Germinal centers were stained as previously described (see, for example, Cattoretti, G. et al., "BCL-6 protein is expressed in germinal-center B cells," Blood 86, pp. 45-53 (1995), which is incorporated by reference herein) with modification.
- antigen-retrieved slides were blocked with 3% pig serum and avidin/biotin blocking kit (Vector Lab), incubated with l ⁇ g/ml of biotin-conjugated PNA (Sigma) for more than 2 hr, and then incubated with biotin-conjugated goat anti-PNA (Vector Lab) for 45 min. Subsequently, the sections were treated with 0.1% NaN 3 and 0.3% H 2 O 2 for 30 min to block the endogenous peroxidase before staining with 1 :400 diluted HRP -conjugated avidin (Dako) for 20 min. After washing, the sections were incubated with HRP developing solution for 20 min, and then embedded with glycerol gelatin. All staining procedures were performed at room temperature.
- Nuclear extracts from anti-CD3-activated CD4+ T cells or transfected HEK293 cells (5 ⁇ g) were incubated on ice for 30 minutes with a 32 P-labeled oligonucleotide spanning an E-box site from the mouse Cd401g promoter in 25 mM HEPEs, pH 7.9, 50 mM KCl, 4% (weight/volume) Ficoll, 5 ⁇ M ZnCl 2 , 0.1 mM dithiothreitol, 0.02% (volume/volume) Nonidet-P40, 5 mM MgCl 2 , 10 ⁇ g/ml of BSA and 10 ng/ ⁇ l of poly(dI:dC) (deoxyinosinerdeoxycytidine).
- Nonspecific binding was assessed in the presence of a 100-fold excess of unlabeled oligonucleotide corresponding to the labeled oligonucleotide with or without E-box mutation and 2.5 ⁇ g anti-TFE3 (BD Pharmingen) or anti-TFEB (Santa Cruz). Samples with antibody were incubated for an additional 30 minutes. Reaction products were separated by native 5% (weight/volume) PAGE at 4°C and were visualized by autoradiography.
- CD40L promoter reporter gene plasmid construction transient transfection. and reporter gene assay
- the 5 extended (“full-length”: to -1535 bp and -1562 bp, relative to the mouse and human ATG, respectively) and truncated mouse and human CD40L promoter fragments (to -382 bp and -944 bp from the ATG, respectively) were PCR amplified from genomic DNA and inserted into the pGL3 luciferase reporter vector (Promega).
- Mouse Cd40lg promoter reporter gene plasmids with E-box mutations were constructed by site-directed mutagenesis (Stratagene) with specific primers.
- Jurkat cells were transfected by electroporation with lO ⁇ g of CD40L promoter reporter gene plasmid, 0.05 ⁇ g renilla control plasmid and 8 ⁇ g TDN expressing plasmid for each sample, and subsequently activated with PMA and ionomycin as described (see, for example, Lobo, F.M., Xu, S., Lee, C. & Fuleihan, R.L., "Transcriptional activity of the distal CD40 ligand promoter," Biochem Biophys Res Commun 279, pp. 245-250 (2000), incorporated herein by reference).
- Firefly luciferase (Luc) and Renilla renifo ⁇ nis luciferase (Rlhic) activities were measured from cell extracts with the Dual Luciferase Reporter Assay System (Promega) and TD-20/20 Luminometer (Turner Designs).
- Primary mouse CD4 T cells were transfected with 2.5 ⁇ g of Cd401g promoter reporter gene plasmids, 0.01 ⁇ g of RLluc and 1.5 ⁇ g of empty pEBB or pEBB-TDN expression plasmid for each sample using the mouse T cell Nuclefector kit (Amaxa).
- the cells were activated for eight hours with 5 ⁇ g/ml of mAb to CD3 ⁇ and then were lysed for luminornetry as described herein. Luciferase activity was always normalized to Rlluc activity. Ia all experiments, the total amount of pEBB expression vector DNA was equalized by balancing cDNA-containing pEBB with empty pEBB,
- the blots were developed by enhanced chemiluminescence (ECL). Coimmunoprecipitation was done according to a protocol from BD-Pharmingen. Purified CD4+ splenocytes (2 x 107) were lysed in imraunoprecipitation buffer (1% (weight/volume) Triton X-100,
- the spleen IgM anti-SRBC response was assayed 5 days after intraperitoneal administration of 0.5-7.5 ⁇ l SRBC in 200 ⁇ l PBS following the protocol of the Jerne plaque assay (see, for example, Jerne, N.K. et al., "Plaque forming cells: methodology and theory,” Transplant Rev 18, pp. 130-191 (1974), incorporated by reference herein).
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Genetics & Genomics (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Techniques for treating immune dysfunction in a patient by regulation of CD40 Ligand (CD40L) expression are provided. For example, one technique includes suppressing TFE3 and/or TFEB in the patient to thereby suppress CD40L expression in the patient. In addition, the technique may include administering a TFE3 inhibitor and/or a TFEB inhibitor for suppressing TFE3 and/or TFEB, respectively, in the patient.
Description
METHOD FOR TREATING IMMUNE DYSFUNCTION BY REGULATION OF CD40 LIGAND EXPRESSION
Cross-Reference to Related Application s) This application claims the benefit of U.S. Provisional Application Serial No. 60/750,611 , filed on December 15, 2005, the disclosure of which is incorporated by reference herein.
Field of the Invention
The present invention relates to immunology and, more particularly, to the treatment of immune dysfunction by the regulation of CD40 ligand (CD40L) expression.
Background of the Invention
The immune system is a responsive protection system comprised of cells in bone marrow, the thymus, and the lymphatic system of ducts and nodes, spleen, and blood. Aspects of the immune system include the innate immune response and the adaptive immune response. An innate immune response is nonspecific, and includes, for example, skin, cilia in mucous membranes, tears, saliva, nasal secretions, and phagocytic cells that migrate to infected areas and engulf pathogens. An adaptive immune response is a specific response against an individual antigen, and includes humoral and cellular systems. The humoral system produces antibodies to eliminate pathogens and their products. The cellular system eliminates pathogens that have invaded cells and regulates the body's entire immune response.
The humoral system response involves B cells, which are specialized white blood cells produced in the bone marrow. Every B cell contains multiple copies of one kind of antibody as a surface receptor for antigen. When the antibody on the surface of a B cell binds to an antigen, the cell can be stimulated to undergo a process called clonal selection. This process includes proliferation and differentiation, where the cells produced make the same antibody, but become memory cells and plasma cells. Memory cells insure that subsequent infections by that particular pathogen receive a quicker and more efficient response. Plasma cells secrete large quantities of the antigen-specific antibody. The antigen-specific antibody forms complexes with free pathogens and their harmful products, inactivating pathogens and stimulating other innate systems including phagocytic cells and complement to eliminate the danger from extracellular fluids.
The cellular system responds to cells containing pathogen display antigen fragments on their cell surfaces. Receptors on the surface of CD8 cells (cytotoxic T cells) can detect the presence of pathogen specific antigen fragments and activate a killing response known as
apoptosis that leads to the death of the infected cell. CD8 cells must interact with CD4 cells (helper T cells) to regulate destruction of infected cells. CD4 cells regulate other ceils of the immune system through secretion of molecules called cytokines. Furthermore, CD4 cells are generally required for the clonal selection of B cells, as described above. CD40 ligand (CD40L, also referred to as CD154, and gp39 (glycoprotein of 39 kiloDaltons) is a critical effector molecule expressed mainly by activated CD4+ T cells and is essential for thymus (T)-dependent immunity. Surface CD40L expressed by activated T cells is required to activate B cells and monocytes via CD40 molecules expressed on those cells. Humans and mice that lack CD40L function suffer from the X-linked form of hyper immunoglobulin M (IgM) syndrome, a severe immune deficiency disease characterized by pyogenic bacterial infections due to the inability of B cells to produce affinity matured antibodies and isotypes other than IgM, and by opportunistic infections due to defective macrophage activation.
Conversely, abnormal CD40L expression by T cells is associated with multiple human immunological and inflammatory diseases such as, but not limited to, systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). CD40L is also expressed by other cells, such as, for example, endothelial cells and platelets, where it is thought to have a role in inflammation and hemostasis.
Given the relationship of CD40L to human disease and the importance of its controlled expression, knowledge of the transcription factors that regulate CD40L expression in vivo is critically important for understanding normal immune regulation. Accordingly, there exists a need for techniques to manipulate CD40L expression as a way of treating human disorders in which such CD40L expression is abnormal, and yet does not suffer from one or more of the problems exhibited by conventional treatment methodologies.
Summary of the Invention
Principles of the present invention provide techniques for treating immune dysfunction by regulation of CD40 Ligand expression.
For example, in one aspect of the invention, a technique treating at least one of an immunological disease and an inflammatory disease in a patient includes the step of suppressing at least one of transcription factor binding to μ enhancer site 3 (TFE3) and transcription factor EB (TFEB) in the patient to thereby suppress CD40L expression. TFE3 and TFEB may be suppressed, in accordance with a preferred embodiment of the invention, by blocking the synthesis of at least one of TFE3 and TFEB, and/or by blocking an ability of the TFE3 and TFEB molecules to interact with each other and/or interact (for example, bond) with deoxyribonucleic
acid (DNA). Shown in the drawings that support the present invention are the effects of two types of TFE3/TFEB inhibitors on CD40L expression, a trans-dominant negative protein (TDN) and stem-loop RNA mediated interference (slRNA).
These and other features, objects and advantages of the present invention will become apparent from the following detailed description of illustrative embodiments thereof, which is to be read in connection with the accompanying drawings.
Brief Description of the Drawings
FIG. Ia depicts an illustrative immunoblot of TFE3 and TFEB in primary CD4+ mouse splenocytes and human Jurkat T cells, according to an embodiment of the present invention;
FTG. Ib depicts an illustrative time course of TFE3 and TFEB protein expression in mouse CD4 T cells after TCR stimulation, according to an embodiment of the present invention;
FIG. Ic depicts an illustrative analysis of Mitf protein expression in mouse CD4 and human Jurkat T cells before and after stimulation and in mouse thymocytes, according to an embodiment of the present invention;
FIG. Id depicts an illustrative semi-quantitative reverse transcriptase dependent- polymerase chain reaction (RT-PCR) assay to measure relative abundance of Tcfe3, Tcfeb, and Mitf RHSTA transcripts in resting and stimulated CD4+ T cells over a time course, according to an embodiment of the present invention; FIG. 1 e is an illustrative schematic view depicting the EμPμ-TDN ("Eμ" and "Pμ" refer to the immunoglobulin μ heavy chain intronic enhancer and promoter, respectively) transgene vector used in one or more embodiments of the present invention;
FIG. If depicts an illustrative immunoblot of TDN protein in extracts from total bone marrow (BM), total spleen (Spleen) and total thymocytes (Thymus) from trans-dominant negative-transgenic (TDN-Tg) mice (+) but not in the same types of cells isolated from non-transgenic (non-Tg) littermates (-), according to an embodiment of the present invention;
FIG. Ig depicts illustrative flow cytometry histograms of permeabilized lymphocytes from non-Tg and the T cell-specific TDN-Tg mice for intensity of staining with an anti-hemagglutinin (-HA) antibody that detects the TDN protein, according to an embodiment of the present invention;
FIG. Ih depicts an illustrative immunoblot of TDN protein present in extracts of purified CD4+ T cells, but not splenic B cells, from TDN-transgenic mice, according to an embodiment of the present invention;
FIGS. 2a and 2b are illustrative graphical views of cell counts and flow cytometry depicting how numbers and a distribution of major B and T cell populations is normal in TDN-transgenic mice, according to an embodiment of the present invention;
FIGS. 3a and 3b are illustrative graphical views using imrnunohistochemistry and flow cytometry to depict impaired germinal center formation in TDN-transgenic mice, according to an embodiment of the present invention;
FIGS. 4a through 4d are illustrative graphical views of ELISA assays depicting impaired T-dependent, but normal T-independent, humoral responses in TDN-transgenic mice, according to an embodiment of the present invention; FIG. 5 a depicts an illustrative flow cytometry demonstrating impaired surface expression of CD40L by TDN-transgenic (TDN) as compared to non-Tg control (wild type (WT)) CD4+ T splenocytes stimulated with monoclonal antibodies (niAb) to CD3 and analyzed at 8 hours, according to an embodiment of the present invention;
FIG. 5b depicts an illustrative quantification of flow cytometry histogram data for percent CD4+ T cells positive for CD40L (left) and CD25 (right) after S hours of stimulation with various amounts of mAb to CD3 (Anti-CD3), according to an embodiment of the invention;
FIGS. 5c and 5d depict illustrative flow cytometry demonstrating comparable levels of ICOS (Inducible Co-stimulator of T cells) and CD28, respectively, by control (wild type (wt)) and TDN-transgenic (TDN) CD4+ splenocytes, according to an embodiment of the present invention; FIG. 5e depicts an illustrative graphical representation of ELISA (enzyme linked immunoadsorbance assay) analyses showing comparable levels of interleukin-4 (IL-4) secretion by control (wild type (WT)) and TDN-transgenic (TDN) CD4+ splenocytes, according to an embodiment of the present invention;
FIG. 5 f depicts an illustrative bar graph showing normal proliferation of TDN-Tg T cells in response to CD3/CD28 stimulation, according to an embodiment of the present invention;
FIG. 5g depicts a representative semi-quantitative RT-PCR analysis showing that Smad 7 expression is normal in TDN-Tg splenic T cells, according to an embodiment of the invention;
FIG. 5h depicts an illustration of representative flow cytometry histograms depicting impaired GD40L but normal CD25 expression by TFE3 -deficient (TcfeS-/-) CD4 T cells infected with a lentivirus that expresses an interfering stem-loop RNA against TFEB, according to an embodiment of the present invention;
FIG. 5i depicts an illustrative representation of an immunoblot showing the expected down-regulation of TFEB protein in CD4+ T cells infected with a retrovirus that expresses the
interfering stem-loop TFEB RNA in support of the data in FIG. 5h} according to an embodiment of the present invention;
