US20130195882A1 - Mcpip protection against osteoclast production - Google Patents
Mcpip protection against osteoclast production Download PDFInfo
- Publication number
- US20130195882A1 US20130195882A1 US13/362,261 US201213362261A US2013195882A1 US 20130195882 A1 US20130195882 A1 US 20130195882A1 US 201213362261 A US201213362261 A US 201213362261A US 2013195882 A1 US2013195882 A1 US 2013195882A1
- Authority
- US
- United States
- Prior art keywords
- mcpip
- expression
- patient
- composition
- induced
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Abandoned
Links
- 238000004519 manufacturing process Methods 0.000 title claims description 36
- 210000002997 osteoclast Anatomy 0.000 title claims description 16
- 101100545174 Mus musculus Zc3h12a gene Proteins 0.000 title 1
- 238000000034 method Methods 0.000 claims abstract description 57
- 101100545173 Homo sapiens ZC3H12A gene Proteins 0.000 claims abstract description 18
- 230000002757 inflammatory effect Effects 0.000 claims abstract description 12
- 208000024891 symptom Diseases 0.000 claims abstract description 12
- 230000001747 exhibiting effect Effects 0.000 claims abstract description 9
- 230000002917 arthritic effect Effects 0.000 claims abstract description 6
- 230000002401 inhibitory effect Effects 0.000 claims abstract description 6
- 208000012659 Joint disease Diseases 0.000 claims abstract description 5
- 230000001009 osteoporotic effect Effects 0.000 claims abstract description 5
- 102100023882 Endoribonuclease ZC3H12A Human genes 0.000 claims abstract 17
- 230000014509 gene expression Effects 0.000 claims description 116
- 210000004027 cell Anatomy 0.000 claims description 69
- 239000004055 small Interfering RNA Substances 0.000 claims description 48
- 108020004459 Small interfering RNA Proteins 0.000 claims description 41
- 239000000203 mixture Substances 0.000 claims description 25
- 108020004999 messenger RNA Proteins 0.000 claims description 15
- 108091027967 Small hairpin RNA Proteins 0.000 claims description 11
- 206010039073 rheumatoid arthritis Diseases 0.000 claims description 9
- 208000001132 Osteoporosis Diseases 0.000 claims description 8
- 230000000692 anti-sense effect Effects 0.000 claims description 8
- 239000002773 nucleotide Substances 0.000 claims description 8
- 125000003729 nucleotide group Chemical group 0.000 claims description 8
- 230000009471 action Effects 0.000 claims description 7
- 208000006386 Bone Resorption Diseases 0.000 claims description 6
- 210000001185 bone marrow Anatomy 0.000 claims description 6
- 230000024279 bone resorption Effects 0.000 claims description 6
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 5
- 208000027866 inflammatory disease Diseases 0.000 claims description 4
- 230000000903 blocking effect Effects 0.000 claims description 3
- 210000005087 mononuclear cell Anatomy 0.000 claims description 3
- 201000008482 osteoarthritis Diseases 0.000 claims description 2
- 230000002159 abnormal effect Effects 0.000 claims 1
- 206010003246 arthritis Diseases 0.000 claims 1
- 230000002685 pulmonary effect Effects 0.000 claims 1
- 230000004069 differentiation Effects 0.000 description 75
- 108010032050 Tartrate-Resistant Acid Phosphatase Proteins 0.000 description 70
- 101710155857 C-C motif chemokine 2 Proteins 0.000 description 67
- 102000000018 Chemokine CCL2 Human genes 0.000 description 67
- 239000002243 precursor Substances 0.000 description 63
- 102000007591 Tartrate-Resistant Acid Phosphatase Human genes 0.000 description 62
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 56
- 230000035882 stress Effects 0.000 description 55
- 108090000625 Cathepsin K Proteins 0.000 description 53
- 102000004171 Cathepsin K Human genes 0.000 description 52
- 239000003642 reactive oxygen metabolite Substances 0.000 description 49
- 230000004900 autophagic degradation Effects 0.000 description 45
- 108090000623 proteins and genes Proteins 0.000 description 42
- DFYRUELUNQRZTB-UHFFFAOYSA-N apocynin Chemical compound COC1=CC(C(C)=O)=CC=C1O DFYRUELUNQRZTB-UHFFFAOYSA-N 0.000 description 30
- 102000004072 Beclin-1 Human genes 0.000 description 28
- 102000004169 proteins and genes Human genes 0.000 description 28
- 108090000524 Beclin-1 Proteins 0.000 description 26
- 238000003119 immunoblot Methods 0.000 description 25
- 102100030013 Endoribonuclease Human genes 0.000 description 22
- 108091006081 Inositol-requiring enzyme-1 Proteins 0.000 description 22
- 230000000694 effects Effects 0.000 description 22
- 238000001890 transfection Methods 0.000 description 21
- 108010017007 glucose-regulated proteins Proteins 0.000 description 20
- 230000006698 induction Effects 0.000 description 20
- 210000001616 monocyte Anatomy 0.000 description 18
- 230000005764 inhibitory process Effects 0.000 description 16
- 229930188866 apocynin Natural products 0.000 description 15
- 239000003112 inhibitor Substances 0.000 description 15
- 230000001404 mediated effect Effects 0.000 description 15
- CETPSERCERDGAM-UHFFFAOYSA-N ceric oxide Chemical compound O=[Ce]=O CETPSERCERDGAM-UHFFFAOYSA-N 0.000 description 14
- 229910000422 cerium(IV) oxide Inorganic materials 0.000 description 14
- 238000003197 gene knockdown Methods 0.000 description 14
- 210000004379 membrane Anatomy 0.000 description 14
- 239000012528 membrane Substances 0.000 description 14
- 230000005945 translocation Effects 0.000 description 12
- 230000015572 biosynthetic process Effects 0.000 description 11
- 108010002998 NADPH Oxidases Proteins 0.000 description 10
- 102000004722 NADPH Oxidases Human genes 0.000 description 10
- 239000000411 inducer Substances 0.000 description 10
- 239000003550 marker Substances 0.000 description 10
- 238000003753 real-time PCR Methods 0.000 description 10
- 238000004458 analytical method Methods 0.000 description 9
- 150000001875 compounds Chemical class 0.000 description 9
- 230000001965 increasing effect Effects 0.000 description 9
- BHTRKEVKTKCXOH-LBSADWJPSA-N tauroursodeoxycholic acid Chemical compound C([C@H]1C[C@@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC(=O)NCCS(O)(=O)=O)C)[C@@]2(C)CC1 BHTRKEVKTKCXOH-LBSADWJPSA-N 0.000 description 9
- 102000006495 integrins Human genes 0.000 description 8
- 108010044426 integrins Proteins 0.000 description 8
- 239000013598 vector Substances 0.000 description 8
- 238000011529 RT qPCR Methods 0.000 description 7
- 230000004913 activation Effects 0.000 description 7
- 230000001413 cellular effect Effects 0.000 description 7
- 201000010099 disease Diseases 0.000 description 7
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 7
- 239000013604 expression vector Substances 0.000 description 7
- 230000008569 process Effects 0.000 description 7
- 101150102163 ATG7 gene Proteins 0.000 description 6
- 101000990902 Homo sapiens Matrix metalloproteinase-9 Proteins 0.000 description 6
- 102100030412 Matrix metalloproteinase-9 Human genes 0.000 description 6
- 108091034117 Oligonucleotide Proteins 0.000 description 6
- 102000014128 RANK Ligand Human genes 0.000 description 6
- 108010025832 RANK Ligand Proteins 0.000 description 6
- YJQCOFNZVFGCAF-UHFFFAOYSA-N Tunicamycin II Natural products O1C(CC(O)C2C(C(O)C(O2)N2C(NC(=O)C=C2)=O)O)C(O)C(O)C(NC(=O)C=CCCCCCCCCC(C)C)C1OC1OC(CO)C(O)C(O)C1NC(C)=O YJQCOFNZVFGCAF-UHFFFAOYSA-N 0.000 description 6
- 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 6
- 239000000463 material Substances 0.000 description 6
- FNEZBBILNYNQGC-UHFFFAOYSA-N methyl 2-(3,6-diamino-9h-xanthen-9-yl)benzoate Chemical compound COC(=O)C1=CC=CC=C1C1C2=CC=C(N)C=C2OC2=CC(N)=CC=C21 FNEZBBILNYNQGC-UHFFFAOYSA-N 0.000 description 6
- 230000036542 oxidative stress Effects 0.000 description 6
- ZHSGGJXRNHWHRS-VIDYELAYSA-N tunicamycin Chemical compound O([C@H]1[C@@H]([C@H]([C@@H](O)[C@@H](CC(O)[C@@H]2[C@H]([C@@H](O)[C@@H](O2)N2C(NC(=O)C=C2)=O)O)O1)O)NC(=O)/C=C/CC(C)C)[C@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1NC(C)=O ZHSGGJXRNHWHRS-VIDYELAYSA-N 0.000 description 6
- MEYZYGMYMLNUHJ-UHFFFAOYSA-N tunicamycin Natural products CC(C)CCCCCCCCCC=CC(=O)NC1C(O)C(O)C(CC(O)C2OC(C(O)C2O)N3C=CC(=O)NC3=O)OC1OC4OC(CO)C(O)C(O)C4NC(=O)C MEYZYGMYMLNUHJ-UHFFFAOYSA-N 0.000 description 6
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 5
- 206010061218 Inflammation Diseases 0.000 description 5
- 241000699666 Mus <mouse, genus> Species 0.000 description 5
- HATRDXDCPOXQJX-UHFFFAOYSA-N Thapsigargin Natural products CCCCCCCC(=O)OC1C(OC(O)C(=C/C)C)C(=C2C3OC(=O)C(C)(O)C3(O)C(CC(C)(OC(=O)C)C12)OC(=O)CCC)C HATRDXDCPOXQJX-UHFFFAOYSA-N 0.000 description 5
- 230000011759 adipose tissue development Effects 0.000 description 5
- 239000000074 antisense oligonucleotide Substances 0.000 description 5
- 238000012230 antisense oligonucleotides Methods 0.000 description 5
- 230000004054 inflammatory process Effects 0.000 description 5
- 239000002105 nanoparticle Substances 0.000 description 5
- 230000037361 pathway Effects 0.000 description 5
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 5
- 238000010186 staining Methods 0.000 description 5
- 239000003826 tablet Substances 0.000 description 5
- IXFPJGBNCFXKPI-FSIHEZPISA-N thapsigargin Chemical compound CCCC(=O)O[C@H]1C[C@](C)(OC(C)=O)[C@H]2[C@H](OC(=O)CCCCCCC)[C@@H](OC(=O)C(\C)=C/C)C(C)=C2[C@@H]2OC(=O)[C@@](C)(O)[C@]21O IXFPJGBNCFXKPI-FSIHEZPISA-N 0.000 description 5
- QDLHCMPXEPAAMD-QAIWCSMKSA-N wortmannin Chemical compound C1([C@]2(C)C3=C(C4=O)OC=C3C(=O)O[C@@H]2COC)=C4[C@@H]2CCC(=O)[C@@]2(C)C[C@H]1OC(C)=O QDLHCMPXEPAAMD-QAIWCSMKSA-N 0.000 description 5
- QDLHCMPXEPAAMD-UHFFFAOYSA-N wortmannin Natural products COCC1OC(=O)C2=COC(C3=O)=C2C1(C)C1=C3C2CCC(=O)C2(C)CC1OC(C)=O QDLHCMPXEPAAMD-UHFFFAOYSA-N 0.000 description 5
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 4
- 206010051728 Bone erosion Diseases 0.000 description 4
- 108010057466 NF-kappa B Proteins 0.000 description 4
- 102000003945 NF-kappa B Human genes 0.000 description 4
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 4
- 230000033115 angiogenesis Effects 0.000 description 4
- 210000000988 bone and bone Anatomy 0.000 description 4
- 239000003153 chemical reaction reagent Substances 0.000 description 4
- 210000000805 cytoplasm Anatomy 0.000 description 4
- 230000009368 gene silencing by RNA Effects 0.000 description 4
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 4
- 230000007246 mechanism Effects 0.000 description 4
- 230000002018 overexpression Effects 0.000 description 4
- 230000001105 regulatory effect Effects 0.000 description 4
- 238000012360 testing method Methods 0.000 description 4
- 230000003827 upregulation Effects 0.000 description 4
- 238000001262 western blot Methods 0.000 description 4
- 206010065687 Bone loss Diseases 0.000 description 3
- 206010022489 Insulin Resistance Diseases 0.000 description 3
- 108091054455 MAP kinase family Proteins 0.000 description 3
- 102000043136 MAP kinase family Human genes 0.000 description 3
- 239000012828 PI3K inhibitor Substances 0.000 description 3
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 3
- 208000035896 Twin-reversed arterial perfusion sequence Diseases 0.000 description 3
- 239000011575 calcium Substances 0.000 description 3
- 230000024245 cell differentiation Effects 0.000 description 3
- 239000013592 cell lysate Substances 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 238000012637 gene transfection Methods 0.000 description 3
- 230000002518 glial effect Effects 0.000 description 3
- 230000006872 improvement Effects 0.000 description 3
- 238000001802 infusion Methods 0.000 description 3
- 210000002540 macrophage Anatomy 0.000 description 3
- 102000002574 p38 Mitogen-Activated Protein Kinases Human genes 0.000 description 3
- 108010068338 p38 Mitogen-Activated Protein Kinases Proteins 0.000 description 3
- 229940043441 phosphoinositide 3-kinase inhibitor Drugs 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 239000000047 product Substances 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 230000000638 stimulation Effects 0.000 description 3
- 230000004083 survival effect Effects 0.000 description 3
- 238000002560 therapeutic procedure Methods 0.000 description 3
- 230000035897 transcription Effects 0.000 description 3
- 238000013518 transcription Methods 0.000 description 3
- 230000004906 unfolded protein response Effects 0.000 description 3
- 230000003612 virological effect Effects 0.000 description 3
- 102100022900 Actin, cytoplasmic 1 Human genes 0.000 description 2
- 108010085238 Actins Proteins 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 102100031151 C-C chemokine receptor type 2 Human genes 0.000 description 2
- 101710149815 C-C chemokine receptor type 2 Proteins 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- HEDRZPFGACZZDS-UHFFFAOYSA-N Chloroform Chemical compound ClC(Cl)Cl HEDRZPFGACZZDS-UHFFFAOYSA-N 0.000 description 2
- 229920002261 Corn starch Polymers 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 102000004190 Enzymes Human genes 0.000 description 2
- 108090000790 Enzymes Proteins 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- 102000008607 Integrin beta3 Human genes 0.000 description 2
- 108010020950 Integrin beta3 Proteins 0.000 description 2
- KFZMGEQAYNKOFK-UHFFFAOYSA-N Isopropanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 2
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 2
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 2
- 102000038030 PI3Ks Human genes 0.000 description 2
- 108091007960 PI3Ks Proteins 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 241000700584 Simplexvirus Species 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 101710185494 Zinc finger protein Proteins 0.000 description 2
- 102100023597 Zinc finger protein 816 Human genes 0.000 description 2
- 210000001789 adipocyte Anatomy 0.000 description 2
- 150000001413 amino acids Chemical class 0.000 description 2
- 229940121363 anti-inflammatory agent Drugs 0.000 description 2
- 239000002260 anti-inflammatory agent Substances 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 230000027455 binding Effects 0.000 description 2
- 230000031018 biological processes and functions Effects 0.000 description 2
- 230000008512 biological response Effects 0.000 description 2
- 210000002798 bone marrow cell Anatomy 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 239000013000 chemical inhibitor Substances 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 239000008120 corn starch Substances 0.000 description 2
- 229940099112 cornstarch Drugs 0.000 description 2
- 230000003247 decreasing effect Effects 0.000 description 2
- 238000003745 diagnosis Methods 0.000 description 2
- 239000007888 film coating Substances 0.000 description 2
- 238000009501 film coating Methods 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 230000029226 lipidation Effects 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 230000033001 locomotion Effects 0.000 description 2
- 235000019359 magnesium stearate Nutrition 0.000 description 2
- 239000008108 microcrystalline cellulose Substances 0.000 description 2
- 229940016286 microcrystalline cellulose Drugs 0.000 description 2
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 230000001590 oxidative effect Effects 0.000 description 2
- 230000001575 pathological effect Effects 0.000 description 2
- 210000005259 peripheral blood Anatomy 0.000 description 2
- 239000011886 peripheral blood Substances 0.000 description 2
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N phenylmethanesulfonyl fluoride Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 2
- 239000013612 plasmid Substances 0.000 description 2
- 238000003752 polymerase chain reaction Methods 0.000 description 2
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 2
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 2
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 2
- 230000034190 positive regulation of NF-kappaB transcription factor activity Effects 0.000 description 2
- 210000000229 preadipocyte Anatomy 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 2
- 239000000375 suspending agent Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 239000000454 talc Substances 0.000 description 2
- 229910052623 talc Inorganic materials 0.000 description 2
- 235000012222 talc Nutrition 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 2
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-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
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical group C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- SARKXLKWFKNUMR-UHFFFAOYSA-N 4-benzamido-5-chloro-2-methylbenzenediazonium;chloride Chemical compound [Cl-].C1=C([N+]#N)C(C)=CC(NC(=O)C=2C=CC=CC=2)=C1Cl SARKXLKWFKNUMR-UHFFFAOYSA-N 0.000 description 1
- 235000019489 Almond oil Nutrition 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 108020005544 Antisense RNA Proteins 0.000 description 1
- 241000219194 Arabidopsis Species 0.000 description 1
- 208000032791 BCR-ABL1 positive chronic myelogenous leukemia Diseases 0.000 description 1
- 208000008035 Back Pain Diseases 0.000 description 1
- 208000010392 Bone Fractures Diseases 0.000 description 1
- 102000001902 CC Chemokines Human genes 0.000 description 1
- 108010040471 CC Chemokines Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000282461 Canis lupus Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 101000950671 Chelon ramada Myosin light chain 3, skeletal muscle isoform Proteins 0.000 description 1