FIG. 5j depicts an illustrative real-time RT-PCR analysis of ribonucleic acid (RNA) from CD4+ T cells showing reduced abundance of CDAQIg transcripts in CD4+ T cells from TDN-transgenic mice relative to non-transgenic littermates at various times after incubation with mAb to CD3 (horizontal axis), according to an embodiment of the invention;
FIG. 6a depicts an illustrative flow cytometry histogram demonstrating indistinguishable CD40 expression in B cells from non-transgenic and TDN-transgenic mice, according to an embodiment of the present invention; FIGS. 6b and 6c depict illustrative flow cytometry histograms, demonstrating equivalent expressions of CD86 and major histocompatibility complex (MHC) class II, respectively, after stimulation of B cells from non-transgenic and TDN-transgenic mice with mAb to CD40, according to an embodiment of the present invention;
FIG. όd depicts an illustrative ELISA measuring immunoglobulin G (IgG) responses to TNP-KLH (Trinitrophenol-keyhole limpet hemocyanin) which shows equivalent responses in non-transgenic and TDN-transgenic mice treated with an agonist mAb to CD40 during immunization, according to an embodiment of the present invention;
FIG. 7a depicts an illustrative schematic alignment of promoters for the gene encoding
CD40L from humans, mice and rats, showing MiT (the MitfTTFE family) sites (a subset of E-boxes; "E" refers to "enhancer" boxes from the IgH enhancer) where TFE3 and TFEB bind to and activate the CD40lg and CD40LG promoters, according to an embodiment of the present invention
FIG. 7b depicts an illustrative Chromatin immunoprecipitation (ChIP) of a CD40lg promoter fragment by anti-TFE3 and anti-TFEB antibodies, as well as the illustrative controls for the ChIP assays, according to an embodiment of the present invention;
FIG. 7c depicts illustrative electrophoretic mobility-shift assays (EMSA) demonstrating the binding of native TFE3 and TFEB proteins present in CD4 T cell extracts to individual MiT sites from the Cd40lg promoter, according to an embodiment of the present invention;
FIG. 7d depicts illustrative electrophoretic mobility shift assays (EMSAs) of nuclear extracts of human embryonic kidney (HEK293) cells transfected with control (-), TFE3, or TFEB pEBB expression vectors showing TEE3 and TFEB binding to the MiT sites from the Cd40lg promoter, according to an embodiment of the present invention;
FIG. 8a is a bar graph depicting illustrative luciferase assays to measure Cd40lg promoter activity in CD4 T cells from non-Tg and TDN-Tg mice, showing that a CD40lg fragment
containing the MiT sites depends on endogenous TFE3 and TFEB to enhance promoter activity, according to an embodiment of the present invention;
FIG. 8b is a bar graph depicting illustrative luciferase assays showing the contributions of individual and combinations of MiT sites to Cd40lg promoter activity in non-Tg, TDN-Tg, and Tcfei-f- CD4 T cells, according to an embodiment of the present invention;
FlG. 8c depicts immunological assays that show that native TFE3 and TFEB in CD4 T cells can form heterodimers, an interaction that is blocked by the TDN protein, according to an embodiment of the present invention;
FIG. 9a is a bar graph depicting illustrative luciferase assay showing the importance of MiT sites and endogenous TFE3 and TFEB for the human CD40LG and mouse CD40lg promoter activity in human Jurkat T cells, according to an embodiment of the present invention;
FIG. 9b is an illustrative graphical view of a flow cytometry histogram depicting how inhibition of TFE3 and TFEB activity by expression of the TDN protein blocks endogenous CD40L induction after activation of human Jurkat T cells but that CD25 induction is not affected, according to an embodiment of the present invention;
FIG. 10a depicts a table illustrating oligonucleotides for EMSA in FIGS. 7, including SEQ. ID NOs: 1-16, according to an embodiment of the present invention;
FIG. 10b depicts a table illustrating oligonucleotides for cloning Cd40lg and CD40LG promoters, including SEQ. ID NOs: 17-22, according to an embodiment of the present invention; FIG. 10c depicts a table illustrating oligonucleotides for E-box mutagenesis, including
SEQ. ID NOs: 23-30, according to an embodiment of the present invention;
FIG. 11a depicts an immunoblot showing that TFE3 and TFEB proteins are expressed in B cells, according to an embodiment of the present invention;
FIG. lib depicts a representative immunoblot showing TDN protein in B cell containing lymphoid organs from B-cell-specific TDN-Tg mice, according to an embodiment of the present invention;
FIG. lie depicts representative flow cytometry histograms showing that the TDN protein is expressed only in B, but not T cells in "B-cell-specific TDN-Tg" mice, according to an embodiment of the present invention; FIG. 1 Id depicts representative flow cytometry dot plots showing that major B and T cell populations in these "B cell-specific TDN-Tg" mice are normal according to an embodiment of the present invention; and
FlG. 12 is a flow chart illustrating a technique for treating at least one of an immunological disease and an inflammatory disease in a patient, according to an embodiment of the present invention.
Detailed Description of the Invention
One or more embodiments of the present invention meet the above-noted need by providing methods of treating an immune disease in which CD40L expression is abnormal or otherwise drives pathology. The invention, in an illustrative embodiment thereof, beneficially exploits an advantageous role of transcription factor binding to immunoglobulin heavy chain μ (IGHM) enhancer (μE) site 3 (TFE3) and transcription factor EB (TFEB) in regulating CD40L expression. One or more embodiments of the present invention teach, for example, that TFE3 and TFEB directly activate CD40L at the transcriptional level in vivo without significantly affecting the synthesis of other activation and effector molecules, Biactivation of TFE3 and TFEB attenuates the induction of CD40L expression on T cells. TFE3 and TFEB are broadly expressed transcription factors related to the transcription factor Mitf. Although they have been linked to cytokine signaling pathways in non-lymphoid cells, their function in T cells is unknown. TFE3 -deficient mice are phenotypically normal, whereas TFEB deficiency causes early embryonic death. One or more embodiments of the present invention illustrate that combined inactivity of TFE3 and TFEB in T cells result in a hyper-immunoglobulin M (hyper-IgM) syndrome due to impaired expression of the CD40 ligand gene (Cd40lg) by CD4+ T cells.
As previously stated, abnormal CD40L expression by T cells is associated with multiple human immunological and inflammatory diseases such as, but not limited to, systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). CD40L is also expressed by other cells, such as, for example, endothelial cells and platelets, where it is thought to have a role in inflammation and hemostasis.
As used herein, "CD40L" refers to the protein, "Cd40lg" refers to the name of the mouse gene encoding the CD40L protein, and "CD40LG" refers to the human gene. Similarly, TFE3 and TFEB refer to the proteins, whereas Tcfe3 and Tcfeb refer to the mouse genes. Native TFE3 and TFEB bound to multiple cognate sites in the promoter of the gene encoding CD40 ligand, and maximum Cd40lg promoter activity and gene expression required TFE3 or TFEB. Consequently, as illustrated by one or more embodiments of the invention, TFE3 and TFEB are direct, physiological and mutually redundant activators of Cd40lg expression in activated CD4+ T cells critical for T cell-dependent antibody responses.
The teachings of one or more embodiments of the present invention relate generally to the regulation of CD40L expression. Control of CD40L expression by T cells is complex and highly regulated, befitting its central role in immune activation and modulation. Although one or more embodiments of the present invention is described herein in the context of T cells and B cells, it is contemplated that the techniques of the invention may be used for controlling CD40L expression by immune cells in general. The phrase "immune cells" as used herein is intended to include (but is not limited to) hematopoietic and endothelial cells.
CD40L is rapidly and transiently induced upon stimulation of naive T cells via the T cell receptor (TCR), with surface levels peaking by 6-8 hours and then declining. This profile of TCR-dependent CD40L induction primarily reflects transcriptional activity. CD40L expression can be prolonged and augmented by additional stimulatory input from co-stimulatory and/or accessory molecules such as, but not limited to, CD28, CD2, LFA-I (lymphocyte-function associated antigen-1), and CD43, and interleukins, such as TL-2, IL-12, and IL-15, via mechanisms that involve enhanced transcription, increased CD40L messenger-RNA (mRNA) stability, or both. Consequently, the overall dynamics of CD40L expression is dependent on TCR stimulatory conditions, activation and/or effector status, developmental stage of the T cell, and age.
Mice engineered to express CD40L via heterologous promoters in T cells have been shown to develop lymphoproliferative disorders and tumors, highlighting the importance of regulated CD40L transcription (see, for example, Brown, M.P. et al., "Thymic lymphoproliferative disease after successful correction of CD40 ligand deficiency by gene transfer in mice," Nat Med 4, pp. 1253-1260 (1998), and Sacco, M.G. et al., "Lymphoid abnormalities in CD40 ligand transgenic mice suggest the need for tight regulation in gene therapy approaches to hyper immunoglobulin M (IgM) syndrome," Cancer Gene Ther. 7, pp. 1299-1306 (2000), the disclosures of which are incorporated by reference herein).
Multiple transcription factors and cognate promoter and enhancer regulatory elements responsive to the TCR and co-stimulatory receptor stimulation have been identified that regulate the transcription of CD40L. Like many effector and activation molecules induced by T cell stimulation, such as, for example, IL-2 and tumor necrosis factor (TNF) family genes, CD40L induction is dependent on subunits of nuclear factors of activated T-cells (NFAT), a Ca2+-dependent transcription factor complex. Additional regulators include subunits of AP-I (Activator Protein- 1) and NFkB (Nuclear Factor that binds to the kappa enhancer B site) and Egr-1 (early growth response-1), which were also shared by other TCR and/or co-stimulator responsive genes, and AKNA. Binding sites for other transcription factors, such as, for example,
STAT5, GATA-3, Oct-1 subunits, have also been identified (see, for example, Crow, M.KL & Kirou, K.A, "Regulation of CD40 ligand expression in systemic lupus erythematosus," Curr Opin Rheumatol 13, pp. 361-369 (2001), and Cron, R. Q, "CD154 transcriptional regulation in primary human CD4 T cells," Immunol Res 27, pp. 185-202 (2003), which are incorporated by reference herein), but their physiological contribution to CD40L transcription is not yet known.
Despite these commonalities, the dynamics of CD40L expression and its responsiveness to various stimuli are distinct from other molecules induced by T cell activation and therefore suggest that the regulation of CD40L transcription in T cells may involve as yet unknown factors/ or elements. Similarly, relatively less is known about how CD40L transcription is regulated in non-lymphoid cells.
Given the relationship of CD40L to human disease and the importance of its controlled expression, knowledge of the transcription factors that regulate CD40L expression in vivo is critically important for understanding normal immune regulation and for designing strategies to manipulate CD40L expression as a way to treat the multiple human disorders in which CD40L expression is abnormal or undesirable.
In accordance with one embodiment of the present invention, the transcription factors TFE3 and TFEB are demonstrated to be physiological transcription regulators of CD40L expression in T cells. TFE3 is a member of the helix-loop-helix family of transcription factors that binds to the mu-E3 (μE3) motif of the immunoglobulin heavy-chain enhancer and is expressed in many cell types. TFEB is a transcription factor with a basic region-DNA binding domain, a helix-loop-helix and leucine zipper dimerization domains, and a nuclear localization signal, thought to be located adjacent to the helix-loop-helix domain. TFEB is ubiquitously expressed.
TFE3 and TFEB are closely related members of a functionally interactive DNA binding family known as Mitf/TFE (MiT), that includes the microphthalmia transcription factor Mitf and TFEC (Transcription factor E-box C). MiT proteins bind to so called μE3 sites, a subset of E-boxes that match a general CANNTG consensus sequence, with those binding to TFE3 in vitro first identified and characterized in immunoglobulin heavy-chain and T cell receptor (TCR) enhancers. DNA binding is mediated by nearly identical basic regions (BRs) and requires dimer (either homodimer or heterodimer) formation between MiT family members mediated by conserved helix-loop-helix (FILH) and leucine zipper (LZ) domains.
MiT proteins share similar structures and are often expressed together, yet genetic studies have demonstrated both overlapping and non-overlapping functions for MiT proteins in different cell-types. Mitf, a well-characterized family member, is expressed mainly in pigment and
myeloid cells, where it is involved in melanocyte and mast cell development as a transcription mediator of the c-kit pathway,- and is a negative regulator of B cell activation and terminal differentiation. TFE3 and Mitf serve redundant roles in osteoclast development as transcriptional mediators of the macrophage colony-stimulating factor (m-csf) pathway. These functions of Mitf correspond to its cell-type and/or lineage restricted expression pattern.
Like expression of Mitf, TFEC expression is restricted mainly to the myeloid lineage, but TFEC-deficient mice are phenotypically normal, even though TFEOdeficient macrophages have lower expression of a subset of interleukin 4 (IL-4) -responsive transcripts, including the transcript for granulocyte-macrophage colony-stimulating factor. In contrast, TFE3 and TFEB are more broadly expressed, and relatively less is understood about their individual biological functions, primarily because germline TFE3-deficient animals have no reported defects. Studies of cultured non-lymphoid cell lines have indicated that TFE3 facilitates the activation of a subset of genes dependent on transcription factor Smad3 (Mothers against decapentaplegic homolog 3), in response to transforming growth factor-β (TGF-β) including those encoding components of the extracellular matrix and Smad7, a TGF-β pathway inhibitor. Ectopic over-expression of TFE3 in hepatocytes in mice promotes glycogen synthesis. However, mice deficient in TFE3 are phenotypically normal, with no defects noted in development, reproduction or the immune response. In contrast, TFEB deficient embryos die early in gestation because of defects in placental vascularization. The function of TFEB in adults is not known.