- 108010077544 Chromatin Proteins 0.000 description 1
- 208000010833 Chronic myeloid leukaemia Diseases 0.000 description 1
- 241001062872 Cleyera japonica Species 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- 230000004568 DNA-binding Effects 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 108700041152 Endoplasmic Reticulum Chaperone BiP Proteins 0.000 description 1
- 102100021451 Endoplasmic reticulum chaperone BiP Human genes 0.000 description 1
- 241000792859 Enema Species 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 229940123457 Free radical scavenger Drugs 0.000 description 1
- 102000003688 G-Protein-Coupled Receptors Human genes 0.000 description 1
- 108090000045 G-Protein-Coupled Receptors Proteins 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 239000012981 Hank's balanced salt solution Substances 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000897480 Homo sapiens C-C motif chemokine 2 Proteins 0.000 description 1
- 101000973495 Homo sapiens E3 ubiquitin-protein ligase MIB2 Proteins 0.000 description 1
- 101001052506 Homo sapiens Microtubule-associated proteins 1A/1B light chain 3A Proteins 0.000 description 1
- 101001071230 Homo sapiens PHD finger protein 20 Proteins 0.000 description 1
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 1
- 206010023232 Joint swelling Diseases 0.000 description 1
- -1 LC3-II Proteins 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 239000012741 Laemmli sample buffer Substances 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 1
- 240000007472 Leucaena leucocephala Species 0.000 description 1
- GDBQQVLCIARPGH-UHFFFAOYSA-N Leupeptin Natural products CC(C)CC(NC(C)=O)C(=O)NC(CC(C)C)C(=O)NC(C=O)CCCN=C(N)N GDBQQVLCIARPGH-UHFFFAOYSA-N 0.000 description 1
- 239000012097 Lipofectamine 2000 Substances 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 208000033761 Myelogenous Chronic BCR-ABL Positive Leukemia Diseases 0.000 description 1
- ACFIXJIJDZMPPO-NNYOXOHSSA-N NADPH Chemical compound C1=CCC(C(=O)N)=CN1[C@H]1[C@H](O)[C@H](O)[C@@H](COP(O)(=O)OP(O)(=O)OC[C@@H]2[C@H]([C@@H](OP(O)(O)=O)[C@@H](O2)N2C3=NC=NC(N)=C3N=C2)O)O1 ACFIXJIJDZMPPO-NNYOXOHSSA-N 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 102000004316 Oxidoreductases Human genes 0.000 description 1
- 108090000854 Oxidoreductases Proteins 0.000 description 1
- 102100036878 PHD finger protein 20 Human genes 0.000 description 1
- 108010016731 PPAR gamma Proteins 0.000 description 1
- 239000002033 PVDF binder Substances 0.000 description 1
- 208000002193 Pain Diseases 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 229930182555 Penicillin Natural products 0.000 description 1
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 1
- 102100038825 Peroxisome proliferator-activated receptor gamma Human genes 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 102000000574 RNA-Induced Silencing Complex Human genes 0.000 description 1
- 108010016790 RNA-Induced Silencing Complex Proteins 0.000 description 1
- 230000004570 RNA-binding Effects 0.000 description 1
- 102000006382 Ribonucleases Human genes 0.000 description 1
- 108010083644 Ribonucleases Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 229940124639 Selective inhibitor Drugs 0.000 description 1
- 102000012479 Serine Proteases Human genes 0.000 description 1
- 108010022999 Serine Proteases Proteins 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 239000004141 Sodium laurylsulphate Substances 0.000 description 1
- 206010041541 Spinal compression fracture Diseases 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 102000004142 Trypsin Human genes 0.000 description 1
- 108090000631 Trypsin Proteins 0.000 description 1
- IOMLBTHPCVDRHM-UHFFFAOYSA-N [3-[(2,4-dimethylphenyl)carbamoyl]naphthalen-2-yl] dihydrogen phosphate Chemical compound CC1=CC(C)=CC=C1NC(=O)C1=CC2=CC=CC=C2C=C1OP(O)(O)=O IOMLBTHPCVDRHM-UHFFFAOYSA-N 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 239000000362 adenosine triphosphatase inhibitor Substances 0.000 description 1
- 239000000556 agonist Substances 0.000 description 1
- 239000008168 almond oil Substances 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 230000000840 anti-viral effect Effects 0.000 description 1
- 239000008135 aqueous vehicle Substances 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000003305 autocrine Effects 0.000 description 1
- 210000004957 autophagosome Anatomy 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 244000309464 bull Species 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 229960005069 calcium Drugs 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 229960001714 calcium phosphate Drugs 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 210000004413 cardiac myocyte Anatomy 0.000 description 1
- 210000000845 cartilage Anatomy 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000004640 cellular pathway Effects 0.000 description 1
- 229910000420 cerium oxide Inorganic materials 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 238000002038 chemiluminescence detection Methods 0.000 description 1
- 210000003483 chromatin Anatomy 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 229940075614 colloidal silicon dioxide Drugs 0.000 description 1
- 239000003184 complementary RNA Substances 0.000 description 1
- 238000013270 controlled release Methods 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000034994 death Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 239000003405 delayed action preparation Substances 0.000 description 1
- 238000010217 densitometric analysis Methods 0.000 description 1
- 238000001514 detection method Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 230000008034 disappearance Effects 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000002158 endotoxin Substances 0.000 description 1
- 239000007920 enema Substances 0.000 description 1
- 229940079360 enema for constipation Drugs 0.000 description 1
- 230000007705 epithelial mesenchymal transition Effects 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 235000019441 ethanol Nutrition 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 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
- 238000002474 experimental method Methods 0.000 description 1
- 239000013613 expression plasmid Substances 0.000 description 1
- 208000028327 extreme fatigue Diseases 0.000 description 1
- 239000003925 fat Substances 0.000 description 1
- 239000012091 fetal bovine serum Substances 0.000 description 1
- 210000002950 fibroblast Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 230000007760 free radical scavenging Effects 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 230000003394 haemopoietic effect Effects 0.000 description 1
- 230000011132 hemopoiesis Effects 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 238000010842 high-capacity cDNA reverse transcription kit Methods 0.000 description 1
- 102000046768 human CCL2 Human genes 0.000 description 1
- 102000050964 human ZC3H12A Human genes 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 1
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 1
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 1
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 1
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 1
- UFVKGYZPFZQRLF-UHFFFAOYSA-N hydroxypropyl methyl cellulose Chemical compound OC1C(O)C(OC)OC(CO)C1OC1C(O)C(O)C(OC2C(C(O)C(OC3C(C(O)C(O)C(CO)O3)O)C(CO)O2)O)C(CO)O1 UFVKGYZPFZQRLF-UHFFFAOYSA-N 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 230000001939 inductive effect Effects 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 238000010255 intramuscular injection Methods 0.000 description 1
- 239000007927 intramuscular injection Substances 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 239000002555 ionophore Substances 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 210000001503 joint Anatomy 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 229960001375 lactose Drugs 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- GDBQQVLCIARPGH-ULQDDVLXSA-N leupeptin Chemical compound CC(C)C[C@H](NC(C)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C=O)CCCN=C(N)N GDBQQVLCIARPGH-ULQDDVLXSA-N 0.000 description 1
- 108010052968 leupeptin Proteins 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 229920006008 lipopolysaccharide Polymers 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 208000028755 loss of height Diseases 0.000 description 1
- 239000007937 lozenge Substances 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 206010025482 malaise Diseases 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 229960001855 mannitol Drugs 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 108091070501 miRNA Proteins 0.000 description 1
- 239000002679 microRNA Substances 0.000 description 1
- 239000011859 microparticle Substances 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 239000007758 minimum essential medium Substances 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 239000003068 molecular probe Substances 0.000 description 1
- 125000004573 morpholin-4-yl group Chemical group N1(CCOCC1)* 0.000 description 1
- 230000004031 neuronal differentiation Effects 0.000 description 1
- 229930027945 nicotinamide-adenine dinucleotide Natural products 0.000 description 1
- 230000006610 nonapoptotic cell death Effects 0.000 description 1
- 239000002687 nonaqueous vehicle Substances 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 210000004940 nucleus Anatomy 0.000 description 1
- 230000030648 nucleus localization Effects 0.000 description 1
- 230000011164 ossification Effects 0.000 description 1
- 230000002177 osteoclastogenic effect Effects 0.000 description 1
- 239000007800 oxidant agent Substances 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- BMMGVYCKOGBVEV-UHFFFAOYSA-N oxo(oxoceriooxy)cerium Chemical compound [Ce]=O.O=[Ce]=O BMMGVYCKOGBVEV-UHFFFAOYSA-N 0.000 description 1
- 210000003134 paneth cell Anatomy 0.000 description 1
- 230000003076 paracrine Effects 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229940049954 penicillin Drugs 0.000 description 1
- 229950000964 pepstatin Drugs 0.000 description 1
- 108010091212 pepstatin Proteins 0.000 description 1
- FAXGPCHRFPCXOO-LXTPJMTPSA-N pepstatin A Chemical compound OC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C(C)C)NC(=O)[C@H](C(C)C)NC(=O)CC(C)C FAXGPCHRFPCXOO-LXTPJMTPSA-N 0.000 description 1
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 229920000642 polymer Polymers 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- 230000001124 posttranscriptional effect Effects 0.000 description 1
- LJCNRYVRMXRIQR-OLXYHTOASA-L potassium sodium L-tartrate Chemical compound [Na+].[K+].[O-]C(=O)[C@H](O)[C@@H](O)C([O-])=O LJCNRYVRMXRIQR-OLXYHTOASA-L 0.000 description 1
- 229940074439 potassium sodium tartrate Drugs 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 210000005238 principal cell Anatomy 0.000 description 1
- 235000010232 propyl p-hydroxybenzoate Nutrition 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 239000002516 radical scavenger Substances 0.000 description 1
- 150000003254 radicals Chemical class 0.000 description 1
- 108091006084 receptor activators Proteins 0.000 description 1
- 230000019254 respiratory burst Effects 0.000 description 1
- 230000016914 response to endoplasmic reticulum stress Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 230000009919 sequestration Effects 0.000 description 1
- 238000007493 shaping process Methods 0.000 description 1
- 239000002924 silencing RNA Substances 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 1
- 235000011006 sodium potassium tartrate Nutrition 0.000 description 1
- 229940080313 sodium starch Drugs 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 235000010356 sorbitol Nutrition 0.000 description 1
- 206010041569 spinal fracture Diseases 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 239000012192 staining solution Substances 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 229960005322 streptomycin Drugs 0.000 description 1
- 238000010254 subcutaneous injection Methods 0.000 description 1
- 239000007929 subcutaneous injection Substances 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 230000002459 sustained effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 201000004595 synovitis Diseases 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 239000006188 syrup Substances 0.000 description 1
- 235000020357 syrup Nutrition 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 1
- 230000036346 tooth eruption Effects 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 102000035160 transmembrane proteins Human genes 0.000 description 1
- 108091005703 transmembrane proteins Proteins 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 235000012431 wafers Nutrition 0.000 description 1
- 230000004580 weight loss Effects 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
- C12N15/1136—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/713—Double-stranded nucleic acids or oligonucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/28—Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/02—Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/08—Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
- A61P19/10—Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- 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
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/11—Antisense
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/14—Type of nucleic acid interfering N.A.
Definitions
- the invention relates to cellular factors for modulating cellular pathways. More particularly, the invention relates to cellular transcription factors that affect inflammation and the differentiation of osteoclasts, and methods for their therapeutic use.
- osteoclast is the principal cell type responsible for bone resorption in inflammatory joint diseases (Harris, 1990; Sakiyama et al., 2001; Sato and Takayanagi, 2006; Mundy, 2007).
- Rheumatoid arthritis is characterized by the presence of inflammatory synovitis accompanied by the destruction of the joint cartilage and bone (Harris, 1990; Mundy, 2007; Sugimura and Li, 2010).
- OCs are bone-resorbing cells that differentiate from hematopoietic precursors of the monocyte/macrophage lineage (Sakiyama et al., 2001; Boyce et al., 2007).
- OCs are multinuclear giant cells that stain positive for tartrate-resistant acid phosphatase (TRAP) and serine protease cathepsin K (CTSK) (Kiviranta et al., 2001; Boyce et al., 2007).
- TRIP tartrate-resistant acid phosphatase
- CSK serine protease cathepsin K
- MCP-1 Monocyte chemotactic protein-1
- NF- ⁇ B nuclear factor- ⁇ B
- MCP-1 The cellular effect of MCP-1 is mediated by the CCR2, a G-protein-coupled receptor that is induced by the receptor activator of RANKL (Gerszten et al., 2001; Kim et al., 2005).
- the signaling process initiated by MCP-1 binding to CCR2 leads to changes in gene expression.
- MCPIP zinc-finger protein
- MCPIP mediates OC differentiation
- MCPIP mediates several biological processes initiated by MCP-1. including cardiomyocyte death (Younce and Kolattukudy, 2010; Younce et al., 2010), adipogenesis (Younce et al., 2009), angiogenesis (Niu et al., 2008), and glial differentiation of neuroprogenitor cells (Vrotsos et al., 2009). It has been discovered, as disclosed herein, that MCP-1 induces differentiation of monocytic cells into OC precursors via MCPIP.
- MCPIP mediates differentiation of OC precursors via induction of oxidative stress that causes endoplasmic reticulum (ER) stress that leads to autophagy involved in osteoclastogenesis.
- FIG. 1 MCP-1 induction of OC precursor differentiation, mediated by MCPIP.
- BMCs were transfected with FLAG-tagged MCPIP (WT-MCPIP) or FLAG-tagged empty vector (MAT-FLAG).
- TRAP-expressing cells were stained and viewed under the Nikon microscope (A).
- BMNCs treated with MCP-1 alone or with MCPIP siRNA or Scr siRNA.
- TRAP-expressing cells were stained and TRAP-positive cell proportion was measured (B). At least three fields ( ⁇ 500 cells), were chosen. *P ⁇ 0.05 and **P ⁇ 0.01 versus control; # P ⁇ 0.05 versus Scr.
- C Immunoblotting showing the expression of TRAP, CTSK, and MCPIP induced by MCP-1.
- FIG. 2 MCPIP overexpression induces OC-related marker TRAP and CTSK expression.
- A Real-time PCR showing the transcription of TRAP, CTSK, and MCPIP (upper panel) and ⁇ V integrin, ⁇ 3 integrin, and MMP9 (lower panel) induced by MCPIP transfection. *P ⁇ 0.05 and **P ⁇ 0.01 versus control or GFP; # P ⁇ 0.05 versus day 2 or 6.
- C Percentage of TRAP-positive cells. *P ⁇ 0.01 versus GFP; # P ⁇ 0.01 versus none or Scr.
- FIG. 3 ROS production involvement in MCPIP/MCP-1-induced OC precursor differentiation.
- A Western blot showed that MCP-1 treatment or MCPIP transfection induces p47 PHOX expression. *P ⁇ 0.05 versus control or GFP; # P ⁇ 0.05 versus Scr.
- B Immunoblotting showed that MCP-1 or MCPIP overexpression induces an increase in cytoplasmic membrane-associated p47 PHOX . * P ⁇ 0.05 versus control or GFP; # P ⁇ 0.05 versus Scr.
- C ROS production induced by MCP-1 or MCPIP transfection, or siRNA was detected by using DHR123. *P ⁇ 0.05 versus control or GFP; # P ⁇ 0.05 versus Scr.
- FIG. 4 MCPIP induction of ER stress via ROS production involved in OC precursor differentiation.
- A Immunoblotting shown that MCP-1 or MCPIP transfection induces the expression of GRP78 and IRE-1 (ER stress markers) by inducing ROS production.
- B Blockage of ER stress by IRE-1 siRNA abolished MCPIP-induced mRNA and protein expression of TRAP and CTSK. *P ⁇ 0.05 versus GFP, # P ⁇ 0.05 versus Scr.
- C Blockage of ER stress by TUDC abolished MCPIP-induced mRNA and protein expression of TRAP and CTSK. *P ⁇ 0.05 versus GFP, # P ⁇ 0.05 versus MCPIP alone.
- FIG. 5 MCPIP induction of autophagy via ROS production and ER stress involved in OC precursor differentiation.
- A Immunoblotting shown that MCPIP induces autophagy characterized with the marker Beclin-1 expression and ROS/ER stress inhibitor inhibited MCPIP-induced expression of Beclin-1.
- B and C Immunoblotting shown that autophagy blocker 3′-MA and LY294002 blocked MCPIP-induced OC-related gene TRAP and CTSK expression but not GRP78.
- D Real-time PCR shown that autophagy blocker 3′-MA and LY294002 blocked MCPIP-induced expression of TRAP and CTSK but not GRP78. *P ⁇ 0.05 versus GFP, # P ⁇ 0.05 versus MCPIP alone.
- E Blocking autophagy by Beclin-1 siRNA inhibited MCPIP-induced mRNA and protein expression of TRAP and CTSK but not GRP78. *P ⁇ 0.05 versus MCPIP alone.
- FIG. 6 MCPIP-induced OC precursor differentiation inhibited by PI3K inhibitor, wortmannin.
- A qRT-PCR showing induction of Beclin-1 and TRAP and CTSK at the transcript level by MCPIP transfection, and the inhibition effect of wortmannin on MCPIP. *P ⁇ 0.05 versus MCPIP alone.
- B Immunoblotting show that MCPIP induced Beclin-1, and TRAP and CTSK, and that their induction was suppressed by wortmannin.
- C The intensities of immunoblots were measured and normalized to (3-actin of the corresponding group. *P ⁇ 0.05 compared with MCPIP alone.
- B Effect of p47 PHOX antisense oligonucleotides on expression of p47 PHOX and its translocation from cytoplasm to membrane [* P ⁇ 0.05 compared to non-sense oligonucleotides treated cells (“p47/NS”)].
- C Effect of p47 PHOX antisense oligonucleotides on MCPIP-induced TRAP and CTSK expression [* P ⁇ 0.05 compared to non-sense oligonucleotides treated cells (“p47/NS”)].
- D Effect of apocynin on expression of p47PHOX and its translocation from cytoplasm to membrane [* P ⁇ 0.05 compared to apocynin-untreated cells (“None”)].
- E Effect of apocynin on MCPIPinduced TRAP and CTSK expression [* P ⁇ 0.05 compared to wellcynin-untreated cells (“None”)].
- MCPIP induces ER stress via ROS production which involvement in OC precursor differentiation. After treatment, cell lysate was collected and analysed using immunoblot with appropriate antibody and results were quantified against (3-actin.
- A MCP-1 induces ER stress marker GRP78 and IRE-1 expression via MCPIP (* P ⁇ 0.05).
- B Effect of p47PHOX antisense oligonucleotides, CeO2 and apocynin on MCPIP-induced expression of GRP78 and IRE-1 (* P ⁇ 0.05).
- C inhibition of ER stress by using knock-down of IRE-1 attenuates MCPIP-induced expression of TRAP and CTSK (* P ⁇ 0.05).
- TUDC pretreatment of TUDC inhibits MCPIP-induced expression of TRAP and CTSK and ER stress marker IRE-1 and GRP78 (* P ⁇ 0.05).
- E ER stress inducer tunicamycin and thapsigargin induce expression of TRAP and CTSK (* P ⁇ 0.05).
- MCPIP induces autophagy via ROS production and ER stress which involvement in OC precursor differentiation. After treatment, cell lysate was collected and analysed using immunoblot with appropriate antibody and results were quantified against (3-actin.
- A MCPIP induces autophagy characterized with the marker beclin-1 expression and ROS inhibitor or ER stress inhibitor inhibited MCPIP-induced expression of beclin-1. (* P ⁇ 0.05).
- B autophagy blocker 3′-MA blocked MCPIP-induced osteoclast-related gene TRAP and CTSK expression but not GRP78 (* P ⁇ 0.05).
- C autophagy blocker LY-294002 blocked MCPIP induced osteoclast-related gene TRAP and CTSK expression but not GRP78. (* P ⁇ 0.05).
- D blockage of autophagy by beclin-1 specific siRNA inhibited MCPIP-induced osteoclast-related gene TRAP and CTSK expression but not GRP78 both in protein and mRNA level. * P ⁇ 0.05.
- MCPIP—induced autophagy marker Atg7 is necessary for induction of OC precursor differentiation.
- A qRT-PCR shows that transfection of MCPIP expression vector resulted in significantly increased mRNA levels of Atg7, Beclin-1, TRAP, and CTSK. Atg7-specific siRNA abolished MCPIP-induced expression of these genes. * P ⁇ 0.05 compared with MCPIP alone.
- B Immunoblot shows that transfection with MCPIP expression vector caused induction of beclin-1, TRAP, and CTSK at the protein levels. Upregulation of these marker genes were inhibited by Atg7-specific siRNA but were not affected by scrambled siRNA.
- C The intensity of each protein was measured and normalized to (3-actin of the corresponding group. * P ⁇ 0.05 compared with MCPIP alone.
- MCPIP MCP-1-induced protein
- MCP-1 induces MCPIP in human peripheral blood monocytes which induces differentiation of monocytes into osteoclast cells (OC) or OC precursors.
- OC osteoclast cells
- NF K B is a master controller of inflammation in the body. Agonists of MCPIP would enhance its anti-inflammatory potentcy; it is herein discovered that inhibition of the activation of NF K B by MCPIP provides a significant potential as an anti-inflammatory agent. Therefore, inhibition of the activation of NF K B by inhibiting MCPIP has a significant use as an anti-inflammatory agent.
- MCPIP inhibits inflammation, which plays a critical role in diseases including angiogenesis, i.e., the physiological process involving the growth of new blood vessels from pre-existing vessels, adipogenesis, the process of cell differentiation by which preadipocytes become adipocytes, and osteoclastogenesis, i.e., the development of osteoclasts.
- angiogenesis i.e., the physiological process involving the growth of new blood vessels from pre-existing vessels, adipogenesis, the process of cell differentiation by which preadipocytes become adipocytes, and osteoclastogenesis, i.e., the development of osteoclasts.
- MCPIP plays a critical role in the differentiation of osteoclasts involved in inflammatory bone loss that occurs in many inflammatory diseases such as, for example, rheumatoid arthritis.
- elevated MCP-1 levels have been identified as a direct cause of insulin resistance.
- MCPIP has been discovered to mediate insulin resistance, and thus, MCPIP inhibitors enhance insulin
- MCPIP plays a critical role in differentiation of osteoclasts involved in inflammatory bone loss that occurs in many inflammatory diseases such as rheumatoid arthritis. It is disclosed herein that differentiation of monocytes to OC precursors is mediated by MCPIP. Therefore, the blocking or inhibition of MCPIP provides a promising therapeutic strategy for preventing differentiation of monocytes to OC precursors, and ultimately the prevention of bone resorption. Bone resorption is a process by which osteoclasts break down bone and release minerals, resulting in a transfer of calcium from bone fluid to the blood. This discovery provides a particular benefit in inflammatory joint diseases.
- inflammatory bone erosion is involved in many pathological conditions (Lu et al., 2007; Ha et al., 2010).
- the novel inventive features described and contemplated herein provide a new insight into the mechanism by which MCP-1 induces differentiation of monocytic cells into TRAP- and CTSK-expressing cells that can proceed to differentiate into functional OCs in the presence of RANKL, and demonstrate that MCPIP is a novel target for therapy of inflammatory bone erosion, in one exemplary embodiment.
- a method of treating a condition in a patient in need includes administering to the patient a therapeutically effective amount of a composition that inhibits the expression or action of MCPIP.
- the patient in need may be exhibiting pre-arthritic symptoms including but not limited to pain or tenderness in a joint which is aggravated by movement or activity, inflammation indicated by joint swelling, stiffness, redness, and/or warmth, joint deformity, loss of range of motion or flexibility in a joint, or unexplained weight loss.
- Additional pre-arthritic symptoms include extreme fatigue, lack of energy, weakness or a feeling of malaise, a non-specific fever, or crepitus, i.e., creaky, popping or snapping joints.
- the patient in need may be exhibiting pre-osteoporotic symptoms, including but not limited to back pain caused by a fractured or collapsed vertebra, loss of height over time, a stooped posture, or a bone fracture that occurs much more easily than expected.
- the patient in need may also be exhibiting symptoms of an existing arthritic or osteoporotic disease, or both.
- the patient in need may be exhibiting symptoms of any arthritic or osteoporotic related-diseases, any inflammatory joint diseases, or any pre-disease symptoms of these related diseases.
- the condition may include an osteoporosis-related condition, and in a particular embodiment, rheumatoid arthritis.
- therapeutically effective amount refers to an amount sufficient to elicit the desired biological response.
- the desired biological response can be an overall improvement in the condition being treated.
- the overall improvement can be associated with improvement in individual symptoms.
- Subject or patient refers to animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, pigs, dogs, cats, rabbits, guinea pigs, rats, mice or other bovine, ovine, equine, canine, feline, rodent or murine species.
- the subject is a human.
- the compounds for use in the method of the invention can be formulated for oral, transdermal, sublingual, buccal, parenteral, rectal, intranasal, intrabronchial, intrapulmonary, or ocular administration. Oral administration is preferred.
- the compounds can be of the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., polyvinylpyrrolidone, hydroxypropylcellulose or hydroxypropylmethylcellulose); fillers (e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrates (e.g., sodium starch glycollate); or wetting agents (e.g., sodium lauryl sulphate).
- binding agents e.g., polyvinylpyrrolidone, hydroxypropylcellulose or hydroxypropylmethylcellulose
- fillers e.g., cornstarch
- the tablets can be coated using suitable methods and coating materials such as OPADRY film coating systems available from Colorcon, West Point, Pa. (e.g., OPADRY, OY Type, OY—C Type, Organic Enteric OY—P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRYTM White, 32K18400).
- suitable methods and coating materials such as OPADRY film coating systems available from Colorcon, West Point, Pa. (e.g., OPADRY, OY Type, OY—C Type, Organic Enteric OY—P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRYTM White, 32K18400).
- the oral form is a tablet containing MCPIP and a pharmaceutically acceptable excipient, such as, but not limited to mannitol, corn starch, microcrystalline cellulose, colloidal silicon dioxide, polyvinyl pyrrolidone, talc, magnesium stearate, and the like which are optionally coated with an OPADRY film coating.
- a pharmaceutically acceptable excipient such as, but not limited to mannitol, corn starch, microcrystalline cellulose, colloidal silicon dioxide, polyvinyl pyrrolidone, talc, magnesium stearate, and the like which are optionally coated with an OPADRY film coating.
- Liquid preparation for oral administration can be in the form of solutions, syrups or suspensions.
- the liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agent (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl p-hydroxy benzoates or sorbic acid).
- suspending agents e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats
- emulsifying agent e.g., lecithin or acacia
- non-aqueous vehicles e.g., almond oil, oily esters or ethyl alcohol
- preservatives e.g., methyl or propyl p-hydroxy benzoates or sorb
- the compounds for use in the method of the invention can be in the form of tablets or lozenges formulated in a conventional manner.
- the compounds for use in the method of the invention can be formulated for injection or infusion, for example, intravenous, intramuscular or subcutaneous injection or infusion, or for administration in a bolus dose and/or continuous infusion.
- Suspensions, solutions or emulsions in an oily or aqueous vehicle, optionally containing other formulatory agents such as suspending, stabilizing and/or dispersing agents can be used.
- the compounds for use in the method of the invention can be in the form of suppositories or enemas.
- tablets can be formulated in conventional manner.
- the compounds (e.g. protein or delivery vehicle) for use in the method of the invention can be formulated in a sustained release preparation.
- the compounds can be formulated with a suitable polymer or hydrophobic material which provides sustained and/or controlled release properties to the active agent compound.
- the compounds for use the method of the invention can be administered in the form of microparticles for example, by injection or in the form of wafers or discs by implantation.
- an expression vector is a viral or a non-viral expression vector.
- Viral expression vectors which may be used advantageously in the method of the invention include, but are not limited to, an adeno associated virus (AAV) vector, a lentivirus vector, an adenovirus vector, and a herpes simplex virus (HSV) vector.
- AAV adeno associated virus
- HSV herpes simplex virus
- the composition comprises siRNA or miRNA specific for MCPIP, an antisense nucleotide specific for MCPIP, and/or shRNA.
- the composition comprises an antibody specific to MCPIP.
- a method of inhibiting osteoclast production (or a method of reducing osteoclast precursor cells) in a patient in need includes administering a therapeutically effective amount of a composition that inhibits the expression or action of MCPIP in the patient.
- the patient in need in an embodiment, may be exhibiting symptoms of rheumatoid arthritis, osteoarthritis, and/or osteoporosis.
- administering a therapeutically effective amount of a composition includes a composition comprising: a composition that inhibits the expression or action of MCPIP, and a pharmaceutically acceptable excipient.
- the composition includes an MCPIP siRNA, an shRNA, an antibody specific to MCPIP, and/or an antisense nucleotide specific for MCPIP.
- siRNA small interfering/short interfering/silencing RNA
- SiRNA most often is involved in the RNA interference pathway where it interferes with the expression of a specific gene.
- siRNA also act in RNA interference-related pathways, e.g., as an antiviral mechanism or in shaping the chromatin structure of a genome.
- ShRNA short hairpin or small hairpin RNA
- shRNA refers to a sequence of RNA that makes a tight hairpin turn and is used to silence gene expression via RNA interference. It uses a vector introduced into cells and a U6 or H1 promoter to ensure that the shRNA is always expressed.
- the shRNA hairpin structure is cleaved by cellular machinery into siRNA which is then bound to the RNA-induced silencing complex. This complex binds to and cleaves mRNAs which match the siRNA that is bound to it.
- MCPIP can also be blocked by subjecting procured cells to an antibody specific to MCP-1.
- An antisense nucleotide may also be used to block or inhibit expression, in particular, the expression of MCP-1. Expression may also be inhibited with the use of a morpholino oligomer or phosphorodiamidate morpholino oligomer (PMO).
- PMOs are an antisense technology used to block access of other molecules to specific sequences within nucleic acid. PMOs are often used as a research tool for reverse genetics, and function by knocking down gene function. This is achieved by preventing cells from making a targeted protein or by modifying splicing of pre-mRNA.
- MCP-1 induces TRAP-positive OC precursor formation from human peripheral blood mononuclear cells (Kim et al., 2006a).
- 50 ng/ml MCP-1 induced TRAP-positive OC precursor cell formation from human bone marrow mononuclear cells (BMCs) ( FIG. 1B ).
- Immunoblotting FIG. 1C
- PCR real-time polymerase chain reaction
- FIG. 1D showed that MCP-1 treatment induced expression of OC markers TRAP and CTSK.
- MCP-1 did not affect the expression of the OC functional markers aV integrin, (33 integrin, and MMP9 ( FIG. 1E ).
- MCPIP small interfering RNA siRNA
- FIGS. 1C and D show that 50 ng/ml MCP-1 induced up-regulation of MCPIP protein and mRNA levels, which can be suppressed by treatment with MCPIP small interfering RNA (siRNA) ( FIGS. 1C and D).
- MCPIP small interfering RNA siRNA
- FIG. 1A Compared with FLAG-tagged empty vector (MAT-FLAG), expression of FLAG-tagged MCPIP (WT-FLAG) induced TRAP-positive OC precursor cell formation ( FIG. 1A ).
- MCPIP siRNA also significantly inhibited the formation of OC precursor cells that expressed TRAP and CTSK but showed no effects on expression of aV integrin, (33 integrin, and MMP9 ( FIG. 1C-E ).
- MCP-1-induced differentiation of OC precursors is mediated by MCPIP
- forced expression of MCPIP in monocytes might be expected to induce the formation of TRAP-positive OC precursor cells without MCP-1.
- BMCs were transfected to test this concept, as can be seen in FIG. 2 , with MCPIP-GFP expression vector. Increased expression of MCPIP was found 24 h after transfection at mRNA levels as measured by real-time PCR and its expression reached the peak at 4 days after transfection (see FIG. 2A , upper panel). The expression of TRAP and CTSK was induced at 2 days after MCPIP transfection and reached the peak at 4 days.