Given the extensive amino acid sequence similarities and overlapping expression profiles, a possible explanation for the former observation (that is, that Tcfe3-/- mice are normal) is that TFEB and TFE3 are functionally redundant in many tissues and, to varying degrees, TFEB may compensate for a TFE3-deficiency. By selectively inactivating these TFE3 and TFEB molecules, the present invention, in accordance with an illustrative embodiment thereof, exploits a central role of TFE3 and TFEB in the immune system via their control of CD40L expression, and thereby provides a beneficial methodology for treating immune dysfunction. The phrase "immune dysfunction" as used herein is intended to refer to any syndrome in which activity of imrnunocytes is undesirable, including, but not limited to, leukemia, lymphoma, lupus and rheumatoid arthritis, transplant rejection, asthma and other allergic diseases, autoimmune diseases, etc.
One or more embodiments of the present invention demonstrate that TFE3 and TFEB are critical for T cell function and humoral immunity through their direct control of CD40L expression. This was demonstrated by expression of the TDN protein in mouse T cells in vivo or
in human T cells in culture, which blocked existing TFE3 and TFEB proteins expressed therein, and by de novo inhibition of TFEB protein synthesis via expression of an interfering RNA against TFEB in Tcfe3-/~ T cells. TDN expression in mouse T cells in vivo via the EμPμ transgene resulted in hyper-IgM syndrome caused by defective CD40L expression. As mentioned previously, like the induction of many effector and activation molecules by
T cell stimulation, such as IL-2 and TNF family members, CD40L induction is dependent on subunits of NFAT, a calcium-responsive transcription factor complex. Additional regulators include the AT-Hook ("AT-Hook" refers to the AT-rich sequences this type of transcription factor binds to and the shape of the protein itself) transcription factor AKNA and subunits of transcription factors AP-I, NF-kB and Egr-1, which are also shared by other TCR and/or co-stirnulator-responsive genes. However, unlike the aforementioned genes, Cd40lg is the only one identified that requires TFE3 or TFEB. That requirement may contribute to the unique expression profile of Cd40lg compared with that of other TCR-responsive genes. Identifying the conditions and pathways that control TFE3 and TFEB activity is therefore important in understanding the control of CD40L-deρendent immunity and immunopathology.
In activated CD4+ T cells, TFE3 and TFEB were mutually redundant in controlling Cd40lg transcription, as a combined deficiency in TFE3 and TFEB inhibited TCR-dependent CD40L induction in the primary T cell culture systems, whereas individual deficiencies had a minor or no effect. Thus, one or more embodiments of the present invention have demonstrated physiological functional redundancy between TFE3 and TFEB in vivo, which may be a more general property of these proteins in other cell types. A degree of functional redundancy has also been reported among NFAT components in activating Cd40lg transcription, as a combined deficiency of NFATcI and NFATc2 (individual members of the NFAT family, respectively) results in greater impairment in CD40L expression than does individual deficiency. However, it remains possible that, in certain conditions, TFE3 and TFEB each have a unique contribution to the activation of Cd40lg. For example, only TFEB protein was induced after TCR stimulation of primary cells, most likely through post-transcriptional mechanisms, and TFEB bound to the Cd40lg promoter independently of TFE3. There were also differences in the relative importance of a subset of E-boxes to Cd401g promoter activity in wild-type versus Tcfe3-/- (Tcfe3 refers to the gene encoding TFE3 T cells. Consequently, there may be physiological contexts in which TFEB or TFE3 may be more advantageous, such as, for example, for NFAT subunits in T cell subsets or in other cell types expressing CD40L.
The very low, rather than absent, CD40L expression in activated T cells lacking TFE3 and TFEB activity is consistent with the limited degree of germinal center formation in
TDN-transgenic mice, in contrast to that of mice with genetic Cd40tg deficiency, in which no germinal centers are found, We believe that the remaining CD40L expression was not due to incomplete mactivation of TFE3 and TFEB by the TDN protein, given that TDN protein expression inhibited the binding of endogenous TFE3 or TFEB to the Cd40lg promoter to basal binding amounts, as evaluated by electrophoretic mobility-shift assay and chromatin immunoprecipitation. It is, however, possible that other μE3 (also referred to as "MiT") site-binding transcription factors not in the MiT family may bind to the promoter in their absence. These may include upstream stimulating factor (USF-I) and c-Myc, which are from distinct and non-interactive helix-loop-helix families. USF-I binds to a promoter target site normally occupied by TFE3 in Tcfe3-/- but not wild-type fibroblasts. It has been suggested that c-Myc can also activate the Cd40lg promoter, but its effect is apparently indirect. Yet regardless of MiT site occupancy, we postulate that the absence of TFE3 and TFEB raises the activation threshold of the Cd40lg promoter by the remaining activators, such as NFAT and API . In that scenario, very large and sustained amounts of the other transcription activators, coupled with synergistic mechanisms such as mRNA and protein stabilization, could allow sufficient CD40L accumulation to overcome the TFE3 and TFEB deficiency in certain T cell clones highly activated by TCR, co-stimulators and interleukins.
The normal distribution of the main T cell populations in non-immunized TDN-transgenic mice suggested that other possible MiT target genes in T cells were not required for T cell development itself beyond the double-positive stage when the TDN protein was first detected. Those findings are consistent with the reported lack of developmental defects of MitfΛ'ιnAi (Mitf refers to the gene encoding the Mitf protein, and the Mi/Mi refers to the particular mutant allele of MiIf) T cells and indicate that TFE3 and TFEB are also not essential. Also unaffected were the expression of several TCR-responsive genes other than Cd40lg, and antibody responses to the thymus-independent antigen. Those data suggest that MiT deficiency caused a relatively restricted T cell defect, in contrast to deficiencies in NFAT and
NF-B subunits, which can radically affect T cell development, effector function or homeostasis.
Given those observations, we believe a deficit in CD40L expression is sufficient to account for the impaired T cell-dependent antibody responses noted in TDN-transgenic mice. Further, in some humans with X-linked hyper-IgM syndrome, the genetic defect leads to low rather than no or mutant CD40L expression. Nevertheless, one or more embodiments of the invention do not rule out the importance of other putative TFE3 and TFEB target genes in T helper-cell function not specifically addressed herein, or their importance in positive and negative
selection. Although TFE3 has been directly and indirectly linked to several cytokine signaling pathways that control cell growth and differentiation in non-lymphoid cells, including c-Kϊt (c-kit is a tyrosine kinase receptor that is mutated in malignancies and is activated by "steel factor," and is also referred to as the c-kit ligand. Another name for c-kit is CDl 17. C-kit is critical for the development of mast cells and melanocytes) and transforming growth factor-β, cell type-specific and gene-specific, regulation by each MiT protein is an established characteristic of this family. Pathway involvement established in one cell type cannot be directly extrapolated to other cell types and must be determined experimentally.
Many human diseases are associated with abnormal CD40L expression. For example, CD40L expression is often constitutively increased on T cells from patients with systemic lupus erthythematosus and rheumatoid arthritis. In systemic lupus erthythematosus, this occurs without abnormal expression of other markers of T cell activation. Transcriptional and post-transcriptional mechanisms that normally enhance CD40L expression are all thought to contribute to that phenomenon. It has been shown that the Ras GTPase-mitogen-activated protein kinase (MAPK) pathway is necessary for the maintenance of abnormal CD40L expression by T cells from patients with systemic lupus erthythematosus. Given that in non-lymphoid cells, the transcriptional activity of TFE3 and TFEB can each be regulated by the Ras-mitogen-activated protein kinase (-MAPK) pathway, for example, in response to activation of the m-csf receptor, a key issue will be whether the activity of TFE3 and TFEB can be regulated by MAPK activation in T cells (for example, in response to the TCR and IL-15 receptor) and if there are differences in their activity in normal versus autoimmune T cells. MAPK activation of TFE3 and TFEB can be defined, at least in part, as phosphorylation which enables TEF3 and TEFB to interact with other molecules critical for transcription. FIG. Ia depicts an illustrative imrmmoblot of TFE3 and TFEB protein in un-stiimilated and stimulated primary CD4+ mouse splenocytes (via CD3 mAb) and human Jurkat T cells (stimulated via phorbol myristate acetate [PMA] plus ionomycin), according to an embodiment of the present invention. The GAPDH immunoblot is also shown as a loading control.
FIG. Ib depicts an illustrative time course of TFE3 and TFEB protein expression after TCR stimulation, according to an embodiment of the present invention. CD4+ T cells were stimulated with the CD3 mAb and aliquots taken at each time point for analysis by immunoblot. These data are extensions of the data in FIG. Ia.
FIG. Ic depicts an illustrative analysis of Mi tf protein expression, according to an embodiment of the present invention. Extracts from resting and LPS stimulated CD 19+ B cells,
resting and CD3-stimulated (8 hours) CD4+ T cells, un-stimυlated and pharmacologically stimulated Jurkat T cells, and resting thymocytes, were probed by immunoblot with a Mitf mAb. The cross-reactive band corresponding in size to the A isoform of Mitf is indicated with the line extending from "Mitf." This designation is also based on the disappearance of this band in stimulated B cells. The GAPDH immunoblot is also shown as a loading control.
FIG. Id depicts an illustrative semi-quantitative reverse transcriptase (RT) dependent-polymerase chain reaction (PCR) to measure relative abundance of Tcfe3, Tcfeb, (the mouse genes encoding TFE3 and TFEB proteins, respectively) and Mitf mRNA transcripts in resting and stimulated CD4+T cells over a time course, according to an embodiment of the present invention. Reverse transcriptase, in this context, is required to convert the mRNA into a DNA copy (cDNA) that is then used as a template for amplification in the PCR reaction. The identities of the bands are indicated. The titrations illustrate the relative band intensities of PCRs programmed with the same amount of 0 hour complementary DNA (cDNA) used for the PCR series (1/1), one-third the amount (1/3), or three-fold more (3/1). Immunoblot (FIGS. Ia, Ib, and Ic) and RT-PCR (FIG. Id) analyses were used to assess the MiT protein and mRNA expression profile, respectively, in T cells. Of the four MiT proteins, only TFE3 and TFEB proteins were detected in CD4+ splenic mouse T cells and in the human transformed T cell line Jurkat, as shown in FIG. Ia5 which depicts an exemplary immunoblot of TFE3 and TFEB in primary CD4+ mouse splenocytes and Jurkat T cells. With reference to FIGS. Ia3 Ib, Ic, and Id, by way of example only, CD4+ splenic T cells were stimulated by incubation with an anti-CD3 mAb for 8 hrs. Jurkat T cells were stimulated with phorbol myristate acetate (PMA)/ionomycin (Iono) for 20 hours. Glyseraldehyde-3-ρhosphate dehydrogenase (GAPDH) protein was the loading control. The TFEB and GAPDH antibodies recognized epitopes on the corresponding proteins from mouse and human and were used for probing both extracts. However, TFE3 proteins from mouse and human extracts were revealed with species restricted anti-TFE3 antibodies. Consequently, band intensities cannot be directly compared between mouse and human. The exemplary images shown in FIGS. la-Id are representative of at least three independent experiments.
Notably, TFEB protein levels increased (five-fold) in response to TCR-engagement in parallel with CD40L induction in primary mouse T cells, whereas TFEB levels were already elevated in un-stimulated Jurkat cells and did not change in response to stimulation. In contrast, TFE3 levels remained relatively constant in both cases, as apparent from FIG. Ia, The increase in TFEB was at the post-transcriptional level, as relative amounts of steady-state Tcfeb (the mouse gene encoding TFEB protein) mRNA were unchanged (FIG. Id). A band was detected
that was interpreted to be the A isoform of Mitf in un-stimulated CD4+ T cells, but we did not detect it in TCR-stimulated cells (FIG. Ic). This corresponds to a decrease in Miff mRNA (FIG. Id).
We did not detect TFEC expression in any of these samples by either immunoblot or RT-PCR (data not shown). In the human transformed T cell line Jurkat, both TFE3 and TFEB proteins were present and their abundance did not change in response to pharmacological stimulation (FIG. Ia). We did not detect Mitf or TFEC in these cells (FIG. Ic; TFEC data not shown). Thus, in TCR-activated (via CD3 mAb) mouse CD4+ T cells and pharmacologically activated (via PMA/iono) Jurkat T cells, TFE3 and TFEB were the only MiT family members expressed.
One or more embodiments of the present invention illustrate that T-celϊ-specific inactivation of endogenous TFE3 and TFEB in vivo via transgenesis results in a phenotype resembling X-linked hyper-IgM syndrome*
FIG. Ie is an illustrative schematic view depicting an EμPμ-TDN ("Eμ" and "Pμ" refer to the immunoglobulin μ heavy chain intronic enhancer and promoter, respectively) transgene vector used to create the TDN-transgenic (Tg) mice, according to an embodiment of the present invention. The construction of the TDN protein has been described (see, for example, Huan, C, Sashital, D., Hailemariam, T., Kelly, M.L. & Roman, C.A.J., Renal carcinoma associated transcription factors TFE3 and TFEB are leukemia-inhibitory factor-responsive transcription activators of E-cadherin," J Biol Chem 13, p. 13 (2005), which is incorporated by reference herein). It contains the HLH-Z region from TFE3 but lacks the DNA binding BR and transcriptional activation domains. A SV40-nuclear localization signal was added to compensate for the loss of that activity by deletion of the BR, and a HA-tag was added for immunodetection.
FIG. If depicts an illustrative immunoblot of extracts from total bone marrow (BM), total spleen (Spleen) and total thymocytes (Thymus) from TDN-transgenic mice (+) and non-transgenic littermates (-), according to an embodiment of the present invention. Non-transgenic mice are control littermates that did not receive the chromosome containing the transgene from the transgenic parent. FIG. If depicts, in part, TDN expression in total extracts of HEKL293 cells transfected with a plasmid expressing the TDN cDNA (lane labeled HEK TDN) for comparison. The anti-HA antibody detects the cognate hemagglutinin (HA) epitope built into the TDN protein. FIG. 1 f shows that the TDN protein is only detected in lymphoid organs that contain T cells from TDN-Tg mice and not in non-Tg mice, which established, in part, that TDN expression in this transgenic line was T cell specific.
FIG. Ig depicts illustrative flow cytometry histograms of permeabilized lymphocytes from non-Tg and T cell-specific TDN-Tg mice for staining with an anti-HA antibody that detects the TDN protein, according to an embodiment of the present invention. In the exemplary histograms in FIG. Ig, filled curves are HA staining profiles of non-Tg (control) cells, and line overlays are profiles of cells from TDN-Tg mice from bone marrow, spleen, and thymocytes. B and T lymphocyte populations are delineated using lineage-specific markers.
FIG. Ih depicts an illustrative immunoblot of extracts of purified splenic B cells and CD4+ T cells from TDN-transgenic mice (-), showing that the TDN protein is expressed only by splenic T cells but not in B cells from TDN-Tg mice, according to an embodiment of the present invention. FIG. Ih includes, in part, an immunoblot for the hemagglutinin (HA) epitope built into the TDN protein, and GAPDH, a loading control. Data are representative of at least three independent experiments.
FIGS. 2a and 2b are illustrative graphical views depicting how the numbers and a distribution of major B and T cell populations is normal in TDN-transgenic mice, according to an embodiment of the present invention.
TDN-transgenic mice exhibited humoral immune defects that were consistent with a hyper IgM syndrome caused by a deficit in CD40L expression. An EμPμ transgene, as described, for example, in Tepper, R.I. et al, "IL-4 induces allergic-like inflammatory disease and alters T cell development in transgenic mice," Cell 62, pp.457-467 (1990), which is incorporated by reference herein, was used to direct expression of the TDN protein. In one or more embodiments of the present invention, transgenic mouse lines were analyzed in which the TDN was expressed exclusively in T cells, which was shown in FIGS. If-Ih. TDN expression was found to be highest in thymocytes and lower in mature single positive splenic T cells. No TDN protein was detected in B cells or noτi-lymphoid cells and tissues (see, for example, FIGS. If-Ih). Analysis of lymphoid compartments in TDN-transgenic mice by cell counting and flow cytometry revealed that B and T cell development proceeded, with normal to nearly normal numbers and distributions of major splenic B and T cell populations (Spleen) and thymocytes (Thymus) at 5 weeks of age. This is shown in FIGS. 2a and 2b. Li hyper-IgM syndrome due to CD40L deficiency, antigen-independent lymphocyte development is normal. Rather, the hallmark phenotype includes defects in germinal center (GC) formation and antibody responses to T-dependent antigens. Therefore, the ability to form GCs was determined in transgenic and control mice intraperitoneally challenged with sheep red blood cells (SRBC).
FIGS. 3a and 3b are illustrative graphical views depicting impaired germinal center formation in TDN-transgenic mice immunized with SRBCs, according to an embodiment of the present invention.
Comparison of spleen sections on day 8-9 post inoculation revealed that GC formation was substantially reduced in TDN-transgenic mice compared to controls, with many of the detectable GCs smaller and poorly formed, an example of which is depicted in FIG. 3a (PNA and PNA plus B220 stains; PNA (peanut agglutinin) and B220 (also referred to as B cell antigen of 220 kiloDalton molecular weight) identify GC B cells). This was confirmed in a more quantitative way, for example, by flow cytometry, which showed an approximate 5 -fold reduction of CD 19-+ PNA+ splenic GC B cells compared to controls, as depicted in FIG. 3b. In contrast, FIG. 3a (PNA plus CD4 stains, the latter which identifies CD4 T cells) illustrates that TDN-transgenic T cells are present in the normal, expected locations in the spleens of immunized mice, adjacent to B cells. This is another property of T cells that could have been affected and caused the immune deficiency, but FIGS. 3a and 3b illustrate that it was not such a cause. That is, if T cells cannot make it to the proper place in the spleen, they cannot interact with B cells even if they had expressed CD40L. FIGS. 3a and 3b depict that T cells can make it to the proper place in the spleen, thereby illustrating that this property is normal and therefore does not account for the GC defect.
FIGS. 4a through 4d are illustrative graphical views of ELISAs depicting impaired T-dependent, but normal T-independent, humoral (antibody) responses in TDN-transgenic mice, according to an embodiment of the present invention.
Impaired GC formation is indicative of a lapse in Tπ-cell (helper T cells, primarily CD4+ T cells) dependent B cell activation and is predictive of impaired T-dependent antibody responses. Consistent with this, serum levels of total IgG and IgA isotypes in naive TDN-transgenic mice were reduced compared to controls, whereas total IgM was normal if not slightly elevated, as shown in FIG.4a. This prediction was then tested directly by evaluating the humoral responses of mice that were challenged with the T-independent and T-dependent antigens TNP-Ficoll and TNP-KLH, respectively. As shown in FIG. 4b, TNP-specifϊc antibody responses to TNP-Ficoll were comparable in TDN-transgenic and control mice. However, TDN-transgenic mice failed to mount a characteristic humoral response to the T-dependent antigen TNP-KLH compared to normal- as evidenced by dramatically reduced TNP -specific IgG antibodies at 14 and 21 days post immunization compared to controls, as illustrated in FIG. 4c. Similarly, per capita SRBC-specific plasma cell formation was also proportionally reduced, as shown in FIG. 4d. Thus, TFE3 and TFEB inactivation in T cells mediated by transgenic TDN
expression led to poor thymus dependent B cell responses to model antigens in a manner consistent with a CD40L deficiency.
Analysis of TDN-transgenic mice from two other independently generated T cell-specific lines also showed comparable T cell and B cell numbers and distribution but similarly impaired germinal center formation and generation of plasma cells in response to SRBC challenge compared with that of non-transgenic littermates. In these ways, T cell-specific TDN-transgenic mice showed the hallmark phenotypes of hyper-lgM syndrome, in which antigen-independent lymphocyte development and antibody responses to T cell-independent antigens are intact, but germinal center formation and IgG responses to T cell-dependent antigens are defective. The normal distribution and locations of major T cell populations in unimmunized TDN transgenic mice suggests that any other possible target genes of TFE3 and/or TFEB in T cells were not required for T cell development per se beyond the double positive (DP) stage when the TDN protein was first clearly detected (see, for example, FIGS. 2 and 3a). Also unaffected were antibody responses to the thymus independent antigen TNP-Ficoll (FIG. 4), which suggests that any T cell functions that influence this response were largely intact in the absence of TFE3 and TFEB activity. As detailed below, one or more embodiments of the invention illustrate that defective CD40L expression by TDN-transgenic T cells underlies the humoral immune defect in TDN-transgenic mice.
FIG. 5a depicts an illustrative flow cytometry of surface expression of CD40L by control (wild-type (WT)) and TDN-transgenic (TDN) CD4+ splenocytes stimulated, by way of example, with the anti-CD3 mAb and analyzed by flow cytometry for CD40L marker expression, according to an embodiment of the present invention. Filled curves represent CD40L staining of un-stimulated cells, and grey lines represent CD3 -stimulated cells. Numbers above bracketed lines indicate CD40L+ cells. Data are representative of more than three experiments. FIG. 5a depicts how surface expression of CD40L is impaired in TDN-transgenic T cells.
The expression of CD40L was much lower on the surface of TDN-transgenic CD4+ T cells than that of non-transgenic (WT) cells after TCR stimulation. Representative data is of T cells from one TDN-Tg line of mice, but is similar in the three lines studied.
However, the induction of other molecules including inducible co-stimulator (ICOS), CD25 CD28, and CD69 was indistinguishable between them, as shown, for example, in the following FIGS. 5b-d (and data not shown).
FIG. 5b depicts an illustrative quantification in bar graph form of histogram data as in FIG. 5a for percent CD4+ T cells positive for CD40L (left) and CD25 (right) after S hours of stimulation with various amounts of mAb to CD3 (Anti-CD3), according to an embodiment of the
invention. In FTG. 5b, n = 6 mice for total CD40L analyses, and n = 3 mice for analysis of both
CD40L and CD25 in the same experiment. The error bars depicted in the bar graph show the s.e.m. (standard error of the mean) for those values.
FIGS. 5c and 5d depict illustrative flow cytometry histograms of expressions of ICOS and CD28, respectively, showing that their expression is unaffected in TDN-Tg T cells compared to non-Tg (WT)5 according to an embodiment of the present invention. Tn FIGS. 5c and 5d, filled curves represent the relative intensity of ICOS or CD28 on freshly isolated CD4+ T cells. Green lines depicted in FIG. 5d represent the intensity on cells left un-stimulated in culture for 2 days.
Grey lines depicted in FIG. 5c and grey lines depicted in FIG. 5d represent the intensity on cells stimulated for 2 days with mAb to CD3. Data are representative of three separate experiments. FIG. 5e depicts an illustrative graphical representation of ELISA measurements of
IL-4 secretion, according to an embodiment of the present invention.
As shown in FIG. 5e, in vitro IL-4 secretion by TDN-transgenic T cells is normal. CD4+
T cells from control and transgenic mice were cultured with plate-bound anti-CD3 mAb for 2 days, followed by IL-2 for 7 days, and then re-stimulated with anti-CD3 without IL-2 for 2 days.
Data for n = 3 independent experiments, and standard error bars are shown.
FIG. 5f depicts an illustrative bar graph showing that the proliferative response of TDN-Tg
CD4 T cells in vitro to mitogenic stimulation by CD3/CD28 beads is indistinguishable from control non-Tg CD4 T cells. Purified T cells were placed in culture, CD3/CD28 beads added, and cell counts taken over a time course. Data for n = 3 independent experiments, and standard error bars are shown. We show that T cell proliferation in response to CD3 and CD28 co-stimulation is indistinguishable between non-Tg and TDN-Tg T cells. These proliferation data support one of the major the assertions depicted in FIGS. 2, 3a and 5, namely, that TDN-Tg CD4 T cells (that is,
T cells that lack TFE3/TFEB function) are relatively normal. FIG. 5g depicts an illustrative RT-PCR showing that Srnad7 expression is unaffected in
TDN-Tg and Tφ3-/~ splenic CD4 T cells. Smad7 is analyzed here because it is regulated by TFE3 and TFEB in non-hematopoietic cells.
In sum, FIG. 5 of the present invention teaches us that inactivation of TFE3 and TFEB had no effect on TCR-dependent CD25, CD69, or CD28 expression, IL-4 secretion, IL-2 responsiveness or CD3/CD28 responsiveness of CD4+ T cells, or Smad7 expression. The data therefore suggest that the effect of TFE3 and/or TFEB inactivation was relatively restricted to
CD40L and did not globally affect T cell responses to stimulation, in contrast to deficiencies in some NFAT and NFkB subunits.
One or more embodiments of the present invention illustrate that CD40L expression by CD4 T cells depends on endogenous TFE3 or TFEB. That is, they are mutually redundant activators of CD40L expression in CD4 T cells.
FIG. 5h depicts illustrative flow cytometry histograms showing CD40L and CD25 staining profiles of un-stimulated (filled curve) and TCR-stimulated (line overlay) green fluorescent protein (GFP+) cells from normal and TcfeS-/- mice (for three independent experiments), in which GFP shows cells that express either a control virus or a virus that expresses an inhibitory (interfering) stem-loop RNA that blocks TFEB synthesis, according to an embodiment of the present invention. Primary CD4+ splenic T cells were isolated from normal and Tcfe3-/- mice infected with the indicated stem-loop-interfering RNA/GFP lentivirus and incubated with IL-2 for about 4-6 days. Cells were then stimulated with plate-bound anti-CD3 mAb for about 8 hours and analyzed for CD40L or CD25 surface expression by flow cytometry.
Germline Tcfe3-/- mice have no reported defects in immune function, and the very early embryonic lethality caused by germline TFEB-deficicncy has precluded the study of Tcfeb-/- T cells. Therefore, the remaining models consistent with the biochemical, promoter, and genetic data prior to the experiment in FIG.5h were that TFEB itself is important for CD40L expression, or there exists a functional redundancy between TFE3 and TFEB with respect to CD40L expression. However, the TDN, by blocking both TFE3 and TFEB, does not allow discrimination between these models. To address this issue, in FTG. 5h CD40L expression was evaluated in primary splenic CD4+ T cells from wild-type (WT) and Tcfe3-/- mice rendered TFEB-deficient by repressing endogenous TFEB expression via retroviral introduction of a TFEB-specific stem-loop interfering RNA (slRNA).
FIG. 5i depicts an illustrative representation of an immunoblot confirming down-regulation of TFEB in CD4+ T cells infected with a retrovirus that expresses the inhibitory stem-loop TFEB RNA as in FIG. 5h, according to an embodiment of the present invention. FIG. 5i depicts an immunoblot of extracts from sorted GFP+ cells infected with a control (left) or TFEB-slRNA lentivirus, for TFEB and GAPDH. The immunoblot is representative of two independent experiments.
With continued reference to FIG. 5h, induction of CD40L by TCR-stimulation with an anti-CD3 mAb was comparable between wt T cells infected with the control or TFEB slRNA lentiviruses and Tcfe3-/- T cells infected with the control lentivirus, suggesting TFEB itself was not critical if TFE3 was present. In contrast, CD40L induction on Tcfe3-/- T cells infected with the TFEB slRNA vector was significantly impaired compared to control infected and wt cells. Induction of CD25 was unaffected by the TFEB slRNA in all cases. In addition, growth of wt
and TcfeS-/- T cells expressing the TFEB slRNA was indistinguishable from control-infected cells. These results support the model that TFE3 and TFEB have important and mutually redundant roles in the TCR-dependent induction of CD40L by primary mouse splenic T cells. Data presented herein excludes the possibility that impaired CD40L expression was a secondary 5 consequence of MiT-defϊcient T cell development or persistent TDN protein expression.