- ROS Reactive oxygen species
- Immunoblot analysis shows that MCP-1 treatment and MCPIP expression increased the expression of p47 PHOX ( FIG. 3A ) and its translocation from the cytoplasm into the membrane ( FIG. 3B ).
- ROS production caused by MCP-1 treatment and MCPIP expression was assessed by DHR123 staining and results showed that MCP-1 treatment and MCPIP transfection remarkably increased ROS generation.
- the effects of MCP-1 treatment or forced MCPIP expression on the expression and translocation of p47 PHOX and ROS generation were inhibited by MCPIP siRNA ( FIG. 3A-C ).
- apocynin an inhibitor of NADPH oxidase on MCPIP-induced induction of OC-related genes TRAP and CTSK was further tested herein. It was found that apocynin suppressed MCPIP-induced ROS production ( FIG. 3D ) and expression of TRAP and CTSK ( FIG. 3F and Supplementary Figure S 2 E). Moreover, apocynin inhibited MCPIP-induced expression and membrane translocation of p47 PHOX ( FIG. 3F and Supplementary Figure S 2 D).
- MCP-1 treatment induces ER stress in BMCs.
- Immunoblot analysis showed that MCP-1 treatment induced expression of the ER stress markers 78 kDa glucose regulated protein (GRP78) and inositol-requiring enzyme-1 (IRE-1) ( FIG. 4A , left panel, and Supplementary Figure S 3 A). This induction was inhibited by MCPIP siRNA ( FIG. 4A , left panel, and Supplementary Figure S 3 A), indicating that MCP-1 induced ER stress via MCPIP.
- GRP78 glucose regulated protein
- IRE-1 inositol-requiring enzyme-1
- MCPIP-induced ER stress was inhibited by IRE-1 siRNA ( FIG. 4B and Supplementary Figure S 3 C) and the ER stress-specific inhibitor tauroursodeoxycholate (TUDC) ( FIG. 4C and Supplementary Figure S 3 D).
- IRE-1 siRNA FIG. 4B and Supplementary Figure S 3 C
- TUDC ER stress-specific inhibitor tauroursodeoxycholate
- ER stress inducers might induce differentiation of OC precursors without other inducers.
- two ER stress inducers tunicamycin (TU) and thapsigargin (TH), induced expression of GRP78, IRE-1, TRAP, and CTSK at both protein and mRNA levels ( FIGS. 4D and E and Supplementary Figure S 3 E).
- TU tunicamycin
- TH thapsigargin
- Increased expression of Beclin-1 is a commonly used marker of autophagy (Wang, 2008). Autophagy has been implicated in differentiation in some cellular contexts (Baerga et al., 2009; Singh et al., 2009). However, it was heretofore unclear whether autophagy has involvement in OC differentiation. Here it was found that forced expression of MCPIP increased expression of Beclin-1; this effect was suppressed by MCPIP siRNA, but not by non-specific scramble (Scr) siRNA ( FIG. 5A , left panel and Supplementary Figure S 4 A). This result revealed that MCPIP induced autophagy in BMCs during differentiation into OC precursor cells.
- MCPIP-induced ROS production and ER stress are involved directly in MCPIP-mediated autophagy.
- MCPIP-expressing cells were treated with CeO 2 nanoparticles that can trap free radicals, NADPH inhibitor apocynin, p47/AS, ER stress inhibitor TUDC, or IRE-1 siRNA.
- Immunoblot analysis showed that MCPIP-induced expression of Beclin-1 was inhibited significantly by inhibition of oxidative stress and ER stress and knockdown of genes involved in these stresses ( FIG. 5A , right panel, and Supplementary Figure S 4 A).
- MCP-1 The role of MCP-1 in differentiation of human bone marrow monocytes to OC precursors has been discovered for the first time herein. Furthermore, it has also been discovered herein that this process is mediated via the induction of MCPIP. MCP-1 induces the differentiation of monocytic cells into TRAP and CTSK-positive cells that do not express other OC functional markers such as aV integrin, (33 integrin, and MMP9 and do not exhibit bone resorption (Kim et al., 2006b). The differentiation into functional OCs requires RANKL in addition to MCP-1. Thus, the MCP-1 induced differentiation yields what might be considered osteoclastogenic cells or OC precursors.
- MCPIP mediates MCP-1-induced adipogenesis (Younce et al., 2009), glial differentiation of neuroprogenitor cells (Vrotsos et al., 2009), and angiogenesis (Niu et al., 2008).
- forced expression of MCPIP resulted in high expression NADPH oxidase subunit p47 PHOX and an increased level of membrane-associated p47 PHOX , causing ROS production. It is discovered herein that this oxidative stress causes ER stress that leads to autophagy involved in OC differentiation.
- ROS are associated with multiple cellular functions such as cell proliferation, differentiation, and apoptosis (Wolf, 2005).
- the present results demonstrate that MCP-1 treatment and forced expression of MCPIP induce ROS generation during MCPIP-induced OC precursor differentiation.
- CeO 2 nanoparticles function as a free radical scavenger (Niu et al., 2007; Tsai et al., 2007; Younce and Kolattukudy, 2010). It has also been identified herein that CeO 2 inhibits the MCP-1- or MCPIP-induced ROS production and expression of OC-related genes.
- NADPH oxidase is considered the most important source of ROS by respiratory burst in a monocyte/macrophage system (Decoursey and Ligeti, 2005; Bedard and Krause, 2007).
- NAD(P)H oxidase is a multiple subunit enzyme complex. Assembly of transmembrane subunits and cytosolic subunits of enzyme complex is the first important step for its activation. In this step, p47 PHOX is the most important component which is phosphorylated, translocated from cytoplasm to the membrane to interact with gp91 PHOX (Decoursey and Ligeti, 2005; Bedard and Krause 2007; Leto et al., 2009).
- MCP-1 treatment or forced expression of MCPIP resulted in expression of p47 PHOX and increased the membrane-associated p47 PHOX level
- knockdown of MCPIP in MCP-1-treated cells decreased the expression and translocation of p47 PHOX .
- NADPH oxidase activity inhibitor apocynin and knockdown of p47 PHOX with antisense oligonucleotides inhibited membrane translocation of p47 PHOX , ROS production, and expression of OC-related genes CTSK and TRAP.
- MCPIP increases ROS production and induces expression of OC-related genes by increasing expression and translocation of p47 PHOX .
- MCPIP may serve as a novel regulator for several genes at the transcriptional and post-transcriptional level because of its DNA and RNA binding property. Therefore, MCPIP may regulate directly the expression of p47 PHOX as a transcriptional factor. Secondly, it has been reported that MCPIP activates MAPK signal pathway (Younce and Kolattukudy, 2010). Activation of MAPK may be an important reason for the MCPIP-induced expression and translocation of p47 PHOX oxidase.
- ER stress results from the accumulation of misfolded proteins which leads to the induction of the unfolded protein response (UPR) (Malhotra and Kaufman, 2007). ROS production is known to cause proteins to aggregate and misfold.
- UPR unfolded protein response
- MCP-1 treatment and forced expression of MCPIP induced ER stress via generation of ROS in the monocytes during induction of OC differentiation.
- the important role of ER stress in monocyte differentiation into OC precursors was demonstrated by the findings that inhibition of ER stress inhibited differentiation and known ER stress inducers caused differentiation.
- ER stress inhibitor TUDC and knockdown of IRE-1 showed that inhibition of ER stress leads to inhibition of MCPIP-induced OC precursor differentiation.
- thapsgargin and tunicamycin that are known important inducers of UPR/ER stress induce differentiation of monocytes into OC precursors without MCP-1 or any other inducers.
- Thapsgargin an inhibitor of ER-specific Ca-ATPase, has previously been shown to induce OC differentiation from RAW264.7 macrophage cells and mouse bone marrow cells (Takami et al., 1997; Yip et al., 2005).
- Autophagy is generally thought of as a survival mechanism, although its dysregulation has been linked to non-apoptotic cell death (Wang, 2008; Glick et al., 2010). Since differentiation involves disappearance of one set of proteins and appearance of a new set of proteins, a self-digestion process such as autophagy could be involved in this process. In fact, autophagy has been reported to be an important event for differentiation of the chronic myelogenous leukemia K562 cells (Colosetti et al., 2009), adipocytes (Malhotra and Kaufman, 2007; Singh et al., 2009; Goldman et al., 2010), paneth cells (Stappenbeck, 2010), and neuronal differentiation (Zeng and Zhou, 2008).
- Beclin-1 is a critical component in the class III PI3K complex (PI3KC3) that induces the formation of autophagosomes in mammalian systems (Wang, 2008). It has been demonstrated that Beclin-1 bridges autophagy and differentiation, and the process of autophagy and differentiation requires up-regulation of Beclin-1 (Wang, 2008). Inhibition of differentiation by PI3K inhibitors and knockdown of Beclin-1 and Atg7 strongly suggest that MCPIP induced OC precursor differentiation via autophagy. Emerging data now indicate that ER stress is a potent inducer of autophagy (Sakaki and Kaufman, 2008). Herein, the inventors have discovered that MCPIP induces OC precursor differentiation via autophagy which depends on MCPIP-induced ROS production and ER stress.
- ER stress can activate p38 MAPK signal pathway to induce autophagy (Kim et al., 2008). Moreover, ER stress activates phosphorylation of protein kinase-like ERK (PERK), an ER-localized transmembrane protein, and induction of IRE-1 is necessary for radiation-induced autophagy in mouse embryonic fibroblasts (Kim et al., 2010). Activation of NF- ⁇ B and MAPK signal pathway is necessary for OC differentiation (Huang et al., 2006).
- PERK protein kinase-like ERK
- MCPIP inhibits activation of NF- ⁇ B induced by IL- ⁇ (Skalniak et al., 2009) and lipopolysaccharide stimulation (Liang et al., 2008) while over-expression of MCPIP can cause activation of JNK and p38 (Younce and Kolattukudy, 2010).
- activation of p38 instead of NF- ⁇ B signaling is probably involved in MCPIP-mediated differentiation of monocytes into OC precursors.
- the overall pathway involved in the MCP-1/MCPIP-mediated OC precursor differentiation is shown in Supplementary Scheme 51.
- Inflammatory bone erosion is involved in many pathological conditions (Lu et al., 2007; Ha et al., 2010).
- the novel inventive features recited herein provide a new insight into the mechanism by which MCP-1 induces differentiation of monocytic cells into TRAP- and CTSK-expressing cells that can proceed to differentiate into functional OCs in the presence of RANKL, and suggest that MCPIP is a novel target for therapy of inflammatory bone erosion.
- Human BMCs were from Stemcell Technologies. ⁇ -MEM, FBS, HBSS, trypsin, recombinant human M-CSF (300-25), human MCP-1, and Trizol reagent were purchased from Invitrogen. Anti- ⁇ -actin, CTSK monoclonal antibodies, CeO 2 nanoparticles, apocynin, TUDC, 3′-MA, and LY294002 were from Sigma-Aldrich. Anti-TRAP, p47 PHOX , Fas, IRE-1, GRP78, Beclin-1, LC3 polyclonal antibodies, goat anti-rabbit and mouse secondary antibodies, and specific siRNA for IRE-1 and Beclin-1 were purchased from Santa Cruz Biotechnology. Specific siRNA for MCPIP and negative control siRNA were obtained from Ambion. Anti-MCPIP polyclonal antibody was prepared as indicated before (Zhou et al., 2006; Younce and Kolattukudy, 2010).
- BMCs were cultured in ⁇ -MEM supplemented with 10% FBS containing 30 ng/ml M-CSF, 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin in 5% CO 2 at 37° C.
- OC precursor cells were induced after 3-day culture as Ha et al. (2010). At this point, cells were treated with 50 ng/ml MCP-1, inhibitors, or gene transfection.
- CeO 2 (10 ⁇ M), apocynin (100 ⁇ g/ml), TUDC (100 ⁇ M), 3′-MA (50 ⁇ M), and LY294002 (20 ⁇ M) were added 6 h before gene transfection.
- OC precursor cells were transfected with 1 ⁇ g GFP or MCPIP-GFP eukaryotic expression plasmids for gene-gain-function assay by using Fugene 6.
- chemically synthesized siRNA duplex 100 nM targeting MCPIP, IRE-1, or Beclin-1 was transfected into OC precursor cells using DharmaFECT (Dharmacon) for 24 h prior to transfection with MCPIP-GFP or GFP plasmid.
- DharmaFECT Dharmacon
- a scrambled siRNA was used as a negative control.
- oligonucleotides For knockdown expression of p47 PHOX , specific antisense (AS) (5′-CCAGCAGGGCGATGTGACGGATGAA-3′) and sense (5′-ATGGGGGACACCTTCATCCGTCAC-3′) oligonucleotides were designed and synthesized by phosphorothioate modification by Integrated DNA Technologies. The oligonucleotides were transfected into OC precursor cells using Lipofectamine 2000 for 24 h before MCPIP—GFP plasmid transfection.
- AS specific antisense
- sense 5′-ATGGGGGACACCTTCATCCGTCAC-3′
- TRAP histological staining for TRAP as described previously (Kim et al., 2006a). Briefly, following fixation, cells were stained with freshly prepared TRAP staining solution (naphthol AS-MX phosphate, fast red violet LB salt, and potassium sodium tartrate). On each coverslip, totally at least 500 cells were examined and the TRAP-positive cells were counted in 3-5 fields (20 ⁇ objective), and the percentage of TRAP-positive cells was calculated.
- Oxidant production in OC precursor cells was assessed by measuring the oxidation of intracellular DHR123 (Molecular Probes) as described previously (Younce et al., 2010). ROS production was expressed as folds compared with control cells expressing GFP alone.
- OC precursor cells were harvested, sonicated on ice in a buffer containing 100 mM KCl, 3 mM NaCl, 3.5 mM MgCl 2 , 10 mM HEPES, 1 mM EGTA, 10 ⁇ g/ml pepstatin, 10 ⁇ g/ml leupeptin, and 0.5 mM PMSF; lysates were centrifuged at 600 g for 10 min at 4° C. to remove nuclei and unbroken cells. The supernatant was then ultracentrifuged at 100000 g for 1 h at 4° C. Membranes were washed in the same buffer, quantified (Lemarie), and resuspended in Laemmli sample buffer, before western blot analysis.
- SDS-PAGE Sodium dodecyl sulfate-polyacrylamide gel electrophoresis
- Samples (30 ⁇ g of protein) were subjected to 12% SDS-PAGE for most of target protein and 18% SDS-PAGE for LC3-II and LC3, transferred onto PVDF membranes (Millipore), and assayed for MCPIP, p47 PHOX , TRAP, CTSK, GRP78, IRE-1, Beclin-1, LC3-II, and ⁇ -actin or Fas (loading control) protein expression by chemiluminescence detection (Pierce ECL kit) according to the manufacturer's instructions.
- the specific protein bands were quantified by densitometric analysis with GS-690 Image Densitometer (Bio-Rad).
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Biomedical Technology (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicinal Chemistry (AREA)
- Immunology (AREA)
- Biochemistry (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Biophysics (AREA)
- Cell Biology (AREA)
- Developmental Biology & Embryology (AREA)
- Epidemiology (AREA)
- Physics & Mathematics (AREA)
- Microbiology (AREA)
- Plant Pathology (AREA)
- Physical Education & Sports Medicine (AREA)
- Rheumatology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Virology (AREA)
- Hematology (AREA)
- Endocrinology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Pain & Pain Management (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Disclosed herein are methods of treating a patient at risk of developing an inflammatory joint disease. In exemplary embodiments, the method involves inhibiting MCPIP levels in a patient in need, wherein said patient in need is exhibiting pre-arthritic or pre-osteoporotic symptoms.
Description
- The invention relates to cellular factors for modulating cellular pathways. More particularly, the invention relates to cellular transcription factors that affect inflammation and the differentiation of osteoclasts, and methods for their therapeutic use.
- There is strong evidence that osteoclast (OC) is the principal cell type responsible for bone resorption in inflammatory joint diseases (Harris, 1990; Sakiyama et al., 2001; Sato and Takayanagi, 2006; Mundy, 2007). Rheumatoid arthritis (RA) is characterized by the presence of inflammatory synovitis accompanied by the destruction of the joint cartilage and bone (Harris, 1990; Mundy, 2007; Sugimura and Li, 2010). OCs are bone-resorbing cells that differentiate from hematopoietic precursors of the monocyte/macrophage lineage (Sakiyama et al., 2001; Boyce et al., 2007). OCs are multinuclear giant cells that stain positive for tartrate-resistant acid phosphatase (TRAP) and serine protease cathepsin K (CTSK) (Kiviranta et al., 2001; Boyce et al., 2007).