These one or more embodiments of the present invention therefore represent the first known demonstration of a physiological functional redundancy between TFE3 and TFEB and the first known demonstration of their role in controlling T cell function via their joint regulation of CD40L (see, for example, FIGS. 5, 7, 8, and 9). It remains possible that under certain conditions o each has a unique contribution to the activation of CD40L based on observations that only TFEB was induced after TCR stimulation of primary cells (FIG. Ia and Ib) and that TFEB could bind independently from TFE3 to the CD40L promoter (FIG. 7).
The present invention teaches us that TFE3 and TFEB control the expression of CD40L by directly controlling transcription of the gene encoding CD40L (Cd40lg and 5 CD40LG).
FIG. 5j depicts an illustrative real-time RT-PCR analysis of RNA isolated from CD4+ T cells from wild-type or TDN-transgenic littermate mice at various times after incubation with mAb to CD3 (horizontal axis), according to an embodiment of the invention.
In FIG. 5j, the illustrative bar graph shows the relative amounts of mRNA encoding 0 CD40lg from CD4+ T cells from wild type (wt) (black bars) or TDN-transgenic littermate mice (white bars) at the indicated times after incubation with the anti-CD3 mAb. By way of example only, and without loss of generality, real-time RT-PCR analysis of CD40lg RNA isolated from TDN-transgenic and control CD4+ splenic T cells at various times after TCR-stimulation showed a reduction of Cd40lg transcripts in TDN-Tg T cells at the induction peak to one-third that of 5 non-Tg (control) cells, with unchanged overall kinetics (FIG. 5j). These data substantiate the model that TFEB and TFE3 directly activate CD40lg at the transcriptional level in vivo. Data from n = 3 independent experiments and standard error bars are shown. Similar results were obtained with T cells from other independently generated, T cell-specific, TDN-transgenic lines.
In sum, one or more embodiments of the present invention teach a fundamental 0 relationship between the transcription factors TFE3 and TFEB and T cell function via their direct regulation of CD40L expression. The data are most consistent with a mutually redundant role of these molecules with respect to controlling CD40L transcription, as a combined TFE3/TFEB deficiency inhibited TCR-dependent CD40L induction in the primary T cell culture systems, whereas individual deficiencies had either a minor or no measurable effect (see, for example, FIG.
5h). A degree of functional redundancy has also been observed with NFAT components, as a combined deficiency of NFATcI and NFATc2 resulted in the greatest impairment in CD40L expression compared to individual deficiencies. The transgenic approach to inactivate endogenous TFE3 and TFEB via T cell specific expression of the TDN protein was therefore a key and advantageous strategy to address the redundancy and also served to circumvent the embryonic lethality of germline TFEB deficiency by directing expression to somatic cells.
One or more embodiments of the present invention teaches us that B cells from the T-cell specific TDN-Tg mice (FIGS. 1 e-lh) are not themselves defective and are capable of responding normally to antigen and CD40 signaling (FIG. 6). Consistent with that, CD 19+ B cells from TDN-transgenic mice and non-transgenic mice had similar expression of CD40 and responded similarly to in vitro stimulation, including the CD40-dependent induction of CD86 and major histo-compatibility complex (MHC) class II expression. Moreover, an agonist monoclonal antibody (mAb) to CD40, administered during immunization with TNP-KLH, enhanced day-7 TNP-specific IgG titers of both non-transgenic and TDN-transgenic mice, but most notably rendered the IgG responses indistinguishable from each other. Ih contrast, IgG antibody titers in mice treated with the isotype-matched control mAb were lower, and TDN-transgenic mice also had lower IgG responses than those of non-transgenic mice. These data indicate that B cell responses to T cell help and other aspects of T cell help critical for this T cell-dependent antibody response were intact in the TDN-transgenic mice and supported the conclusion that the humoral immune defect in TDN-transgenic mice was T cell intrinsic and was due to defective CD40L expression.
FIG. 6a depicts an illustrative flow cytometry histogram showing equivalent CD40 expression in B cells from non-transgenic and TDN-transgenic mice, according to an embodiment of the present invention. Filled curves represent unstained cells, and grey lines represent cells stained with mAb to CD40.
FIGS. 6b and 6c depict illustrative flow cytometry histograms showing that expressions of CD86 and MHC class II respectively, by B cells from TDN-Tg and non-Tg mice are equivalent, after stimulation of B cells with mAb to CD40, according to an embodiment of the present invention. In FIGS. 6b and 6c, filled curves represent CD86 or MHC class II staining on un-stimulated cells, and grey lines on stimulated cells.
FTG. 6d depicts illustrative ELISA analyses measuring IgG responses to TNP-KLH in non-transgenic and TDN-transgenic mice treated with an agonist mAb to CD40 during immunization, according to an embodiment of the present invention. TNP-KLH is an experimental antigen to which an antibody response requires T-cell derived CD40L to B cells.
Control mice (filled circles) and TDN-transgenic mice (open circles) were immunized intraperitoneally once with 100 micrograms of TNP-KLH plus 100 micrograms of either isotype-matched control antibody (control) or mAb to CD40 (3/23), delivered intravenously. The same amount of each mAb was administered once a day over 7 days. Data represent TNP-specific IgG isotypes from serum samples obtained on days 0 and 7.
One or more embodiments of the present invention illustrate that TFE3 and TFEB bind to and activate the CD40L promoter.
FIG. 7a depicts an illustrative schematic alignment of CD40L promoters from humans, mice and rats, showing where TFE3 and TFEB bind to and activate the CD40L promoter, according to an embodiment of the present invention.
The human, mouse, and rat CD40L promoters contain a number of E-box like sequences, a subset of which matches known optimal μE3-binding sites for MiT proteins and which are present in the CD40L promoter regions from each species. By way of example only, FIG. 7a depicts an illustrative schematic alignment of CD40L promoters from humans, mice and rats, showing where TFE3 and TFEB bind to and activate the CD40L promoter.
E-boxes are categorized as either ones that match known and optimal MiT transcription factor binding sites and that are present in all promoters (gray boxes with letters) or as non-conserved and non-optimal (dotted open boxes). The optimal MiT sites are further subcategorized based on the precise sequence of the site, as represented by letters A through E. Nucleotide positions of boxes are numbered, with A of the ATG translation start site equal to +1 , as will be understood by those skilled in the art.
FIG. 7b upper panel depicts an illustrative Chromatin immunoprecipitation (ChIP) of a Cd40lg promoter fragment by anti-TFE3 and anfi-TFEB antibodies, according to an embodiment of the present invention. The lower panel depicts illustrative controls for ChIP assays in the upper panel of FIG. 7b, according to an embodiment of the present invention. Titration of input DNA confirms amplification conditions in the ChIP assay are in the semi-quantitative, linear range. Input material (l/900th of total) from the upper panel of FIG. 7b was serially diluted as indicated above each lane and amplified in parallel with the experimental samples from FIG, 7b.
Chromatin immunoprecipitation (ChIP) assays were used to determine whether endogenous TFE3 and TFEB could bind to the CD40L promoter in vivo. ChIP analysis of un-stimulated and TCR-stimulated primary CD4+ splenic T cells from wild-type (wt) mice showed that TFE3 and TFEB each bound to a fragment of the murine Cd40lg promoter under both resting and stimulated conditions. FIG. Tb depicts exemplary Chromatin immunoprecipitation (ChIP) of a Cd40lg promoter fragment by anti-TFE3 and anti-TFEB antibodies. By way of example
only, primary CD4+ splenocytes from normal and Tcfe3-/- mice were un-stimulated or stimulated with anti-CD3, as indicated above each lane (for example, lanes 1-13) in FIG. 7b, upper panel. Shown is a semi-quantitative PCR analysis of starting material (Input, lanes 10-13) and immunoprecipitated material (ChIP, lanes 1-9) from the indicated sources and with the indicated Abs. ChIP using an anti-GAPDH antibody (lane 9) served as an additional negative control for background band intensity. The exemplary data shown in FIG. 7b is representative of three independent experiments.
Material detected in the anti-TFE3 ChIP sample from Tcfe3-/- cells defined background band intensity and was equivalent to the irrelevant antibody ChIP control (for example, compare lanes 3 and 9 in FIG. 7b, upper panel). Interestingly, whereas TFE3 binding did not change, TFEB binding appeared to increase after T cell activation by TCR cross-linking in a manner that mirrored in the increase of TFEB protein after TCR stimulation (see, for example, lanes 5 and 6 in FIG. 7b; FIG. Ia). The ability of TFEB to bind the fragment could be independent of TFE3, as TFEB recruitment to the Cd40lg promoter was unaffected in Tcfe3-/- cells (for example, lane 7 in FIG. 7b, upper panel). Neither TFE3 nor TFEB binding to the Cd40lg promoter was detected by ChIP in TDN-transgenic CD4+ splenic T cells, thus documenting an effective inactivation of endogenous TFE3 and TFEB DNA binding activity in those T cells by the transgenicaliy expressed TDN protein (see, for example, FIG. 7b upper panel, lanes 4 and 8). FIG. 7c (described below) also illustrates how TDN expression in T cells in vivo via transgenesis blocks TFE3 and TFEB binding to the CD40L promoter using a different methodology.
FIG. 7c depicts illustrative electrophoretic mobility-shift assays (EMSAs) to evaluate the binding of native TFE3 and TFEB proteins to the individual MiT sites in the Cd40lg promoter, according to an embodiment of the present invention. Nuclear extracts (NE) from stimulated wild-type and Tcfe3-/- splenic CD4+ T cells were incubated with radio-labeled oligonucleotides spanning a single MiT E-box (sites 1-8) and complexes were resolved by native gel electrophoresis. The top two panels show relative E-box-binding activity in extracts made from WT (left) or Tcfe3-/- (right) CD4 T cells. The markers on the left point out shifted complexes and free probe. The bottom EMSAs represent cold oligonucleotide competition and antibody- interference assays to determine the identity and specificity of E-box-binding complexes in cell extracts.
Each of the panels in the lower sets of EMSAs shows binding activity to one particular E-box probe from WT (left, first six lanes), TDN-Tg (left, last lane) or Tcfe3-/- (right panels) CD4 T cells. Excess unlabeled E-box oligonucleotides, with wild-type (WT) or with point mutations (Mut), and anti-TFE3 or anti-TFEB were included in some binding reactions (+; above lanes).
The exposures for the bottom panels were optimized for each probe. Data are representative of at least three independent experiments.
FIG. 7d depicts illustrative electrophoretic mobility shift assays (EMSAs) of nuclear extracts of human embryonic kidney (HEK293) cells transfected with control (-), TFE3, or TFEB pEBB (pEBB is a plasmid used by many labs to express genes in mammalian cells; the "E" in EBB stands for El alpha, the name of the mammalian gene from where the promoter was obtained) expression vectors showing TFE3 and TFEB binding to the MiT sites from the Cd40lg promoter, according to an embodiment of the present invention. As shown in FIG. 7c, EMSAs with nuclear extracts from primary CD4+ T cells established that native TFE3 and TFEB bound to the multiple MiT consensus E-boxes in the Cd40lg promoter, although to differing relative degrees and sometimes with preferential binding of one or both subunits to individual sites. Similar results were obtained here (FIG. 7d) by analyzing DNA-binding activities in extracts of HEK293 cells over-expressing TFE3 or TFEB. We demonstrated the specificity of the protein-DNA interactions by including TFE3- or TFEB-speciftc antibodies that interfered with DNA binding.
FIG. 8a depicts a bar graph plotting the relative activities of Cd40lg promoter fragments containing (full-length) or lacking (truncated) the MiT E-boxes and in the presence or absence of the TDN protein, as measured by (firefly) luciferase reporter gene assays, and shows that Cd40lg promoter activity depends on endogenous TFE3, TFEB and MiT boxes to enhance promoter activity, according to an embodiment of the present invention. Primary CD4+ splenic T cells from wild-type and TDN-transgenic mice were transfected with mouse Cd40lg promoter-luciferase constructs, either full-length (from the ATG to -1535 base pairs upstream) or truncated (to -382) lacking the MiT sites (rectangles). A separate plasmid that contains Renilla luciferase, molecularly distinct from firefly luciferase and whose expression was dependent on a promoter that does not require TFE3 and TFEB, was co-transfected to provide an internal transfection control for normalizing firefly luciferase values. Then, 2 days later, cells were stimulated for 8 hours with mAb to CD3, then were collected and lysed for luminometry. Data represent relative luciferase values (normalized to Renilla values). Standard error bars are shown, and in FIG. 8 a, n = three independent transfections using cells prepared from three separate sets of wild-type and TDN-transgenic mice.
FTG. 8b depicts illustrative contributions of individual and combinations of MiT E-box sites to promoter activity, according to an embodiment of the present invention. Point mutations abrogating binding of TFE3 and/or TFEB binding (1X1) were introduced into E-box sites (numbered 1-8; left margin) in the context of the full-length mouse Cd40lg promoter. Primary
splenic CD4+ T cells from wild-type and TcfeS-A mice were transfected, stimulated and analyzed for luciferase activity as in. described in FIG. 8a.
Point mutations that abrogated the binding of TFE3 and/or TFEB to individual sites also attenuated Cd40lg promoter activity in primary wild-type and TFE3-deficient CD4+ T cells (FIG. 8b), although to differing degrees. A full-length construct containing mutations of all eight sites had the greatest effect on promoter activity, reducing it to the activity of the truncated promoter. Individual mutation of sites three through seven had a less severe but measurably attenuating effects in both wild-type and TFE3 -deficient T cells. In contrast, mutation of site one or two had no effect in wild-type cells but had a substantial effect in TFE3-deficient T cells. That result was consistent with the preferential binding of TFEB from wild-type extracts to site one and the greater relative binding of TFEB to site two than to all other sites in the absence of TFE3.