- Monocyte chemotactic protein-1 (MCP-1), a CC chemokine commonly found at the site of tooth eruption, RA bone degradation, and bacterially induced bone loss (Wise et al., 2002), is known to induce differentiation of monocytes into TRAP and CTSK-positive precursors of OCs. MCP-1 is expressed by mature OCs and its expression is regulated by nuclear factor-κB (NF-κB) (Kim et al., 2005). Several reports showed that MCP-1 is induced by NF-κB ligand RANKL and promotes OC fusion into multinuclear TRAP-positive cells without bone-resorption activity (Kim et al., 2006a,b), which might be called OC precursors. Recently, it has been reported that MCP-1 plays an important role in regulating OC differentiation in an autocrine/paracrine manner under stimulation by RANKL (Miyamoto et al., 2009). How MCP-1 mediates OC differentiation remains unclear.
- The cellular effect of MCP-1 is mediated by the CCR2, a G-protein-coupled receptor that is induced by the receptor activator of RANKL (Gerszten et al., 2001; Kim et al., 2005). The signaling process initiated by MCP-1 binding to CCR2 leads to changes in gene expression.
- Recently, it was found that this MCP-1 binding leads to the induction of a novel zinc-finger protein called MCPIP in human peripheral blood monocytes (Zhou et al., 2006). The biological functions of MCPIP, however, remain poorly understood.
- The importance of the way in which MCP-1 mediates OC differentiation has been realized, which has been heretofore unclear. The role of MCPIP in biological processes initiated by MCP-1 has also now been discovered. It was previously understood that MCPIP mediates several biological processes initiated by MCP-1. including cardiomyocyte death (Younce and Kolattukudy, 2010; Younce et al., 2010), adipogenesis (Younce et al., 2009), angiogenesis (Niu et al., 2008), and glial differentiation of neuroprogenitor cells (Vrotsos et al., 2009). It has been discovered, as disclosed herein, that MCP-1 induces differentiation of monocytic cells into OC precursors via MCPIP. It is therefore presented that MCPIP mediates differentiation of OC precursors via induction of oxidative stress that causes endoplasmic reticulum (ER) stress that leads to autophagy involved in osteoclastogenesis. These findings implicate that MCPIP is a novel factor involved in OC precursor differentiation, and thus it serves as a new target for diagnosis and treatment of osteoporosis-related disease.
- Figure Legends
-
FIG. 1 . MCP-1 induction of OC precursor differentiation, mediated by MCPIP. (A and B) BMCs were transfected with FLAG-tagged MCPIP (WT-MCPIP) or FLAG-tagged empty vector (MAT-FLAG). TRAP-expressing cells were stained and viewed under the Nikon microscope (A). BMNCs treated with MCP-1 alone or with MCPIP siRNA or Scr siRNA. TRAP-expressing cells were stained and TRAP-positive cell proportion was measured (B). At least three fields (˜500 cells), were chosen. *P<0.05 and **P<0.01 versus control; #P<0.05 versus Scr. (C) Immunoblotting showing the expression of TRAP, CTSK, and MCPIP induced by MCP-1. Data were mean±SD (n=3). *P<0.05 versus control; #P<0.05 versus Scr. (D) Real-time PCR showing the transcription of TRAP, CTSK, and MCPIP induced by MCP-1. *P<0.05 versus control; #P<0.05 versus Scr. (E) Real-time PCR showing the transcription of αV integrin, β3 integrin, and MMP9. -
FIG. 2 . MCPIP overexpression induces OC-related marker TRAP and CTSK expression. (A) Real-time PCR showing the transcription of TRAP, CTSK, and MCPIP (upper panel) and αV integrin, β3 integrin, and MMP9 (lower panel) induced by MCPIP transfection. *P<0.05 and **P<0.01 versus control or GFP; #P<0.05 versusday -
FIG. 3 . ROS production involvement in MCPIP/MCP-1-induced OC precursor differentiation. (A) Western blot showed that MCP-1 treatment or MCPIP transfection induces p47PHOX expression. *P<0.05 versus control or GFP; #P<0.05 versus Scr. (B) Immunoblotting showed that MCP-1 or MCPIP overexpression induces an increase in cytoplasmic membrane-associated p47PHOX. * P<0.05 versus control or GFP; #P<0.05 versus Scr. (C)ROS production induced by MCP-1 or MCPIP transfection, or siRNA was detected by using DHR123. *P<0.05 versus control or GFP; #P<0.05 versus Scr. (D) MCPIP-induced ROS production was inhibited by ROS, NAD(P)H oxidase inhibitors, and p47PHOX knockdown. *P<0.05 versus GFP, #P<0.05 versus MCPIP only. *P<0.05 compared with p47PHOX non-specific (NS) oligonucleotides transfection cells. (E) The effect of CeO2 on MCPIP, TRAP, and CTSK expression induced by MCPIP transfection. Data were mean±SD (n=3). *P<0.05 versus GFP; #P<0.05 versus MCPIP only. (F) The effect of p47PHOX AS and apocynin on the expression of related proteins. m-p47, membrane located p47PHOX; p47, total p47PHOX; p47/NS, non-specific RNA; p47/AS, antisense RNA. Apo, apocynin. -
FIG. 4 . MCPIP induction of ER stress via ROS production involved in OC precursor differentiation. (A) Immunoblotting shown that MCP-1 or MCPIP transfection induces the expression of GRP78 and IRE-1 (ER stress markers) by inducing ROS production. (B) Blockage of ER stress by IRE-1 siRNA abolished MCPIP-induced mRNA and protein expression of TRAP and CTSK. *P<0.05 versus GFP, #P<0.05 versus Scr. (C) Blockage of ER stress by TUDC abolished MCPIP-induced mRNA and protein expression of TRAP and CTSK. *P<0.05 versus GFP, #P<0.05 versus MCPIP alone. (D) Real-time PCR showing the expression of TRAP, CTSK, GRP78, and IRE-1 by ER stress inducer. *P<0.05 versus control. (E) Immunoblotting showing the expression of TRAP, CTSK, GRP78, and IRE-1 by ER stress inducer. TU, tunicamycin; TH, thapsigargin. -
FIG. 5 . MCPIP induction of autophagy via ROS production and ER stress involved in OC precursor differentiation. (A) Immunoblotting shown that MCPIP induces autophagy characterized with the marker Beclin-1 expression and ROS/ER stress inhibitor inhibited MCPIP-induced expression of Beclin-1. (B and C) Immunoblotting shown thatautophagy blocker 3′-MA and LY294002 blocked MCPIP-induced OC-related gene TRAP and CTSK expression but not GRP78. (D) Real-time PCR shown thatautophagy blocker 3′-MA and LY294002 blocked MCPIP-induced expression of TRAP and CTSK but not GRP78. *P<0.05 versus GFP, #P<0.05 versus MCPIP alone. (E) Blocking autophagy by Beclin-1 siRNA inhibited MCPIP-induced mRNA and protein expression of TRAP and CTSK but not GRP78. *P<0.05 versus MCPIP alone. -
FIG. 6 . MCPIP-induced OC precursor differentiation inhibited by PI3K inhibitor, wortmannin. (A) qRT-PCR showing induction of Beclin-1 and TRAP and CTSK at the transcript level by MCPIP transfection, and the inhibition effect of wortmannin on MCPIP. *P<0.05 versus MCPIP alone. (B) Immunoblotting show that MCPIP induced Beclin-1, and TRAP and CTSK, and that their induction was suppressed by wortmannin. (C) The intensities of immunoblots were measured and normalized to (3-actin of the corresponding group. *P<0.05 compared with MCPIP alone. - Supplementary Figure S1. MCPIP induced production of cellular ROS in bone marrow mononuclear cells (BBMN). Cells were transfected with empty vector (MAT-FLAG) or MCPIP-expression vector (WT-MCPIP) and after 24 hr stained with DHR123 that is a cell permeable nonfluorescent reagent that generates a fluorescent product (red) when oxidized by cellular ROS.
- Supplementary Figure S2. ROS production involvement in MCPIP/MCP-1 induced OC precursor differentiation. Bone marrow monocytes were pretreated with CeO2 or apocynin for 6 h or p47PHOXantisense oligonucleotides for 24 h and then cells were transfected with MCPIP or GFP for 4 days. Cell lysate was collected and analysed using immunoblot with appropriate antibody and results were quantified against (3-actin. A, Effect of CeO2 on MCPIP-induced TRAP and CTSK expression [* P<0.05 compared to CeO2-untreated cells (“None”)]. B, Effect of p47 PHOX antisense oligonucleotides on expression of p47 PHOX and its translocation from cytoplasm to membrane [* P<0.05 compared to non-sense oligonucleotides treated cells (“p47/NS”)]. C, Effect of p47 PHOX antisense oligonucleotides on MCPIP-induced TRAP and CTSK expression [* P<0.05 compared to non-sense oligonucleotides treated cells (“p47/NS”)]. D, Effect of apocynin on expression of p47PHOX and its translocation from cytoplasm to membrane [* P<0.05 compared to apocynin-untreated cells (“None”)]. E, Effect of apocynin on MCPIPinduced TRAP and CTSK expression [* P<0.05 compared to apoicynin-untreated cells (“None”)].
- Supplementary Figure S3. MCPIP induces ER stress via ROS production which involvement in OC precursor differentiation. After treatment, cell lysate was collected and analysed using immunoblot with appropriate antibody and results were quantified against (3-actin. A, MCP-1 induces ER stress marker GRP78 and IRE-1 expression via MCPIP (* P<0.05). B, Effect of p47PHOX antisense oligonucleotides, CeO2 and apocynin on MCPIP-induced expression of GRP78 and IRE-1 (* P<0.05). C, inhibition of ER stress by using knock-down of IRE-1 attenuates MCPIP-induced expression of TRAP and CTSK (* P<0.05). D, pretreatment of TUDC inhibits MCPIP-induced expression of TRAP and CTSK and ER stress marker IRE-1 and GRP78 (* P<0.05). E, ER stress inducer tunicamycin and thapsigargin induce expression of TRAP and CTSK (* P<0.05).
- Supplementary Figure S4. MCPIP induces autophagy via ROS production and ER stress which involvement in OC precursor differentiation. After treatment, cell lysate was collected and analysed using immunoblot with appropriate antibody and results were quantified against (3-actin. A, MCPIP induces autophagy characterized with the marker beclin-1 expression and ROS inhibitor or ER stress inhibitor inhibited MCPIP-induced expression of beclin-1. (* P<0.05). B,
autophagy blocker 3′-MA blocked MCPIP-induced osteoclast-related gene TRAP and CTSK expression but not GRP78 (* P<0.05). C, autophagy blocker LY-294002 blocked MCPIP induced osteoclast-related gene TRAP and CTSK expression but not GRP78. (* P<0.05). D, blockage of autophagy by beclin-1 specific siRNA inhibited MCPIP-induced osteoclast-related gene TRAP and CTSK expression but not GRP78 both in protein and mRNA level. * P<0.05. - Supplementary Figure S5 MCPIP—induced autophagy marker Atg7 is necessary for induction of OC precursor differentiation. (A) qRT-PCR shows that transfection of MCPIP expression vector resulted in significantly increased mRNA levels of Atg7, Beclin-1, TRAP, and CTSK. Atg7-specific siRNA abolished MCPIP-induced expression of these genes. * P<0.05 compared with MCPIP alone. (B) Immunoblot shows that transfection with MCPIP expression vector caused induction of beclin-1, TRAP, and CTSK at the protein levels. Upregulation of these marker genes were inhibited by Atg7-specific siRNA but were not affected by scrambled siRNA. (C) The intensity of each protein was measured and normalized to (3-actin of the corresponding group. * P<0.05 compared with MCPIP alone.
- The inventors previously identified the novel transcription factor designated as MCPIP (MCP-1-induced protein). MCPIP was initially isolated from human monocytes after stimulation with MCP-1. The nucleotide (SEQ ID NO: 1) and amino acid (SEQ ID NO: 2) sequences of isolated human MCPIP were deposited with GenBank under accession number AY920403 and the nucleotide (SEQ ID NO: 3) and amino acid (SEQ ID NO: 4) sequences of isolated mouse MCPIP were deposited with GenBank under accession number AY920404.
- The inventors have continued to study the biological relevance of these genes/proteins, and to develop new therapies based on this research. This invention is based on the discovery that MCP-1 induces MCPIP in human peripheral blood monocytes which induces differentiation of monocytes into osteoclast cells (OC) or OC precursors. NFKB is a master controller of inflammation in the body. Agonists of MCPIP would enhance its anti-inflammatory potentcy; it is herein discovered that inhibition of the activation of NFKB by MCPIP provides a significant potential as an anti-inflammatory agent. Therefore, inhibition of the activation of NFKB by inhibiting MCPIP has a significant use as an anti-inflammatory agent. Inhibition of MCPIP inhibits inflammation, which plays a critical role in diseases including angiogenesis, i.e., the physiological process involving the growth of new blood vessels from pre-existing vessels, adipogenesis, the process of cell differentiation by which preadipocytes become adipocytes, and osteoclastogenesis, i.e., the development of osteoclasts. Furthermore, MCPIP plays a critical role in the differentiation of osteoclasts involved in inflammatory bone loss that occurs in many inflammatory diseases such as, for example, rheumatoid arthritis. Additionally, elevated MCP-1 levels have been identified as a direct cause of insulin resistance. MCPIP has been discovered to mediate insulin resistance, and thus, MCPIP inhibitors enhance insulin sensitivity. Additional research has led to the identification of MCPIP as a novel factor involved in OC precursor differentiation, as provided herein, identifies MCPIP as a new target for diagnosis and treatment of osteoporosis-related disease.
- Discovered herein is the effect of inhibition of MCPIP in reducing osteoclast precursor differentiation. MCPIP plays a critical role in differentiation of osteoclasts involved in inflammatory bone loss that occurs in many inflammatory diseases such as rheumatoid arthritis. It is disclosed herein that differentiation of monocytes to OC precursors is mediated by MCPIP. Therefore, the blocking or inhibition of MCPIP provides a promising therapeutic strategy for preventing differentiation of monocytes to OC precursors, and ultimately the prevention of bone resorption. Bone resorption is a process by which osteoclasts break down bone and release minerals, resulting in a transfer of calcium from bone fluid to the blood. This discovery provides a particular benefit in inflammatory joint diseases.
- Ultimately, inflammatory bone erosion is involved in many pathological conditions (Lu et al., 2007; Ha et al., 2010). The novel inventive features described and contemplated herein provide a new insight into the mechanism by which MCP-1 induces differentiation of monocytic cells into TRAP- and CTSK-expressing cells that can proceed to differentiate into functional OCs in the presence of RANKL, and demonstrate that MCPIP is a novel target for therapy of inflammatory bone erosion, in one exemplary embodiment.
- According to one embodiment of the invention, a method of treating a condition in a patient in need includes administering to the patient a therapeutically effective amount of a composition that inhibits the expression or action of MCPIP. The patient in need may be exhibiting pre-arthritic symptoms including but not limited to pain or tenderness in a joint which is aggravated by movement or activity, inflammation indicated by joint swelling, stiffness, redness, and/or warmth, joint deformity, loss of range of motion or flexibility in a joint, or unexplained weight loss. Additional pre-arthritic symptoms include extreme fatigue, lack of energy, weakness or a feeling of malaise, a non-specific fever, or crepitus, i.e., creaky, popping or snapping joints.
- In another embodiment, the patient in need may be exhibiting pre-osteoporotic symptoms, including but not limited to back pain caused by a fractured or collapsed vertebra, loss of height over time, a stooped posture, or a bone fracture that occurs much more easily than expected. The patient in need may also be exhibiting symptoms of an existing arthritic or osteoporotic disease, or both. Furthermore, the patient in need may be exhibiting symptoms of any arthritic or osteoporotic related-diseases, any inflammatory joint diseases, or any pre-disease symptoms of these related diseases. In a further embodiment, the condition may include an osteoporosis-related condition, and in a particular embodiment, rheumatoid arthritis.
- As used herein, therapeutically effective amount refers to an amount sufficient to elicit the desired biological response. In the present invention the desired biological response can be an overall improvement in the condition being treated. The overall improvement can be associated with improvement in individual symptoms.
- Subject or patient, as used herein, refers to animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, pigs, dogs, cats, rabbits, guinea pigs, rats, mice or other bovine, ovine, equine, canine, feline, rodent or murine species. In one embodiment, the subject is a human.
- The compounds for use in the method of the invention can be formulated for oral, transdermal, sublingual, buccal, parenteral, rectal, intranasal, intrabronchial, intrapulmonary, or ocular administration. Oral administration is preferred. For oral administration, the compounds can be of the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., polyvinylpyrrolidone, hydroxypropylcellulose or hydroxypropylmethylcellulose); fillers (e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrates (e.g., sodium starch glycollate); or wetting agents (e.g., sodium lauryl sulphate). If desired, the tablets can be coated using suitable methods and coating materials such as OPADRY film coating systems available from Colorcon, West Point, Pa. (e.g., OPADRY, OY Type, OY—C Type, Organic Enteric OY—P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRY™ White, 32K18400).
- In a particular embodiment, the oral form is a tablet containing MCPIP and a pharmaceutically acceptable excipient, such as, but not limited to mannitol, corn starch, microcrystalline cellulose, colloidal silicon dioxide, polyvinyl pyrrolidone, talc, magnesium stearate, and the like which are optionally coated with an OPADRY film coating.
- Liquid preparation for oral administration can be in the form of solutions, syrups or suspensions. The liquid preparations can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agent (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl p-hydroxy benzoates or sorbic acid).
- For buccal administration, the compounds for use in the method of the invention can be in the form of tablets or lozenges formulated in a conventional manner.
- For parenteral administration, the compounds for use in the method of the invention can be formulated for injection or infusion, for example, intravenous, intramuscular or subcutaneous injection or infusion, or for administration in a bolus dose and/or continuous infusion. Suspensions, solutions or emulsions in an oily or aqueous vehicle, optionally containing other formulatory agents such as suspending, stabilizing and/or dispersing agents can be used.
- For rectal administration, the compounds for use in the method of the invention can be in the form of suppositories or enemas. For sublingual administration, tablets can be formulated in conventional manner.
- For intranasal, intrabronchial or intrapulmonary administration, conventional formulations can be employed.
- Further, the compounds (e.g. protein or delivery vehicle) for use in the method of the invention can be formulated in a sustained release preparation. For example, the compounds can be formulated with a suitable polymer or hydrophobic material which provides sustained and/or controlled release properties to the active agent compound. As such, the compounds for use the method of the invention can be administered in the form of microparticles for example, by injection or in the form of wafers or discs by implantation.
- In accordance with the method of the invention, an expression vector is a viral or a non-viral expression vector. Viral expression vectors which may be used advantageously in the method of the invention include, but are not limited to, an adeno associated virus (AAV) vector, a lentivirus vector, an adenovirus vector, and a herpes simplex virus (HSV) vector.
- In additional embodiments, the composition comprises siRNA or miRNA specific for MCPIP, an antisense nucleotide specific for MCPIP, and/or shRNA. In an alternative embodiment, the composition comprises an antibody specific to MCPIP.
- In another embodiment, a method of inhibiting osteoclast production (or a method of reducing osteoclast precursor cells) in a patient in need is provided. The method includes administering a therapeutically effective amount of a composition that inhibits the expression or action of MCPIP in the patient. The patient in need, in an embodiment, may be exhibiting symptoms of rheumatoid arthritis, osteoarthritis, and/or osteoporosis.
- In another embodiment, administering a therapeutically effective amount of a composition includes a composition comprising: a composition that inhibits the expression or action of MCPIP, and a pharmaceutically acceptable excipient.
- In further embodiments, the composition includes an MCPIP siRNA, an shRNA, an antibody specific to MCPIP, and/or an antisense nucleotide specific for MCPIP.
- Many of the embodiments of the subject invention make reference to particular methods of inhibiting expression. The subject invention is not to be limited to any of the particular methods described. One such method includes siRNA (small interfering/short interfering/silencing RNA). SiRNA most often is involved in the RNA interference pathway where it interferes with the expression of a specific gene. In addition to its role in the RNA interference pathway, siRNA also act in RNA interference-related pathways, e.g., as an antiviral mechanism or in shaping the chromatin structure of a genome.
- Another method by which to inhibit expression and to inhibit the expression of MCPIP in particular is shRNA. ShRNA (short hairpin or small hairpin RNA) refers to a sequence of RNA that makes a tight hairpin turn and is used to silence gene expression via RNA interference. It uses a vector introduced into cells and a U6 or H1 promoter to ensure that the shRNA is always expressed. The shRNA hairpin structure is cleaved by cellular machinery into siRNA which is then bound to the RNA-induced silencing complex. This complex binds to and cleaves mRNAs which match the siRNA that is bound to it.
- MCPIP can also be blocked by subjecting procured cells to an antibody specific to MCP-1. An antisense nucleotide may also be used to block or inhibit expression, in particular, the expression of MCP-1. Expression may also be inhibited with the use of a morpholino oligomer or phosphorodiamidate morpholino oligomer (PMO). PMOs are an antisense technology used to block access of other molecules to specific sequences within nucleic acid. PMOs are often used as a research tool for reverse genetics, and function by knocking down gene function. This is achieved by preventing cells from making a targeted protein or by modifying splicing of pre-mRNA.
- It has been demonstrated that MCP-1 induces TRAP-positive OC precursor formation from human peripheral blood mononuclear cells (Kim et al., 2006a). Here, we found that 50 ng/ml MCP-1 induced TRAP-positive OC precursor cell formation from human bone marrow mononuclear cells (BMCs) (
FIG. 1B ). Immunoblotting (FIG. 1C ) and real-time polymerase chain reaction (PCR) (FIG. 1D ) showed that MCP-1 treatment induced expression of OC markers TRAP and CTSK. However, MCP-1 did not affect the expression of the OC functional markers aV integrin, (33 integrin, and MMP9 (FIG. 1E ). We also found that 50 ng/ml MCP-1 induced up-regulation of MCPIP protein and mRNA levels, which can be suppressed by treatment with MCPIP small interfering RNA (siRNA) (FIGS. 1C and D). Compared with FLAG-tagged empty vector (MAT-FLAG), expression of FLAG-tagged MCPIP (WT-FLAG) induced TRAP-positive OC precursor cell formation (FIG. 1A ). MCPIP siRNA also significantly inhibited the formation of OC precursor cells that expressed TRAP and CTSK but showed no effects on expression of aV integrin, (33 integrin, and MMP9 (FIG. 1C-E ). These results suggest that induction of the TRAP-positive OC precursor cells by MCP-1 treatment of BMCs was mediated via MCPIP. - If MCP-1-induced differentiation of OC precursors is mediated by MCPIP, forced expression of MCPIP in monocytes might be expected to induce the formation of TRAP-positive OC precursor cells without MCP-1. BMCs were transfected to test this concept, as can be seen in
FIG. 2 , with MCPIP-GFP expression vector. Increased expression of MCPIP was found 24 h after transfection at mRNA levels as measured by real-time PCR and its expression reached the peak at 4 days after transfection (seeFIG. 2A , upper panel). The expression of TRAP and CTSK was induced at 2 days after MCPIP transfection and reached the peak at 4 days. However, the expression of aV integrin, (33 integrin, and MMP9 showed no significant changes after MCPIP transfection (seeFIG. 2A , bottom panel). Immunoblot analysis showed that MCPIP overexpression induced expression of TRAP and CTSK (seeFIG. 2B ), and TRAP staining showed that MCPIP expression significantly elevated formation of TRAP-positive cells (FIG. 2C ). When MCPIP siRNA was transfected into BMCs for 24 h prior to MCPIP transfection, MCPIP expression was knocked down and the expression of OC-related genes TRAP and CTSK also were down-regulated (FIG. 2B ). Moreover, the percentage of TRAP-positive cells was lowered by treatment with MCPIP siRNA (FIG. 2C ). These results suggest that MCPIP transfection causes induction of OC-related genes and formation of TRAP-positive OC precursor cells. - Reactive oxygen species (ROS) derived from NADPH oxidase have been suggested to regulate OC differentiation and prolong the survival of OC precursors (Yamasaki et al., 2009). p47PHOX, a regulatory subunit of NADPH oxidase, has been implicated in ROS generation (Decoursey and Ligeti, 2005). Herein, it was tested whether forced expression of MCPIP could induce expression and activation of NADPH oxidase by translocation of p47PHOX into the membrane and produce ROS. Dihydrorhodamine 123 (DHR123) staining revealed ROS production by MCPIP transfected cells but not MAT-FLAG controls (Supplementary Figure S1). Immunoblot analysis shows that MCP-1 treatment and MCPIP expression increased the expression of p47PHOX (
FIG. 3A ) and its translocation from the cytoplasm into the membrane (FIG. 3B ). ROS production caused by MCP-1 treatment and MCPIP expression was assessed by DHR123 staining and results showed that MCP-1 treatment and MCPIP transfection remarkably increased ROS generation. Moreover, the effects of MCP-1 treatment or forced MCPIP expression on the expression and translocation of p47PHOX and ROS generation were inhibited by MCPIP siRNA (FIG. 3A-C ). - To understand the involvement of ROS production in MCPIP-mediated formation of TRAP-positive OC precursor cells, BMCs were treated with CeO2 nanoparticles, an inhibitor of ROS (Tsai et al., 2007) prior to MCPIP transfection. It was found that MCPIP-induced ROS production was significantly inhibited by CeO2 (
FIG. 3D ). Immunoblot and real-time PCR analysis showed that CeO2 inhibited MCPIP-induced TRAP and CTSK expression both at protein and mRNA levels (FIG. 3E and Supplementary Figure S2A). The effect of apocynin, an inhibitor of NADPH oxidase on MCPIP-induced induction of OC-related genes TRAP and CTSK was further tested herein. It was found that apocynin suppressed MCPIP-induced ROS production (FIG. 3D ) and expression of TRAP and CTSK (FIG. 3F and Supplementary Figure S2E). Moreover, apocynin inhibited MCPIP-induced expression and membrane translocation of p47PHOX (FIG. 3F and Supplementary Figure S2D). Furthermore, knock-down of p47PHOX, by its specific antisense oligonucleotides (p47/AS) also decreased ROS production, expression and translocation of p47PHOX, and expression of OC-related genes TRAP and CTSK (FIG. 3F and Supplementary Figure S2B and C). These results suggest that MCPIP causes ROS production by up-regulating p47PHOX expression and its membrane translocation, and that ROS generation is involved in MCP-1-induced OC precursor differentiation. - It has been reported that oxidative stress can induce ER stress (Xue et al., 2005; Malhotra et al., 2008). Therefore, it was tested herein whether MCP-1 treatment induces ER stress in BMCs. Immunoblot analysis showed that MCP-1 treatment induced expression of the ER stress markers 78 kDa glucose regulated protein (GRP78) and inositol-requiring enzyme-1 (IRE-1) (
FIG. 4A , left panel, and Supplementary Figure S3A). This induction was inhibited by MCPIP siRNA (FIG. 4A , left panel, and Supplementary Figure S3A), indicating that MCP-1 induced ER stress via MCPIP. It was also found that forced expression of MCPIP induced ER stress with up-regulation of GRP78 and IRE-1 as indicated by immunoblot (FIG. 4A and Supplementary Figure S3B). To test whether MCPIP induces ER stress through oxidative stress, ROS production was suppressed by CeO2, apocynin, and p47/AS, and then immunoblotting was performed. Results showed that MCPIP-induced expression of GRP78 and IRE-1 was attenuated by CeO2, apocynin, and p47/AS (FIG. 4A and Supplementary Figure S3B). These results suggest that MCP-1 mediated ER stress via MCPIP-induced ROS production. - To understand whether ER stress is involved in MCPIP-induced expression of OC-related genes TRAP and CTSK, MCPIP-induced ER stress was inhibited by IRE-1 siRNA (
FIG. 4B and Supplementary Figure S3C) and the ER stress-specific inhibitor tauroursodeoxycholate (TUDC) (FIG. 4C and Supplementary Figure S3D). Immunoblot and real-time PCR analysis showed that MCPIP-induced expression of TRAP and CTSK was significantly inhibited by IRE-1 siRNA and by TUDC at both protein and mRNA levels but did not affect the expression of MCPIP (FIGS. 4B and C). - If ER stress is critically important for OC differentiation, the inventors identified that ER stress inducers might induce differentiation of OC precursors without other inducers. In fact, two ER stress inducers, tunicamycin (TU) and thapsigargin (TH), induced expression of GRP78, IRE-1, TRAP, and CTSK at both protein and mRNA levels (
FIGS. 4D and E and Supplementary Figure S3E). This result discovered by the inventors suggests that induction of ER stress alone could induce OC precursor differentiation. These results strongly support the conclusion that MCPIP-induced ER stress is involved in MCP-1-mediated OC precursor differentiation. - Increased expression of Beclin-1 is a commonly used marker of autophagy (Wang, 2008). Autophagy has been implicated in differentiation in some cellular contexts (Baerga et al., 2009; Singh et al., 2009). However, it was heretofore unclear whether autophagy has involvement in OC differentiation. Here it was found that forced expression of MCPIP increased expression of Beclin-1; this effect was suppressed by MCPIP siRNA, but not by non-specific scramble (Scr) siRNA (
FIG. 5A , left panel and Supplementary Figure S4A). This result revealed that MCPIP induced autophagy in BMCs during differentiation into OC precursor cells. - ER stress is known to induce autophagy. To test whether MCPIP-induced ROS production and ER stress are involved directly in MCPIP-mediated autophagy, MCPIP-expressing cells were treated with CeO2 nanoparticles that can trap free radicals, NADPH inhibitor apocynin, p47/AS, ER stress inhibitor TUDC, or IRE-1 siRNA. Immunoblot analysis showed that MCPIP-induced expression of Beclin-1 was inhibited significantly by inhibition of oxidative stress and ER stress and knockdown of genes involved in these stresses (
FIG. 5A , right panel, and Supplementary Figure S4A). These results suggested that MCPIP-mediated autophagy was caused by oxidative stress and ER stress during differentiation of BMCs into OC precursor cells. - Recently, it has been shown that the PI3K inhibitors LY294002 and 3′-methyladenine (MA) stop the macroautophagic pathway at the sequestration step in rat hepatocytes (Blommaart et al., 1997; Petiot et al., 2000). In order to investigate whether autophagy is involved in MCPIP-mediated OC precursor differentiation, the effect of LY294002 and 3′-MA on expression of autophagy marker Beclin-1, lipidation of LC3 and expression of OC-related genes, TRAP and CTSK was tested. Real-time PCR analysis showed that 3′-MA and LY294002 significantly inhibited MCPIP-induced expression of Beclin-1, TRAP, and CTSK (
FIG. 5D ). Immunoblot assay demonstrated that 3′-MA and LY294002 inhibited the expression of Beclin-1, TRAP, and CTSK and lipidation of LC-3 (FIGS. 5B and C and Supplementary Figure S4B and C). However, no effect on expression of GRP78 induced by MCPIP was found revealing that inhibition of autophagy does not affect ER stress that is proposed to cause autophagy (FIGS. 5B and C and Supplementary Figure S4B and C). Furthermore, upon inhibition of autophagy by knockdown of Beclin-1 with specific siRNA, MCPIP-induced expression of OC-related markers TRAP and CTSK was markedly suppressed, but scrambled siRNA showed little effects (FIG. 5E and Supplementary Figure S4D). The chemical inhibitors of autophagy and knockdown of Beclin-1 did not affect MCPIP-induced expression of GRP78, a marker of ER stress, which father leads to autophagy. These results strongly suggested that OC precursor cell differentiation induced by MCPIP expression is mediated via induction of ROS production that causes ER stress, which further leads to autophagy. - To further confirm the involvement of autophagy in OC precursor differentiation, a selective inhibitor of PI3K, wortmannin, was tested (Blommaart et al., 2007) on MCPIP-induced differentiation. Wortmannin severely inhibited MCPIP-induced OC precursor differentiation as indicated by the expression of OC markers at mRNA level by quantitative real-time PCR (qRT-PCR) and protein level by immunoblot analysis (
FIG. 6A-C ); inhibition of autophagy was reflected by changes in the Beclin-1 levels. In support of the involvement of autophagy in OC precursor differentiation, knockdown of Atg7 by specific siRNA severely inhibited expression of the OC markers TRAP and CTSK both at mRNA and protein level as measured by qRT-PCR and immunoblot analysis, respectively (Supplementary Figure S5B and C), whereas scrambled siRNA did not significantly affect induction of these proteins. - The role of MCP-1 in differentiation of human bone marrow monocytes to OC precursors has been discovered for the first time herein. Furthermore, it has also been discovered herein that this process is mediated via the induction of MCPIP. MCP-1 induces the differentiation of monocytic cells into TRAP and CTSK-positive cells that do not express other OC functional markers such as aV integrin, (33 integrin, and MMP9 and do not exhibit bone resorption (Kim et al., 2006b). The differentiation into functional OCs requires RANKL in addition to MCP-1. Thus, the MCP-1 induced differentiation yields what might be considered osteoclastogenic cells or OC precursors.