The differential importance of some sites in TFE3 -deficiency versus wild-type cells may indicate that the TFE3 and TFEB redundancy was not complete or that sites had differential responsiveness to TFE3 or TFEB protein abundance. These results indicated that all sites in some context could act in concert for full TFE3- and/or TFEB-dependent enhancement of Cd40lg promoter activity. Thus, we infer that TFE3 or TFEB binding is critical for achieving the physiological regulation of CD40L expression necessary for T cell-dependent antibody responses.
FIG. 8c depicts that native TFE3 and TFEB can form heterodimers and that the TDN protein blocks this interaction, (Le., that heterodimerization, by way of example, is an interaction that is blocked by the TDN protein), according to an embodiment of the present invention, hi FIG. 8c, the graphical depiction on the left represents an immunoblot of stimulated CD4+ splenic T cell extracts from WT, Tcfe3-/- and TDN-Tg mice for TFE3, TFEB, TDN (HA), and GAPDH protein expression. Ih FIG. 8c, the graphical depiction on the right represents extracts from the same sources that were incubated with anti-TFE3, immune complexes that were precipitated, and immuno-precipitates resolved on SDS-PAGE and blotted with anti-TFEB. Ja wild-type CD4+ T cells, native TFE3 and TFEB were immuno-precipitated together (FIG. 8c lane 1), suggesting that, in addition to TFE3 homodimers and TFEB homodimers, that TFE3/TFEB heterodimers can form and may represent a distinct DNA binding species. That interaction was blocked in TDN-transgenic T cells (FIG 8c lane 3). These data confirmed functional inactivation of endogenous TFE3 and TFEB by the transgenically expressed TDN.
FIG. 9a is a bar graph depicting relative levels of Cd40!g and CD40LG promoter activity in human T cells (Jurkat) as represented by exemplary normalized luciferase values for each condition and promoter construct with respect to control renilla values for n = 3 and standard
error bars, according to an embodiment of the present invention. By way of example only, maximal Cd40lg and CD40LG promoter activity required an upstream fragment that contains conserved MiT boxes and endogenous TFE3 and/or TFEB.
To determine whether endogenous TFE3 and TFEB were important for CD40L promoter activity in human T cells, the firefly luciferase reporter gene assay was used. In this assay, murine Cd40lg and human CD40LG promoter fragments were linked to the luciferase gene and transiently transfected into the human T cell line Jurkat. Jurkat T cells were transfected either with full-length (-1.5 kB upstream: human -1562, mouse to -1535 relative to the ATG) or truncated (-382, mouse; -944, human) CD40L promoter-luciferase constructs with control or TDN-expressing plasmid and with Renilla as an internal control. About 2-4 hours later, cells were mock treated or treated with a combination of phorbol myristate acetate (PMA) and ionomycin (Iono), namely, PMA/iono, as above, then harvested 20 hours later for luciferase expression.
Any contribution of endogenous TFE3 and TFEB in Jurkat T cells to CD40L promoter activity was evaluated by comparing promoter activity in the presence or absence of the trans-dominant-negative (TDN) inhibitory protein, which can simultaneously inactivate both endogenous TFE3 and TFEB proteins. As mentioned previously, the TDN protein contains the TFE3 HLH/Zip (helix-loop-helix/leucine zipper) dimerization domains but lacks the DNA binding BR and transcription activation domains and thus forms heterodimers with MiT proteins that are incapable of binding DNA (FIG. 8c).
With continued reference to FIG. 9a, transient transfection of the reporters into Jurkat T cells showed that the activity of the full-length mouse Cd40lg and human CD40LG promoters exhibited about 3-fold higher activity at the induction peak following PMA/iono stimulation than the corresponding truncated promoters that lacked the consensus MiT binding sites. However, simultaneous expression of the TDN protein reduced the activity of the full-length promoters to the levels of the truncated promoter fragments, whereas the activities of the truncated promoters were unaffected by TDN expression. Thus, deletion of an upstream promoter fragment of the Cd40lg and CD40LG genes that contained the predicted MiT consensus sites rendered the promoters unresponsive to inhibition of endogenous TFE3 and TFEB by the TDN. The truncated promoters were still responsive to stimulation, which was attributed to the proximal NF AT/ AP-I site as has been reported (see, for example, Ix>bo, F.M., Xu, S., Lee, C. & Fuleihan, R.L, "Transcriptional activity of the distal CD40 ligand promoter." Biochem Biophys Res Commun, pp. 245-250, 279 (2000), Lindgren, H., Axcrona, K. & Leanderson, T., "Regulation of transcriptional activity of the murine CD40 ligand promoter in response to signals through TCR
and the co-stirnulatory molecules CD28 and CD2," J Immunol 166, pp. 4578-4585 (2001), and Parra, E., Mustelin, T., Dohlsten, M. & Mercola, D., "Identification of a CD28 response element in the CD40 ligand promoter," ./ Immunol 166, pp. 2437-2443 (2001), the disclosures of which are incorporated by reference herein). Therefore, the TDN did not block the activity of these other transcription factors, and established that endogenous TFE3 and/or TFEB were important for maximal induction of CD40LG and Cd40lg promoter activity in human T cells via MiT site-containing sequences.
FIG. 9b is an illustrative graphical view depicting how inhibition of native TFE3 and TFEB activity by expression of trans-dominant negative protein (TDN) blocks endogenous CD40L induction after activation of Jurkat T cells, according to an embodiment of the present invention.
In accordance with an embodiment of the invention, FIG. 9b illustrates that inhibition of TFE3 and TFEB activity by expression of the TDN via the pEBB plasmid blocks CD40L induction after activation of human Jurkat T cells. By way of example only, T cells were transiently transfected with the indicated pEBB-GFP-expression construct (control or TDN-expressing) and two days later stimulated with PMA/iono. About 12 hours later, cells were analyzed, for example, by flow cytometry, or alternative analysis methods, for surface expression of CD40L (left histogram) and CD25 (right histogram). In both histograms, shown are exemplary CD40L and CD25 expression profiles of GFP+ (plasmid transfected) cells that were un-stimulated (filled profiles), cells that received the control plasmid and were stimulated (red line-overlays), and cells that received the TDN plasmid (green line overlay). Data shown is representative of at least three independent experiments.
TDN protein expression selectively inhibited the induction of endogenous CD40L by Jurkat T cells dependent on stimulation by phorbol myristate acetate (PMA) plus ionomycin (iono), whereas the induction of CD25 was unaffected (FIG. 9b). Therefore, one or more embodiments of the present invention teaches us that endogenous TFE3 and/or TFEB are important for maximum induction of Cd40lg and CD40LG promoter activity and endogenous gene expression in mouse and human T cells and that they have evolutionarily conserved functions in that capacity (FIGS. 9a and 9b). In other words, what was defined in mouse CD4 T cells is also operational in human T cells (even though they are transformed).
FIG. 10a depicts a table illustrating oligonucleotides for EMSA in FIG. 7, according to an embodiment of the present invention. The oligonucleotides are very short sequences of DNA that are the same MiT DNA sequences as they exist in the Cd40lg promoter. This is why and how we used them to show that TFE3 and TFEB proteins interact with the MiT sites in the Cd40lg
promoter. FlG. 10a is merely an exemplary embodiment, and one or more embodiments of the present invention are not limited to the MiT sites used therein. One or more embodiments of the present invention any implement other MiT sites to bind to TFE3 and TFEB that do not come from the Cd40lg promoter. FIG. 1 Ob depicts an exemplary table illustrating oligonucleotides for cloning Cd40lg and
CD40LG promoters, according to an embodiment of the present invention.
FIG. 10c depicts an exemplary table illustrating oligonucleotides for E-box mutagenesis for the analyses in FIG. 8b, according to an embodiment of the present invention.
FIGS. 1 Ia-I Ic show that TDN expression in the B cell lineage does not block B cell development.
Furthermore, one or more embodiments of the invention include experiments that are from different lines of transgenic mice in which the TDN inhibitor is expressed in B cells, which make antibodies, and T cells in these mice are normal and do not express the TDN inhibitor. These lines are called "B-cell-specifϊc TDN-Tg mice." FIG. 11a depicts a representative immunoblot showing that TFE3 and TFEB proteins are expressed in B cells, according to an embodiment of the present invention.
FIG. 1 Ib depicts representative immunoblots probing for TDN expression in lymphoid tissues from a different line of EμPμ transgenic mice that are shown here to express the TDN in B cells but not T cells (so called "B cell-specific TDN-Tg mice"), according to an embodiment of the present invention.
FTG. 11c depicts representative flow cytometry, also showing that these mice express the TDN protein only in B, but not T cells, according to an embodiment of the present invention.
FIG. 1 Id depicts representative flow cytometry analyses showing that major B and T cell populations in these "B cell-specific TDN-Tg" mice are normal, according to an embodiment of the present invention.
Lines of EμPμ-TDN transgenic mice were identified that expressed the TDN exclusively in B cells, not T cells. The immunoblot and flow cytometry shown in FIG. 1 Ib and 1 Ic is proof of this principle showing that the TDN inhibitor is made only in B cells in these lines, and that TFE3 and TFEB proteins are expressed in B cells. The flow cytometry analysis of lymphocytes in these mice shows that, remarkably, B cell development is normal (FIG. 11 c). This is important because it suggests that systemic administration of a hypothetical TFE3 and/or TFEB inhibitor will not effect the production of B cells.
FIG. 12 is a flow chart illustrating a technique for treating at least one of an immunological disease and an inflammatory disease in a patient, according to an embodiment of
the present invention. In a preferred embodiment of the present invention, the technique for treating an immune disease in a patient includes step 1202, suppressing at least one of TFE3 and TFEB in a patient to thereby suppress CD40L expression in the patient.
Many human diseases are associated with abnormal CD40L expression (see, for example, Danese, S, & Fioccbi, C, "Platelet activation and the CD40/CD40 ligand pathway, mechanisms and implications for human disease," Crit Rev Immunol 25, pp. 103-121 (2005), and Schattner, EJ., "CD40 ligand in CLL pathogenesis and therapy," LeukLymphomα 37, pp. 461-472 (2000), which are incorporated by reference herein). For example, CD40L expression is typically elevated on T cells from SLE and RA patients. Inappropriate or undesirable expression of CD40L by T cells (or B cells and possibly other cell types) can cause autoantibody production by B cells and/or directly or indirectly (for example, via autoantibody complexes) trigger inflammatory responses mediated by the cellular immune system that destroy tissues and organs. CD40L is also necessary for B cells to become transformed by the Epstein Barr Virus. Interfering with CD40L-CD40 interactions - either genetically (in mice) or pharmacologically (for example, via CsA or interfering CD40L mAb) has had proven efficacy in mouse models and some efficacy in the clinic for ameliorating symptoms of and thus treating many autoimmune/inflammatory diseases.
One or more embodiments of the present invention teaches that TFE3 and TFEB control the expression of CD40L in CD4 T cells and thus provides a new way to control abnormal or undesirable CD40L expression in disease states. In addition, TFE3 and TFEB can each be regulated by the MAPK pathway, which can be activated by stimulation of multiple receptors including the TCR and the 1L-15 receptor, and IL-15 is elevated in RA. Thus, it is possible that the activity of TFE3 and TFEB can be regulated by MAPK activation with respect to CD40L expression by these and other receptors under normal and disease states. Moreover, given the broad distribution of TFE3 and TFEB expression in different cell types, CD40L expression in other cell types, including, but not limited to, B cells, monocytes and platelets, may similarly depend on TFE3 and TFEB and the effects of disease state on their expression, in these cells. For example, B cell abnormalities in some autoimmune diseases may be due to homotypic stimulation via co-expression of CD40 and CD40L on abnormal B cells.
In one or more embodiments of the present invention, the step of suppressing at least one of TFE3 and TFEB may include selectively inactivating TFE3 and/or TFEB, respectively.
Also, in one or more embodiments of the present invention, the step of suppressing at least one of TFE3 and TFEB may alternatively include the step of blocking an ability of at least one of TFE3 and TFEB to interact with DNA.
Furthermore, in one or more embodiments of the present invention, the step of suppressing at least one of TFE3 and TFEB includes the step of blocking an ability of at least one of TFE3 and TFEB synthesis. Suppressing TFE3 and/or TFEB may be achieved, in accordance with an aspect of the invention, by blocking TFE3 and/or TFEB synthesis, respectively, via an interfering RNA (which includes slRNA, as, by way of example only, in an embodiment of the invention). Blocking TFE3 and/or TFEB synthesis would leave other MiT proteins untouched and would be predicted to have the least invasive, least collateral off-target effects.
In one or more embodiments of the present invention, the step of suppressing at least one of TFE3 and TFEB includes at least one of blocking an interaction between TFE3 and TFEB, blocking TFE3 and TFEB from forming dimers, and blocking TFE3 and TFEB from binding with DNA. Blocking TFE3 and TFEB from forming dimers may comprise blocking TFE3 and TFEB from forming dimers with each other, blocking TFE3 from forming dimers with itself, and/or blocking TFEB from forming dimers with itself.
Also, in accordance with one or more embodiments of the present invention, the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient may include the use of the suppressing TDN protein. Blocking TFE3 and/or TFEB protein activity via a TDN-like molecule or oligonucleotide MiT binding site DNA binding blockers may also affect Mitf and TFEC. However, in one or more embodiments of the present invention, molecular inhibitors of this type may have great efficacy if used locally, for example, at the site of a transplant or in bead form and released locally. As described herein, TDN can experimentally block Mitf and TFEC as illustrated, for example, by the depiction herein that Mitf is expressed in thymocytes and un-stimulated T cells. Additionally, Mitf and TFEC may be capable of binding to the MiT sites in the CD40L gene promoters in other cell types that express these molecules. Ih one or more embodiments of the present invention the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient may include some type of interfering RNA (this includes the interfering stem-loop RNA, as was used here, and double stranded oligonucleotide RNAi).
In one or more embodiments of the present invention, the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient may include some type of agent that would block the activation of TFE3 and/or TFEB by MAPK phosphorylation or prevent interactions between TFE3 and TFEB and upstream or downstream effectors that depend on this phosphorylation.
In one or more embodiments of the present invention, tiie technique for treating at least one of an immunological disease and an inflammatory disease in a patient may comprise gene therapy, which may also benefit by including TFE3 and TFEB binding sites in the vector to promote appropriate expression of genes within the gene therapy vector in the patient. In one or more embodiments of the present invention, the immune disease includes at least one of an autoimmune disease and an inflammatory disease.
In one or more embodiments of the present invention, the technique for treating an immune disease in a patient may also include step 1204, administering at least one of a TFE3 inhibitor and a TFEB inhibitor for suppressing at least one of TFE3 and TFEB, respectively, in the patient.
In one or more embodiments of the present invention, at least one of the TFE3 inhibitor and the TFEB inhibitor is administered systemically. Also, in one or more embodiments of the present invention, at least one of the TFE3 inhibitor and the TFEB inhibitor is administered as an inhalant. Furthermore, in one or more embodiments of the present invention, at least one of the
TFE3 inhibitor and the TFEB inhibitor is administered subcutaneously. As way of example and not limitation, the TFE3 inhibitor and the TFEB inhibitor may be administered at site of inflammation and/or rejection.
Materials and Methods
Cell culture. CD4 T cell purification and T cell activation
By way of example only, CD4+ T cells were purified from spleens of 5 week old mice with a mouse CD4 negative isolation kit (Dynal Biotech) to greater than or equal to about 96% purity as determined by flow cytometry. T cells were cultured in RPMI supplemented with 10% (volume/volume) heat-inactivated FCS (fetal calf serum) (Gibco), and 50μM β-mercaptoethanol (Sigma). For the lentiviral stem loop RNAi experiment, 20 units/ml mouse IL-2 (Roche) was added to the culture medium.
For TCR stimulation, the purified mouse spleen CD4+ T cells - either cultured in IL-2 for 4-5 days (slRNAi) or freshly isolated - were incubated in 5-10 μg/ml anti-CD3 mAb (anti-CD3ε mAb 145-2C11, BD Pharmingen)-fixed plates for ~8 hr before harvesting for detection of surface CD40L and CD25 expression.
1 For CD28 expression, the cells were incubated with or without 1 Oμg/ml anti-CD3 for 4Shr after initiation of the culture. For ICOS expression analysis, T cells were incubated for 48 hours
with 10 μg/ml anti-CD3 following the manufacturer's instructions (BD Pharmingen). For the real-time PCR, freshly isolated and purified spleen CD4+ T cells were incubated in 10 μg/ml anti-CD3 fixed plates in RPMI/FBS/BME for 0 hour (hr), 3hr, 6hr, and 9hr before harvesting for RNA extraction and reverse transcription. Approximately 2xlO7 cells were used per time point. For stimulation of Jurkat cells, PMA (20 ng/ml final) and ionomycin (1.5 μM final) were added to the RPMI/FBS/BME culture medium for 12 or 24 hours before harvesting for fluorescent antibody staining and reporter gene assays. IL-4 cytokine analysis was done as described (24). Briefly, CD4+ T cells were removed from anti-CD3 stimulation after 48hr, cultured for 7 days in Roswell Park Memorial Institute medium (RPMI)/ Fetal bovine serum (FBSybeta-mercaptoethanol (BME) containing 100 units/ml mouse IL-2 (Roche), then re-stimulated at 1O5 cells/well in 96-welI plate bound with 5μg/ml anti-CD3 without IL-2 for 48 hours. Supernatants were collected for quantitative enzyme-linked immunosorbent assay (ELISA).
For B cell purification and B cell stimulation, splenic B cells were purified with a mouse B cell Negative Isolation kit (Dynal Biotech) such that 94% or more of the cells were CD 19+. Purified B cells were then analyzed directly for CD40 expression or were stimulated for 48 hours with mAb to CD40 (3/23; BD Pharmingen) and then were analyzed for CD86 and MHC class II surface expression by flow cytometry.
Generation and identification of transgenic mice
To create the TDN-transgenic mice, the 3' hemagglutin (HA) epitope-tagged TDN cDNA was subcloned into an immunoglobulin heavy chain gene enhancer and promoter based transgene cassette (EμPμ) that can direct expression in T cells, B cells or both, depending on the line. Fragments that contained the transgene cassette were excised from the plasmid backbone, purified and microinjected into fertilized FVB oocytes as described in the SUNY-Downstate Transgenic Facility (see, e.g., Hogan, B., Beddington, R., Costantini, F. & Lacy, E, "Manipulating the Mouse Embryo: A Laboratory Manual," Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1994, which is incoipoiated by reference herein).
Founder mice were identified by Southern blotting and PCR of genomic DNA from tails. Transgenic mice analyzed were the progeny of transgenic animals that had been back-crossed at least four generations onto the C57BL/6 strain. Five independent lines were derived that were confirmed to express TDN transgene only in T cells. Three lines were evaluated for recapitulation of impaired induction of CD40L in primary CD4+ T cells. All showed the same defect, and results from one line is shown. All transgenic mice used were 5-6 weeks old and hemizygous for the transgene, and
non-transgenic control mice were sex-matched Iittermates of transgenic mice. The mice were housed under SPF (specific pathogen free) conditions according to guidelines approved by the Division of Laboratory Animal Resources.
Lentivirus-delivered stem-loop KNAi
The control and TFEB slRNA lentiviruses are described in Huan et al. and viral particles produced according to published protocols (see, for example, Rubinson, D.A. et al., "A lenti virus-based system to functionally silence genes in primary mammalian cells, stem cells and transgenic mice by RNA interference;," Nat Genet 33, pp. 401-406 (2003), which is incorporated by reference herein). Briefly, the pLentiLox 3.7 plasmid inserted with TFEB mKNA targeting stem-loop RNAi was co-transfected into 293 cells with the packaging vectors pMDLg/pRRE ("p" refers to "plasmid," and the MDLg and RRE are conventional names for viral genes that encode a function necessary to build viral particles, so called "helper" functions), CMV-VSVG (CVM refers to transcription activation sequences from the cytomegalo virus that are built into plasmids to express other genes, in this case the G protein, from VSV; and VSVG is a name for viral genes that encode a function necessary to build viral particles) and RSV-Rev (RSV refers to transcription activation sequences from the Rous sarcoma viruses that are built into plasmids to express other genes, in this case the Rev protein from HIV; and Rev is a name for viral genes that encode a function necessary to build viral particles). The viruses in culture media were concentrated by centrifugation, and used for infecting purified mouse spleen CD4+ T cells. The infected T cells were cultured with 20 units/ml IL-2 for 4-5 days before evaluating infection efficiency and anti-CD3 stimulation.
Flow Cytometry
To detect cell surface markers, single cell suspensions were treated with Fc receptor block (2-4G2, BD Pharmingen), and were stained with anti-CD19, B220, TCRβ, CD40L, CD25, CD28, CD4, CD8, ICOS and/or CD86 conjugated to FITC5 PE, or Cy-Chrome (BD Pharmingen) or with fluorescein isothiocyanate-conjugated anti-MHC class II (Caltag).. For detection of the TDN protein in transgenic lymphocytes, cells were treated with Fix & Perm Kit (Caltag), before being stained with FITC conjugated anti-HA mAb (3F10, Roche). Cells were analyzed on a FACScan (A BD company name for a particular model of flow cytometer: Fluorescence Activated Cell Sorter/Scan) with CellQuest™ software (a trademark of Becton Dickenson).
Chromatin Irnmunoprecipitation assays f ChIP)
The ChIP assay was done as described (see, for example, Huan, C, Sashital, D., Hailemariam, T., Kelly, M.L. & Roman, C.A.J., "Renal carcinoma associated transcription factors TFE3 and TFEB are leukemia-inhibitory factor-responsive transcription activators of E-cadherin," J Biol Chem 13, p. 13 (2005)), with the following modifications: Purified 1.5 x 107 mouse spleen CD4+ T cells with or without anti-CD3 stimulation for 8 hr were used for each immunoprecipitation, with 5 μg of the following antibodies: anti-TFE3 mAb (G138-312, BD Pharmingen), anti-TFEB (ab2636, abeam), and anti-GAPDH (MAB374, Chemicon). Then, 10% of the precipitated DNA and 0.11% of the input DNA were used as templates for each PCR using the following conditions: 1 minute (min) at 94°C, 1 min at 610C, and 1.5 min at 72°C for 29 cycles. The PCR products were separated by 5% acrylamide gel electrophoresis. The primers used for amplifying mouse CD40 ligand promoter region are: forward, 5 CACAGACAGCATCCCTAGCA 3, and reverse 5 CTAAGCTGAG GCCAAACCAC 3.
Real-time Reverse transcriptase mediated (RT)-PCR
Total RNA from purified CD4+ splenic T cells was isolated with TRI reagent (TR-118, MAR) and contaminant DNA was removed by DNase I (AMP-Dl, Sigma) according to the manufacturer s instructions. Total RNA (5μg) was reverse-transcribed and PCR amplified with QuantiTect SYBR Green RT-PCR kit (Qϊagen) in an Opticon Continuous Fluorescence Detector (MJ Research). Detection of mouse CD40L and GAPDH inRNAs were determined by real-time RT-PCR using following primers, for CD40L, 5' AAAATGGG AAAC AGCTGACG 3', and 5' GGTATTTGCCGCCTTGAGTA 3\ for GAPDH, 5' TCACCACCATGGAGAAGGC 3% and 5' GCTAAGCAGTTGGTGGTGCA 3'. CD40L mRNA expression was normalized to GAPDH mRNA.
Enzvmc-Linked Immunosorbent Assay fELISA) and Immunizations
Total serum levels of IgM, IgGl, IgG2a, IgG2b, IgG3 and IgA were determined using mouse ELISA quantitation kits (Bethyl Lab). Bound Ig was detected using a tetramethylbenzidine (TMB) peroxidase substrate system (KPL). The T cell-independent type π and T cell-dependent immunizations and immune responses assays were done as described (see, for example, Yamazaki, T. et al., "Essential immunoregulatory role for BCAP in B cell development and function," J Exp Med 195, pp. 535-545 (2002), which is incorporated by reference herein).
In vivo stimulation of B cells with mAb to CD40 was done as described (see, for example, McAdam, AJ. et al. "ICOS is critical for CD40-mediated antibody class switching," Nature 409,
102-105 (2001), which is incorporated by reference herein) with modifications. Mice were immunized intraperitoneally with 100 μg TNP-KLH (Biosearch Technologies) in complete Freund's adjuvant (Difco) and were injected intravenously each day for seven days after immunization with 100 μg anti-CD40 (mAb 3/23; BD Pharmingen) or the isotype control rat IgG2a K (mAb R35-95; BD Pharmingen), and then blood was collected to obtain £erum. Serum Igs specific for TNP (Trinitrophenol) were measured in Immulux HB plates (DYNEX) coated with TNP-BSA (Trinitrophenol-bovine serum albumin conjugate) (Biosearch Technologies) with ELISA quantitation kits (Bethyl Lab).
ELISA was performed with EL405 auto plate washer (BIO-TEK), the plates were read at 450nm with μQuant microplate spectrophotometer (BIO-TEK), and data was analyzed with
KCjunior microplate data analysis software (BIO-TEK). For IL-4 cytokine analysis, T cells culture supematants were assayed for cytokine content by Mouse IL-4 Immunoassay Elisa kit
(R&D) following manufacturer's protocol.
Immunocvtochemical analysis of germinal centers
Mice were immunized intraperitoneally with 7.5 μl SRBC (Colorado Serum) in 200 μl PBS. The spleens were harvested S days after immunization and fixed in 10% formalin. Germinal centers were stained as previously described (see, for example, Cattoretti, G. et al., "BCL-6 protein is expressed in germinal-center B cells," Blood 86, pp. 45-53 (1995), which is incorporated by reference herein) with modification.
Briefly, antigen-retrieved slides were blocked with 3% pig serum and avidin/biotin blocking kit (Vector Lab), incubated with lμg/ml of biotin-conjugated PNA (Sigma) for more than 2 hr, and then incubated with biotin-conjugated goat anti-PNA (Vector Lab) for 45 min. Subsequently, the sections were treated with 0.1% NaN3 and 0.3% H2O2 for 30 min to block the endogenous peroxidase before staining with 1 :400 diluted HRP -conjugated avidin (Dako) for 20 min. After washing, the sections were incubated with HRP developing solution for 20 min, and then embedded with glycerol gelatin. All staining procedures were performed at room temperature.
Electrophoretic Mobility-Shift Assay Analysis
These assays were done as described below. Nuclear extracts from anti-CD3-activated CD4+ T cells or transfected HEK293 cells (5 μg) were incubated on ice for 30 minutes with a 32P-labeled oligonucleotide spanning an E-box site from the mouse Cd401g promoter in 25 mM HEPEs, pH 7.9, 50 mM KCl, 4% (weight/volume) Ficoll, 5 μM ZnCl2, 0.1 mM dithiothreitol,
0.02% (volume/volume) Nonidet-P40, 5 mM MgCl2, 10 μg/ml of BSA and 10 ng/ μl of poly(dI:dC) (deoxyinosinerdeoxycytidine). Nonspecific binding was assessed in the presence of a 100-fold excess of unlabeled oligonucleotide corresponding to the labeled oligonucleotide with or without E-box mutation and 2.5 μg anti-TFE3 (BD Pharmingen) or anti-TFEB (Santa Cruz). Samples with antibody were incubated for an additional 30 minutes. Reaction products were separated by native 5% (weight/volume) PAGE at 4°C and were visualized by autoradiography.
CD40L promoter reporter gene plasmid construction, transient transfection. and reporter gene assay The 5 extended ("full-length": to -1535 bp and -1562 bp, relative to the mouse and human ATG, respectively) and truncated mouse and human CD40L promoter fragments (to -382 bp and -944 bp from the ATG, respectively) were PCR amplified from genomic DNA and inserted into the pGL3 luciferase reporter vector (Promega). Mouse Cd40lg promoter reporter gene plasmids with E-box mutations (CANNTG to CTNNTG) were constructed by site-directed mutagenesis (Stratagene) with specific primers. Jurkat cells were transfected by electroporation with lOμg of CD40L promoter reporter gene plasmid, 0.05μg renilla control plasmid and 8μg TDN expressing plasmid for each sample, and subsequently activated with PMA and ionomycin as described (see, for example, Lobo, F.M., Xu, S., Lee, C. & Fuleihan, R.L., "Transcriptional activity of the distal CD40 ligand promoter," Biochem Biophys Res Commun 279, pp. 245-250 (2000), incorporated herein by reference). Firefly luciferase (Luc) and Renilla renifoπnis luciferase (Rlhic) activities were measured from cell extracts with the Dual Luciferase Reporter Assay System (Promega) and TD-20/20 Luminometer (Turner Designs). Primary mouse CD4 T cells were transfected with 2.5 μg of Cd401g promoter reporter gene plasmids, 0.01 μg of RLluc and 1.5 μg of empty pEBB or pEBB-TDN expression plasmid for each sample using the mouse T cell Nuclefector kit (Amaxa). Then, two days after transfection, the cells were activated for eight hours with 5 μg/ml of mAb to CD3ε and then were lysed for luminornetry as described herein. Luciferase activity was always normalized to Rlluc activity. Ia all experiments, the total amount of pEBB expression vector DNA was equalized by balancing cDNA-containing pEBB with empty pEBB,
Protein detection
Purified mouse CD4+ splenic T cells and Jurkat T cells after or without activation were lysed in detergent buffer (]50mM NaCl, 2OmM Tris pH7.4, 1% Triton-100, 0.1% SDS), supplemented with Complete Protease Inhibitor Tablets (Roche). Protein was transferred to PVDF membrane (Immobilon P). The following antibodies were used for western: anti-mouse
TFE3 (BD-Pharmήigen), anti-TFE3 (Santa Cruz Biotechnology), anti-TFEB (abeam), anti-GAPDH (CHEMICON), rabbit anti-goat IgG peroxidase conjugate (Sigma), and goat anti-mouse IgG peroxidase conjugate (Sigma).
The blots were developed by enhanced chemiluminescence (ECL). Coimmunoprecipitation was done according to a protocol from BD-Pharmingen. Purified CD4+ splenocytes (2 x 107) were lysed in imraunoprecipitation buffer (1% (weight/volume) Triton X-100,
150 mM NaCl, 10 mM Tris, pH 7.4, 1 mM EDTA, 1 mM EGTA, pH 8.0, 0.2 mM sodium orthovanadate, 0.5% (weight/volume) Igepal and protease inhibitor 'cocktail' (Roche)). Cell lysates were pre-cleared with protein G-agarose beads (Roche) and then were incubated overnight with 5 μg mAb to TFE3 (BD-Pharmingen). Immunoprecipitates were collected on protein G-agarose beads and were washed before elution in sample buffer for protein detection as described herein.
Plaque forming cells
The spleen IgM anti-SRBC response was assayed 5 days after intraperitoneal administration of 0.5-7.5 μl SRBC in 200 μl PBS following the protocol of the Jerne plaque assay (see, for example, Jerne, N.K. et al., "Plaque forming cells: methodology and theory," Transplant Rev 18, pp. 130-191 (1974), incorporated by reference herein).
While the present invention has been described in accordance with the treatment of immune diseases and disorders described herein, it is to be understood mat the teachings of the present invention are generally applicable to any diseases or disorders necessitating the regulation of CD40L expression. Thus, the teachings of the present invention should not be construed as being limited to the treatment of any particular disease or disorder.
Although illustrative embodiments of the present invention have been described herein, it is to be understood that the invention is not limited to those precise embodiments, and that various other changes and modifications may be made by one skilled in the art without departing from the scope or spirit of the invention.
3S
Claims
1. A method of treating at least one of an immunological disease and an inflammatory disease in a patient, the method comprising the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient.
2. The method of claim 1, wherein the step of suppressing at least one of TFE3 and TFEB comprises the step of selectively inactivating at least one of TFE3 and TFEB, respectively.
3. The method of claim 1, wherein the step of suppressing at least one of TFE3 and TFEB comprises the step of blocking an ability of at least one of TFE3 and TFEB to interact with DNA.
4. The method of claim 1, wherein the step of suppressing at least one of TFE3 and TFEB comprises the step of blocking an ability of at least one of TFE3 and TFEB synthesis.
5. The method of claim 1, wherein the step of suppressing at least one of TFE3 and TFEB comprises the step of blocking an interaction between TFE3 and TFEB.
6. The method of claim 1, wherein the step of suppressing at least one of TFE3 and TFEB comprises the step of blocking TFE3 and TFEB from forming dimers.
7. The method of claim 6, wherein the step of blocking TFE3 and TFEB from forming dimers comprises blocking TFE3 and TFEB from forming dimers with each other.
8. The method of claim 6, wherein the step of blocking TFE3 and TFEB from forming dimers comprises blocking TFE3 from forming dimers with itself and blocking TFEB from forming dimers with itself, respectively.
9. The method of claim 1, wherein the step of suppressing at least one of TFE3 and TFEB comprises the step of blocking TFE3 and TFEB from binding with DNA.
10. The method of claim 1, wherein the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient comprises inactivating at least one of TFE3 and TFEB via TDN protein.
11. The method of claim 1 , wherein the step of suppressing at least one of TFE3 and TFEB in the patient to thereby suppress CD40L expression in the patient comprises blocking at least one of TFE3 and TFEB synthesis via an interfering RNA.
12. The method of claim 1, further comprising the step of administering at least one of aTFE3 inhibitor and a TFEB inhibitor for suppressing at least one of TFE3 and TFEB, respectively, in the patient.
13. The method of claim 12, wherein at least one of the TFE3 inhibitor and the TFEB inhibitor is administered systemically.
14. The method of claim 12, wherein at least one of the TFE3 inhibitor and the TFEB inhibitor is administered as an inhalant.
15. The method of claim 12, wherein at least one of the TFE3 inhibitor and the TFEB inhibitor is administered subcutaneously.
16. The method of claim 15, wherein the TFE3 inhibitor and the TFEB inhibitor is administered at a site of at least one of inflammation and rejection.
17. The method of claim 1, wherein the immunological disease comprises an autoimmune disease.
18. The method of claim 1, wherein the step of suppressing at least one of TFE3 and TFEB comprises the step of blocking a transcriptional activity of at least one of TFE3 and TFEB, respectively.
19. The method of claim 18, wherein the step of blocking a transcriptional activity of at least one of TFE3 and TFEB comprises inhibiting phosphorylation of TFE3 and TFEB.
20. The method of claim 18, wherein the step of blocking a transcriptional activity of at least one of TFE3 and TFEB comprises preventing an interaction with at least one other molecule which controls transcription.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP06840267A EP1968386A4 (en) | 2005-12-15 | 2006-12-14 | Method for treating immune dysfunction by regulation of cd40 ligand expression |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US75061105P | 2005-12-15 | 2005-12-15 | |
US60/750,611 | 2005-12-15 | ||
US11/610,920 US20070178087A1 (en) | 2005-12-15 | 2006-12-14 | Method for Treating Immune Dysfunction by Regulation of CD40 Ligand Expression |
US11/610,920 | 2006-12-14 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2007070856A2 true WO2007070856A2 (en) | 2007-06-21 |
WO2007070856A3 WO2007070856A3 (en) | 2007-11-22 |
Family
ID=38163641
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2006/062111 WO2007070856A2 (en) | 2005-12-15 | 2006-12-14 | Method for treating immune dysfunction by regulation of cd40 ligand expression |
Country Status (3)
Country | Link |
---|---|
US (1) | US20070178087A1 (en) |
EP (1) | EP1968386A4 (en) |
WO (1) | WO2007070856A2 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2218458A1 (en) | 2009-02-13 | 2010-08-18 | Fondazione Telethon | Molecules able to modulate the expression of at least a gene involved in degradative pathways and uses thereof |
Families Citing this family (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN102549158B (en) * | 2009-05-22 | 2017-09-26 | 库尔纳公司 | By suppressing to treat the disease that TFE3 is related to IRS albumen 2 (IRS2) for transcription factor E3 (TFE3) natural antisense transcript |
JP6083735B2 (en) * | 2009-12-31 | 2017-02-22 | カッパーアールエヌエー,インコーポレイテッド | Treatment of insulin receptor substrate 2 (IRS2) related diseases by inhibition of natural antisense transcripts against insulin receptor substrate 2 (IRS2) and transcription factor 3 (TFE3) |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EA002549B1 (en) * | 1997-05-17 | 2002-06-27 | Байоджен, Инк. | Use of a cd40:cd154 binding interruptor to prevent counter adaptive immune responses, particularly graft rejection |
GB0006398D0 (en) * | 2000-03-16 | 2000-05-03 | Novartis Ag | Organic compounds |
US20030212025A1 (en) * | 2001-06-08 | 2003-11-13 | Yen-Ming Hsu | Cd154 variants |
-
2006
- 2006-12-14 US US11/610,920 patent/US20070178087A1/en not_active Abandoned
- 2006-12-14 EP EP06840267A patent/EP1968386A4/en not_active Withdrawn
- 2006-12-14 WO PCT/US2006/062111 patent/WO2007070856A2/en active Search and Examination
Non-Patent Citations (2)
Title |
---|
None |
See also references of EP1968386A2 |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2218458A1 (en) | 2009-02-13 | 2010-08-18 | Fondazione Telethon | Molecules able to modulate the expression of at least a gene involved in degradative pathways and uses thereof |
WO2010092112A1 (en) | 2009-02-13 | 2010-08-19 | Fondazione Telethon | Molecules able to modulate the expression of at least a gene involved in degradative pathways and uses thereof |
JP2012517802A (en) * | 2009-02-13 | 2012-08-09 | フォンダッツィオーネ・テレソン | Molecules capable of regulating the expression of at least one gene involved in the degradation pathway and uses thereof |
AU2010212854B2 (en) * | 2009-02-13 | 2013-10-03 | Fondazione Telethon | Molecules able to modulate the expression of at least a gene involved in degradative pathways and uses thereof |
Also Published As
Publication number | Publication date |
---|---|
US20070178087A1 (en) | 2007-08-02 |
EP1968386A4 (en) | 2009-07-22 |
EP1968386A2 (en) | 2008-09-17 |
WO2007070856A3 (en) | 2007-11-22 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11359020B2 (en) | Agents that modulate immune cell activation and methods of use thereof | |
Reboldi et al. | IgA production requires B cell interaction with subepithelial dendritic cells in Peyer’s patches | |
Huan et al. | Transcription factors TFE3 and TFEB are critical for CD40 ligand expression and thymus-dependent humoral immunity | |
AU2018228501B2 (en) | Therapies based on control of regulatory T cell stability and function via a Neuropilin-1:Semaphorin axis | |
Weber et al. | ICOS maintains the T follicular helper cell phenotype by down-regulating Krüppel-like factor 2 | |
US11219645B2 (en) | Tumor infiltrating lymphocytes for treatment of cancer | |
JP2016001196A (en) | Treatment and diagnostic methods for fibrosis related disorders | |
US10392431B2 (en) | Polynucleotide encoding IL-35 receptor | |
Beer et al. | Impaired immune responses and prolonged allograft survival in Sly1 mutant mice | |
Walker et al. | Established T cell-driven germinal center B cell proliferation is independent of CD28 signaling but is tightly regulated through CTLA-4 | |
Tsukumo et al. | AFF3, a susceptibility factor for autoimmune diseases, is a molecular facilitator of immunoglobulin class switch recombination | |
US20070178087A1 (en) | Method for Treating Immune Dysfunction by Regulation of CD40 Ligand Expression | |
Gao et al. | B cell induction of IL-13 expression in NK cells: role of CD244 and SLAM-associated protein | |
EP1439223A1 (en) | Novel class ii cytokine receptor | |
Whittaker et al. | Analysis of the linker for activation of T cells and the linker for activation of B cells in natural killer cells reveals a novel signaling cassette, dual usage in ITAM signaling, and influence on development of the Ly49 repertoire | |
Panneton | The role of inducible costimulator in autoimmunity | |
US20240262917A1 (en) | Antibodies against alk and methods of use thereof | |
Dasgupta et al. | Insulin receptor substrate (IRS)-2 negatively regulates alternative macrophage activation and allergic lung inflammation | |
Repetny et al. | Binding of LBP‐1a to specific immunoglobulin switch regions in vivo correlates with specific repression of class switch recombination | |
JP4386776B2 (en) | Human monoclonal antibody against costimulatory molecule AILIM and pharmaceutical use thereof | |
Gong | Role of the CBL family of E3-Ubiquitin ligases in the homeostasis of T follicular helper cells | |
Walker | ME (Linda) Joosse Iris Nederlof Lucy SK Walker Janneke N. Samsom | |
Kin | A study of the mechanism by which CD86 regulates IgG1 | |
Huan | TFE3 and TFEB are E-cadherin and CD40 ligand transcription activators | |
WO2016084412A1 (en) | B cell activation inhibitor, and therapeutic agent for autoimmune diseases |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
DPE2 | Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101) | ||
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2006840267 Country of ref document: EP |
|
DPE2 | Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101) |