- It has been demonstrated that MCPIP mediates MCP-1-induced adipogenesis (Younce et al., 2009), glial differentiation of neuroprogenitor cells (Vrotsos et al., 2009), and angiogenesis (Niu et al., 2008). Here, it was found that forced expression of MCPIP resulted in high expression NADPH oxidase subunit p47PHOX and an increased level of membrane-associated p47PHOX, causing ROS production. It is discovered herein that this oxidative stress causes ER stress that leads to autophagy involved in OC differentiation. The involvement of this sequence of processes in OC precursor differentiation is supported by the finding that inhibition of p47PHOX expression, NADPH oxidase activity, ROS production, ER stress, or autophagy by chemical inhibitors or by gene knockdown markedly suppressed MCPIP-induced expression of OC-related genes, TRAP and CTSK.
- ROS are associated with multiple cellular functions such as cell proliferation, differentiation, and apoptosis (Wolf, 2005). Many reports revealed that high level of intracellular ROS also contributes to angiogenesis (Xia et al., 2007), epithelial-mesenchymal transition (Zhang et al., 2009), survival, and differentiation of OCs (Steinbeck et al., 1998; Yamasaki et al., 2009). The present results demonstrate that MCP-1 treatment and forced expression of MCPIP induce ROS generation during MCPIP-induced OC precursor differentiation. It has been reported that CeO2 nanoparticles function as a free radical scavenger (Niu et al., 2007; Tsai et al., 2007; Younce and Kolattukudy, 2010). It has also been identified herein that CeO2 inhibits the MCP-1- or MCPIP-induced ROS production and expression of OC-related genes.
- NADPH oxidase is considered the most important source of ROS by respiratory burst in a monocyte/macrophage system (Decoursey and Ligeti, 2005; Bedard and Krause, 2007). NAD(P)H oxidase is a multiple subunit enzyme complex. Assembly of transmembrane subunits and cytosolic subunits of enzyme complex is the first important step for its activation. In this step, p47PHOX is the most important component which is phosphorylated, translocated from cytoplasm to the membrane to interact with gp91PHOX (Decoursey and Ligeti, 2005; Bedard and Krause 2007; Leto et al., 2009). Herein, it has been found that MCP-1 treatment or forced expression of MCPIP resulted in expression of p47PHOX and increased the membrane-associated p47PHOX level, and knockdown of MCPIP in MCP-1-treated cells decreased the expression and translocation of p47PHOX. Moreover, NADPH oxidase activity inhibitor apocynin and knockdown of p47PHOX with antisense oligonucleotides inhibited membrane translocation of p47PHOX, ROS production, and expression of OC-related genes CTSK and TRAP. Thus, MCPIP increases ROS production and induces expression of OC-related genes by increasing expression and translocation of p47PHOX. As a zinc-finger protein with a nuclear localization sequence (Zhou et al., 2006) and RNase activity (Matsushita et al., 2009; Skalniak et al., 2009), MCPIP may serve as a novel regulator for several genes at the transcriptional and post-transcriptional level because of its DNA and RNA binding property. Therefore, MCPIP may regulate directly the expression of p47PHOX as a transcriptional factor. Secondly, it has been reported that MCPIP activates MAPK signal pathway (Younce and Kolattukudy, 2010). Activation of MAPK may be an important reason for the MCPIP-induced expression and translocation of p47PHOX oxidase.
- ER stress results from the accumulation of misfolded proteins which leads to the induction of the unfolded protein response (UPR) (Malhotra and Kaufman, 2007). ROS production is known to cause proteins to aggregate and misfold. Here, it has been demostrated that MCP-1 treatment and forced expression of MCPIP induced ER stress via generation of ROS in the monocytes during induction of OC differentiation. The important role of ER stress in monocyte differentiation into OC precursors was demonstrated by the findings that inhibition of ER stress inhibited differentiation and known ER stress inducers caused differentiation. ER stress inhibitor TUDC and knockdown of IRE-1 showed that inhibition of ER stress leads to inhibition of MCPIP-induced OC precursor differentiation. It has also been demonstrated herein that thapsgargin and tunicamycin that are known important inducers of UPR/ER stress induce differentiation of monocytes into OC precursors without MCP-1 or any other inducers. Thapsgargin, an inhibitor of ER-specific Ca-ATPase, has previously been shown to induce OC differentiation from RAW264.7 macrophage cells and mouse bone marrow cells (Takami et al., 1997; Yip et al., 2005).
- Autophagy is generally thought of as a survival mechanism, although its dysregulation has been linked to non-apoptotic cell death (Wang, 2008; Glick et al., 2010). Since differentiation involves disappearance of one set of proteins and appearance of a new set of proteins, a self-digestion process such as autophagy could be involved in this process. In fact, autophagy has been reported to be an important event for differentiation of the chronic myelogenous leukemia K562 cells (Colosetti et al., 2009), adipocytes (Malhotra and Kaufman, 2007; Singh et al., 2009; Goldman et al., 2010), paneth cells (Stappenbeck, 2010), and neuronal differentiation (Zeng and Zhou, 2008). Beclin-1 is a critical component in the class III PI3K complex (PI3KC3) that induces the formation of autophagosomes in mammalian systems (Wang, 2008). It has been demonstrated that Beclin-1 bridges autophagy and differentiation, and the process of autophagy and differentiation requires up-regulation of Beclin-1 (Wang, 2008). Inhibition of differentiation by PI3K inhibitors and knockdown of Beclin-1 and Atg7 strongly suggest that MCPIP induced OC precursor differentiation via autophagy. Emerging data now indicate that ER stress is a potent inducer of autophagy (Sakaki and Kaufman, 2008). Herein, the inventors have discovered that MCPIP induces OC precursor differentiation via autophagy which depends on MCPIP-induced ROS production and ER stress. It has been reported that ER stress can activate p38 MAPK signal pathway to induce autophagy (Kim et al., 2008). Moreover, ER stress activates phosphorylation of protein kinase-like ERK (PERK), an ER-localized transmembrane protein, and induction of IRE-1 is necessary for radiation-induced autophagy in mouse embryonic fibroblasts (Kim et al., 2010). Activation of NF-κB and MAPK signal pathway is necessary for OC differentiation (Huang et al., 2006). However, it was reported that MCPIP inhibits activation of NF-κB induced by IL-β (Skalniak et al., 2009) and lipopolysaccharide stimulation (Liang et al., 2008) while over-expression of MCPIP can cause activation of JNK and p38 (Younce and Kolattukudy, 2010). Thus, activation of p38 instead of NF-κB signaling is probably involved in MCPIP-mediated differentiation of monocytes into OC precursors. The overall pathway involved in the MCP-1/MCPIP-mediated OC precursor differentiation is shown in Supplementary Scheme 51.
- Inflammatory bone erosion is involved in many pathological conditions (Lu et al., 2007; Ha et al., 2010). The novel inventive features recited herein provide a new insight into the mechanism by which MCP-1 induces differentiation of monocytic cells into TRAP- and CTSK-expressing cells that can proceed to differentiate into functional OCs in the presence of RANKL, and suggest that MCPIP is a novel target for therapy of inflammatory bone erosion.
- Human BMCs were from Stemcell Technologies. α-MEM, FBS, HBSS, trypsin, recombinant human M-CSF (300-25), human MCP-1, and Trizol reagent were purchased from Invitrogen. Anti-β-actin, CTSK monoclonal antibodies, CeO2 nanoparticles, apocynin, TUDC, 3′-MA, and LY294002 were from Sigma-Aldrich. Anti-TRAP, p47PHOX, Fas, IRE-1, GRP78, Beclin-1, LC3 polyclonal antibodies, goat anti-rabbit and mouse secondary antibodies, and specific siRNA for IRE-1 and Beclin-1 were purchased from Santa Cruz Biotechnology. Specific siRNA for MCPIP and negative control siRNA were obtained from Ambion. Anti-MCPIP polyclonal antibody was prepared as indicated before (Zhou et al., 2006; Younce and Kolattukudy, 2010).
- BMCs were cultured in α-MEM supplemented with 10% FBS containing 30 ng/ml M-CSF, 100 U/ml penicillin, and 100 μg/ml streptomycin in 5% CO2 at 37° C. OC precursor cells were induced after 3-day culture as Ha et al. (2010). At this point, cells were treated with 50 ng/ml MCP-1, inhibitors, or gene transfection. For inhibitor treatment, CeO2 (10 μM), apocynin (100 μg/ml), TUDC (100 μM), 3′-MA (50 μM), and LY294002 (20 μM) were added 6 h before gene transfection.
- Gene Transfection and siRNA Knockdown
- OC precursor cells were transfected with 1 μg GFP or MCPIP-GFP eukaryotic expression plasmids for gene-gain-function assay by using
Fugene 6. For gene silencing, chemically synthesized siRNA duplex (100 nM) targeting MCPIP, IRE-1, or Beclin-1 was transfected into OC precursor cells using DharmaFECT (Dharmacon) for 24 h prior to transfection with MCPIP-GFP or GFP plasmid. A scrambled siRNA was used as a negative control. For knockdown expression of p47PHOX, specific antisense (AS) (5′-CCAGCAGGGCGATGTGACGGATGAA-3′) and sense (5′-ATGGGGGACACCTTCATCCGTCAC-3′) oligonucleotides were designed and synthesized by phosphorothioate modification by Integrated DNA Technologies. The oligonucleotides were transfected into OC precursor cells using Lipofectamine 2000 for 24 h before MCPIP—GFP plasmid transfection. - Three days after MCP-1 treatment or 4 days after MCPIP transfection, cells were fixed for histological staining for TRAP as described previously (Kim et al., 2006a). Briefly, following fixation, cells were stained with freshly prepared TRAP staining solution (naphthol AS-MX phosphate, fast red violet LB salt, and potassium sodium tartrate). On each coverslip, totally at least 500 cells were examined and the TRAP-positive cells were counted in 3-5 fields (20× objective), and the percentage of TRAP-positive cells was calculated.
- Cells were centrifuged after wash with PBS for twice. Cell pellet was resuspended in 1 ml Trizol reagent and the total RNA was extracted with chloroform and isopropanol, purified on Qiagen Mini-Prep column, and treated with DNase. High Capacity cDNA Reverse Transcription kit (Applied Biosystems) was used for cDNA preparation from 1 μg total RNA. qRT-PCR was done in triplicate in an ABI PRISM 7900HT Sequence Detection System with 5% cDNA product, primers (Supplementary Table 51) at 125 nM, and Fast SYBR Green Master Mix (Applied Biosystems). Relative quantitation of PCR products was done by the 2−ΔΔCTmethod, CT=cycles to threshold, and ΔΔCT=(target gene CT)−(β-actin reference gene CT). Final data were described as fold changes against control cells.
- Oxidant production in OC precursor cells was assessed by measuring the oxidation of intracellular DHR123 (Molecular Probes) as described previously (Younce et al., 2010). ROS production was expressed as folds compared with control cells expressing GFP alone.
- OC precursor cells were harvested, sonicated on ice in a buffer containing 100 mM KCl, 3 mM NaCl, 3.5 mM MgCl2, 10 mM HEPES, 1 mM EGTA, 10 μg/ml pepstatin, 10 μg/ml leupeptin, and 0.5 mM PMSF; lysates were centrifuged at 600 g for 10 min at 4° C. to remove nuclei and unbroken cells. The supernatant was then ultracentrifuged at 100000 g for 1 h at 4° C. Membranes were washed in the same buffer, quantified (Lemarie), and resuspended in Laemmli sample buffer, before western blot analysis.
- Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting were performed as described elsewhere. Samples (30 μg of protein) were subjected to 12% SDS-PAGE for most of target protein and 18% SDS-PAGE for LC3-II and LC3, transferred onto PVDF membranes (Millipore), and assayed for MCPIP, p47PHOX, TRAP, CTSK, GRP78, IRE-1, Beclin-1, LC3-II, and β-actin or Fas (loading control) protein expression by chemiluminescence detection (Pierce ECL kit) according to the manufacturer's instructions. The specific protein bands were quantified by densitometric analysis with GS-690 Image Densitometer (Bio-Rad).
- Data are represented as mean±SD of experiments performed on at least three separate occasions. Student's t-test was used to compare the means of normally distributed continuous variables. P<0.05 indicated statistical significance.
- Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art of molecular biology. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described herein. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting.
- Reference is made to standard textbooks of molecular biology that contain definitions and methods and means for carrying out basic techniques, encompassed by the present invention. See, for example, Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York (1982) and Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York (1989); Methods in Plant Molecular Biology, Maliga et al, Eds., Cold Spring Harbor Laboratory Press, New York (1995); Arabidopsis, Meyerowitz et al, Eds., Cold Spring Harbor Laboratory Press, New York (1994) and the various references cited therein.
- Finally, while various embodiments of the present invention have been shown and described herein, it will be obvious that such embodiments are provided by way of example only. Numerous variations, changes and substitutions may be made without departing from the invention herein. Accordingly, it is intended that the invention be limited only by the spirit and scope of the appended claims. The teachings of all patents and other references cited herein are incorporated herein by reference in their entirety to the extent they are not inconsistent with the teachings herein.
-
- 1. Azfer A., Niu J., Rogers et al. Activation of endoplasmic reticulum stress response during the development of ischemic heart disease. Am. J. Physiol. Heart Circ. Physiol. 2006; 291:H1411-H1420.
- 2. Baerga R., Zhang Y., Chen P. H., et al. Targeted deletion of autophagy-related 5 (atg5) impairs adipogenesis in a cellular model and in mice. Autophagy 2009; 5:1118-1130.
- 3. Bedard K., Krause K. H. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol. Rev. 2007; 87:245-313.
- 4. Blommaart E. F., Krause U., Sehellens J. P., et al. The phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002 inhibit autophagy in isolated rat hepatocytes. Eur. J. Biochem. 1997; 243:240-246.
- 5. Boyce B. F., Yao Z., Zhang Q., et al. New roles for osteoclasts in bone. Ann NY Acad. Sci. 2007; 1116:245-254.
- 6. Colosetti P., Puissant A., Robert G., et al. Autophagy is an important event for megakaryocytic differentiation of the chronic myelogenous leukemia K562 cell line. Autophagy 2009; 5:1092-1098.
- 7. Decoursey T. E., Ligeti E. Regulation and termination of NADPH oxidase activity. Cell. Mol. Life. Sci. 2005; 62:2173-2193.
- 8. Gerszten R. E., Friedrich E. B., Matsui T., et al. A role of phosphoinositide 3-kinase in monocyte recruitment under flow conditions. J. Biol. Chem. 2001; 276:26846-26851.
- 9. Glick D., Barth S., Macleod K. F. Autophagy: cellular and molecular mechanisms. J. Pathol. 2010; 221:3-12.
- 10. Goldman S., Zhang Y., Jin S. Autophagy and adipogenesis: implications in obesity and type II diabetes. Autophagy 2010; 6:179-181.
- 11. Ha J., Choi H. S., Lee Y., et al.
CXC chemokine ligand 2 induced by receptor activator of NF-kappa B ligand enhances osteoclastogenesis. J. Immunol. 2010; 184:4717-4724. - 12. Harris E. D. Jr. Rheumatoid arthritis: pathophysiology and implications for therapy. N. Engl. J. Med. 1990; 322:1277-1289.
- 13. Huang H., Rya J., Ha J., et al. Osteoclast differentiation requires TAK1 and MKK6 for NFATc1 induction and NF-kappaB transactivation by RANKL. Cell Death Differ. 2006; 13:1879-1891.
- 14. Kim M. S., Day C. J., Morrison N. A. MCP-1 is induced by receptor activator of nuclear factor-κB ligand, promotes human osteoclast fusion, and rescues granulocyte macrophage colony-stimulating factor suppression of osteoclast formation. J. Biol. Chem. 2005; 280:16163-16169.
- 15. Kim M. S., Day C. J., Selinger C. I., et al. MCP-1-induced human osteoclast-like cells are tartrate-resistant acid phosphatase, NFATc1, and calcitonin receptor-positive but require receptor activator of NFkappaB ligand for bone resorption. J. Biol. Chem. 2006a; 281:1274-1285.
- 16. Kim M. S., Magno C. L., Day C. J., et al. Induction of chemokines and chemokine receptors CCR2b and CCR4 in authentic human osteoclasts differentiated with RANKL and osteoclast like cells differentiated by MCP-1 and RANTES. J. Cell. Biochem. 2006b; 97:512-518.
- 17. Kim D. S., Kim J. H., Lee G. H., et al. p38 mitogen-activated protein kinase is involved in endoplasmic reticulum stress-induced cell death and autophagy in human gingival fibroblasts. Biol. Pharm. Bull. 2008; 33:545-549.
- 18. Kim K. W., Moretti L., Mitchell L. R. et a. Endoplasmic reticulum stress mediates radiation-induced autophagy by perk-eIF2alpha in caspase-3/7-deficient cells. Oncogene 2010; 29:3241-3251.
- 19. Kiviranta R., Morko J., Uusitalo H., et al. Accelerated turnover of metaphyseal trabecular bone in mice overexpressing cathepsin K. J. Bone Miner. Res. 2001; 16:1444-1452.
- 20, Leto T. L., Morand S., Hurt D., et al. Targeting and regulation of reactive oxygen species generation by Nox family NADPH oxidases. Antioxid. Redox Signal. 2009; 11:2607-2619.
- 21. Liang J., Wang J., Azfer A., et al. A novel CCCH-zinc finger protein family regulates proinflammatory activation of macrophages. J. Biol. Chem. 2008; 283:6337-6346.
- 22, Lu Y., Cai Z., Xiao G., et al. Monocyte chemotactic protein-1 mediates prostate cancer-induced bone resorption. Cancer Res. 2007; 67:3646-3653.
- 23. Malhotra J. D., Kaufman R. J., Endoplasmic reticulum stress and oxidative stress: a vicious cycle or a double-edged sword? Antioxid. Redox Signal. 2007; 9:2277-2293.
- 24, Malhotra J. D., Miao H., Zhang K., et al. Antioxidants reduce endoplasmic reticulum stress and improve protein secretion. Proc. Natl. Acad. Sci. USA 2008; 105:18525-18530.
- 25. Matsushita K., Takeuchi O., Standley D. M., et al. Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature 2009; 458:1185-1190.
- 26, Miyamoto K., Ninomiya K., Sonoda K. H., et al. MCP-1 expressed by osteoclasts stimulates osteoclastogenesis in an autocrine/paracrine manner. Biochem. Biophys. Res. Commun. 2009; 383:373-377.
- 27, Mundy G. R. Osteoporosis and inflammation. Nutr. Rev. 2007; 65:5147-5151.
- 28. Niu J., Azfer A., Rogers L. M., et al. Cardioprotective effects of cerium oxide nanoparticles in a transgenic murine model of cardiomyopathy. Cardiovasc. Res. 2007; 73:549-559.
- 29, Niu J., Azfer A., Zhelyabovska O., et al. Monocyte chemotactic protein (MCP)-1 promotes angiogenesis via a novel transcription factor, MCP-1-induced protein (MCPIP). J. Biol. Chem. 2008; 283:14542-14551.
- 30. Petiot A., Ogier-Denis E., Blommaart E. F., et al. Distinct classes of
phosphatidylinositol 3′-kinases are involved in signaling pathways that control macroautophagy in HT-29 cells. J. Biol. Chem. 2000; 275:992-998. - 31. Sakaki K., Kaufman R. J. Regulation of ER stress-induced macroautophagy by protein kinase C. Autophagy 2008; 4:841-843.
- 32. Sakiyama H., Masuda R., Inoue N., et al. Establishment and characterization of macrophage-like cell lines expressing osteoclast-specific markers. J. Bone Miner. Metab. 2001; 19:220-227.
- 33, Sato K., Takayanagi H. Osteoclasts, rheumatoid arthritis, and osteoimmunology. Curr. Opin. Rheumatol. 2006; 18:419-426.
- 34. Singh R., Xiang Y., Wang Y., et al. Autophagy regulates adipose mass and differentiation in mice. J. Clin. Invest. 2009; 119:3329-3339.
- 35, Skalniak L., Mizgalska D., Zarebski A., et al. Regulatory feedback loop between NF-kappaB and MCP-1-induced
protein 1 RNase. FEBS J. 2009; 276:5892-5905. - 36. Stappenbeck T. S. The role of autophagy in Paneth cell differentiation and secretion. Mucosal. Immunol. 2010; 3:8-10.
- 37, Steinbeck M. J., Kim J. K., Trudeau M. J. Involvement of hydrogen peroxide in the differentiation of clonal HD-11EM cells into osteoclast-like cells. J. Cell. Physiol. 1998; 176:574-587.
- 38. Sugimura R., Li L. Shifting in balance between osteogenesis and adipogenesis substantially influences hematopoiesis. J. Mol. Cell. Biol. 2010; 2:61-62.
- 39, Takami M., Woo J. T., Takahashi N., et al. Ca2+-ATPase inhibitors and Ca2+-ionophore induce osteoclast-like cell formation in the cocultures of mouse bone marrow cells and calvarial cells. Biochem. Biophys. Res. Commun. 1997; 237:111-115.
- 40, Tsai Y. Y., Oca-Cossio J., Agering K., et al. Novel synthesis of cerium oxide nanoparticles for free radical scavenging. Nanomedicine (Lond) 2007; 2:325-332.
- 41. Vrotsos E. G., Kolattukudy P. E., Sugaya K. MCP-1 involvement in glial differentiation of neuroprogenitor cells through APP signaling. Brain Res. Bull. 2009; 79:97-103.
- 42.
Wang J. Beclin 1 bridges autophagy, apoptosis and differentiation. Autophagy 2008; 4:947-948. - 43. Wise G. E., Frazier-Bowers S., D'Souza R. N. Cellular, molecular, and genetic determinants of tooth eruption. Crit. Rev. Oral Biol. Med. 2002; 13:323-334.
- 44. Wolf G. Role of reactive oxygen species in angiotensin II-mediated renal growth, differentiation, and apoptosis. Antioxid. Redox Signal. 2005; 7:1337-1345.
- 45. Xia C., Meng Q., Liu L. Z., et al. Reactive oxygen species regulate angiogenesis and tumor growth through vascular endothelial growth factor. Cancer Res. 2007; 67:10823-10830.
- 46. Xue X, Piao J. H., Nakajima A., et al. Tumor necrosis factor alpha (TNFα) induces the unfolded protein response (UPR) in a reactive oxygen species (ROS)-dependent fashion, and the UPR counteracts ROS accumulation by TNFα. J. Biol. Chem. 2005; 280:33917-33925.
- 47. Yamasaki N., Tsuboi H., Hirao M., et al. High oxygen tension prolongs the survival of osteoclast precursors via macrophage colony-stimulating factor. Bone 2009; 44:71-79.
- 48. Yip K. H., Zheng M. H., Steer J. H., et al. Thapsigargin modulates osteoclastogenesis through the regulation of RANKL-induced signaling pathways and reactive oxygen species production. J. Bone Miner. Res. 2005; 20:1462-1471.
- 49. Younce C. W., Kolattukudy P. E. MCP-1 causes cardiomyoblast death via autophagy resulting from ER stress caused by oxidative stress generated by inducing a novel zinc-finger protein, MCPIP. Biochem. J. 2010; 426:43-53.
- 50. Younce C. W., Azfer A., Kolattukudy P. E. MCP-1 (monocyte chemotactic protein-1)-induced protein, a recently identified zinc finger protein, induces adipogenesis in 3T3-L1 pre-adipocytes without peroxisome proliferator-activated receptor gamma. J. Biol. Chem. 2009; 284:27620-27628.
- 51. Younce C. W., Wang K., Kolattukudy P. E. Hyperglycemia-induced cardiomyocyte death is mediated via MCP-1 production and induction of a novel zinc-finger protein MCPIP. Cardiovasc. Res. 2010; 87:665-674.
- 52. Zeng M., Zhou J. N. Roles of autophagy and mTOR signaling in neuronal differentiation of mouse neuroblastoma cells. Cell. Signal. 2008; 20:659-665.
- 53. Zhang K. H., Tian H. Y., Gao X., et al. Ferritin heavy chain-mediated iron homeostasis and subsequent increased reactive oxygen species production are essential for epithelial-mesenchymal transition. Cancer Res. 2009; 69:5340-5348.
- 54. Zhou L., Azfer A., Niu J., et al. Monocyte chemoattractant protein-1 induces a novel transcription factor that causes cardiac myocyte apoptosis and ventricular dysfunction. Circ. Res. 2006; 98:1177-1185.
Claims (22)
1. A method of treating a condition in a patient in need, the method comprising administering to the patient a therapeutically effective amount of a composition that inhibits the expression or action of MCPIP, wherein said condition comprises abnormal osteoclast-induced bone resorption.
2. The method of claim 1 , wherein the patient in need is exhibiting pre-arthritic symptoms.
3. The method of claim 1 , wherein the patient in need is exhibiting pre-osteoporotic symptoms.
4. The method of claim 1 wherein the composition comprises siRNA specific for MCPIP.
5. The method of claim 1 wherein the composition comprises an antisense nucleotide specific for MCPIP.
6. The method of claim 1 wherein the composition comprises shRNA.
7. The method of claim 1 wherein the composition comprises an antibody specific to MCPIP.
8. The method of claim 1 , wherein the composition is administered to the patient via intrabuccal, oral, rectal, pulmonary, ocular, or transdermal administration.
9. The method of claim 1 , wherein the condition comprises an osteoporosis-related condition.
10. The method of claim 1 , wherein the condition comprises Rheumatoid Arthritis.
11. The method of claim 1 , wherein inhibiting MCPIP levels comprises directly administering the composition to the patient in need.
12. The method of claim 1 , wherein administering the composition comprises administering a composition comprising a therapeutically effective amount of the composition that inhibits the expression or action of MCPIP and a pharmaceutically acceptable excipient.
13. A method of inhibiting osteoclast production in a patient in need, comprising:
administering a therapeutically effective amount of a composition that inhibits the expression or action of MCPIP in the patient.
14. The method of claim 13 , wherein the patient in need is exhibiting symptoms of rheumatoid arthritis, osteoarthritis, and/or osteoporosis.
15. The method of claim 13 , wherein administering a therapeutically effective amount of a composition includes a composition comprising: a composition that inhibits the expression or action of MCPIP, and a pharmaceutically acceptable excipient.
16. The method of claim 13 , wherein the composition includes an MCPIP siRNA, an shRNA, an antibody specific to MCPIP, and/or an antisense nucleotide specific for MCPIP.
17. The method of claim 1 , wherein the condition is an inflammatory joint disease.
18. A method of treating an inflammatory disease in a patient in need thereof, said method comprising:
procuring human bone marrow mononuclear cells (BMCs) from said patient to obtain procured BMC's;
treating said procured BMC's, ex vivo, by blocking expression of MCPIP in said cells to obtain treated BMC's;
administering said treated BMC's to said patient.
19. The method of claim 18 , wherein said treating comprises subjecting said procured BMC's to an antisense nucleotide specific to an mRNA sequence encoding MCPIP, an antibody specific to MCPIP, siRNA specific to MCPIP, or shRNA specific to MCPIP.
20. (canceled)
21. (canceled)
22. The method of claim 18 , wherein said inflammatory disease comprises osteoporosis, or arthritis.
Priority Applications (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US13/362,261 US20130195882A1 (en) | 2012-01-31 | 2012-01-31 | Mcpip protection against osteoclast production |
US14/194,254 US9469853B2 (en) | 2005-12-20 | 2014-02-28 | MCPIP protection against osteoclast production |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US13/362,261 US20130195882A1 (en) | 2012-01-31 | 2012-01-31 | Mcpip protection against osteoclast production |
Related Parent Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US12/539,907 Continuation US20100041736A1 (en) | 2005-12-20 | 2009-08-12 | Isolated MCPIP and Methods of Use |
Related Child Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US14/194,254 Continuation US9469853B2 (en) | 2005-12-20 | 2014-02-28 | MCPIP protection against osteoclast production |
Publications (1)
Publication Number | Publication Date |
---|---|
US20130195882A1 true US20130195882A1 (en) | 2013-08-01 |
Family
ID=48870423
Family Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US13/362,261 Abandoned US20130195882A1 (en) | 2005-12-20 | 2012-01-31 | Mcpip protection against osteoclast production |
US14/194,254 Expired - Fee Related US9469853B2 (en) | 2005-12-20 | 2014-02-28 | MCPIP protection against osteoclast production |
Family Applications After (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US14/194,254 Expired - Fee Related US9469853B2 (en) | 2005-12-20 | 2014-02-28 | MCPIP protection against osteoclast production |
Country Status (1)
Country | Link |
---|---|
US (2) | US20130195882A1 (en) |
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN108070564A (en) * | 2017-12-07 | 2018-05-25 | 深圳市人民医院 | A kind of method for proving consideration convey dye siRNA security |
Family Cites Families (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2006079051A2 (en) * | 2005-01-24 | 2006-07-27 | University Of Rochester | Methods of modifying cd11c+ dendritic cell development to form osteoclasts functional in the bone environment |
EP1978999A4 (en) * | 2005-12-20 | 2009-08-19 | Univ Central Florida Res Found | Isolated mcpip and methods of use |
-
2012
- 2012-01-31 US US13/362,261 patent/US20130195882A1/en not_active Abandoned
-
2014
- 2014-02-28 US US14/194,254 patent/US9469853B2/en not_active Expired - Fee Related
Cited By (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN108070564A (en) * | 2017-12-07 | 2018-05-25 | 深圳市人民医院 | A kind of method for proving consideration convey dye siRNA security |
Also Published As
Publication number | Publication date |
---|---|
US20140302044A1 (en) | 2014-10-09 |
US20160257960A9 (en) | 2016-09-08 |
US9469853B2 (en) | 2016-10-18 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Wang et al. | Osteoclast precursor differentiation by MCPIP via oxidative stress, endoplasmic reticulum stress, and autophagy | |
Xu et al. | Resveratrol transcriptionally regulates miRNA-18a-5p expression ameliorating diabetic nephropathy via increasing autophagy. | |
Chen et al. | miR‐136‐3p targets PTEN to regulate vascularization and bone formation and ameliorates alcohol‐induced osteopenia | |
EP2753696B1 (en) | The mirna-212/132 family as a therapeutic target | |
EP2186528A1 (en) | Pharmaceutical containing hif-1 alpha and hif-2 alpha expression inhibitor | |
Wang et al. | microRNA-20b contributes to high glucose-induced podocyte apoptosis by targeting SIRT7 | |
WO2007103114A2 (en) | Notch inhibition in the treatment or prevention of atherosclerosis | |
Chen et al. | Epigallocatechin gallate attenuates uric acid-induced injury in rat renal interstitial fibroblasts NRK-49F by up-regulation of miR-9. | |
US9469853B2 (en) | MCPIP protection against osteoclast production | |
US20160244755A1 (en) | Treating diseases associated with pgc1-alpha by modulating micrornas mir-130a and mir-130b | |
Na et al. | Itraconazole attenuates hepatic gluconeogenesis and promotes glucose uptake by regulating AMPK pathway | |
KR101541974B1 (en) | Composition and method for enhancing differentiation of neuronal stem cells comprising miR29b | |
KR20110082515A (en) | Treatment of scleroderma | |
Zhang et al. | MiR-873-5p regulated LPS-induced oxidative stress via targeting heme oxygenase-1 (HO-1) in KGN cells | |
US11788091B2 (en) | Methods and compositions for diagnosing and treating prostate cancer based on long noncoding RNA overlapping the LCK gene that regulates prostate cancer cell growth | |
US20090137631A1 (en) | Methods and pharmaceutical compositions for regulation of g- and/or gc-rich nucleic acid expression | |
CN112206324A (en) | Application of FHL2 in preparing medicine for treating polycystic ovarian syndrome ovulation disorder | |
KR20190084902A (en) | PHARMACEUTICAL COMPOSITION FOR PREVENTING OR TREATING MUSCULAR DISEASE OR CACHEXIA COMPRISING, AS ACTIVE INGREDIENT, miRNA LOCATED IN Dlk1-Dio3 CLUSTER OR VARIANT THEREOF | |
CN111411112B (en) | Targeting inhibitor of MAPK6P2 gene and application thereof | |
US10443059B2 (en) | Pharmaceutical composition and method for reducing scar formation | |
KR101928427B1 (en) | Composition comprising microRNA-497 for preventing or treating of alcoholic liver diseases and method of screening thereof | |
WO2019051277A1 (en) | Compositions and methods for enhancing weight loss | |
US20230295634A1 (en) | Compositions and methods for the treatment of diseases by enhancing arginase 2 in macrophages | |
KR102012837B1 (en) | Screening method of therapeutic agents for leukaemia by regulating expression of olfactory receptor by G9a and LSD1 | |
Guo et al. | Circ_0004214 prevents human cardiomyocytes from doxorubicin induced cardiotoxicity by governing the miR-22-3p/GATA4 pathway: Circ_0004214 prevents human cardiomyocytes |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AS | Assignment |
Owner name: UNIVERSITY OF CENTRAL FLORIDA RESEARCH FOUNDATION, Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KOLATTUKUDY, PAPPACHAN E.;REEL/FRAME:028052/0190 Effective date: 20120301 |
|
STCB | Information on status: application discontinuation |
Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION |