WO2024102688A1 - Compositions for treating osteoclastogenesis disorders and/or rheumatoid arthritis - Google Patents
Compositions for treating osteoclastogenesis disorders and/or rheumatoid arthritis Download PDFInfo
- Publication number
- WO2024102688A1 WO2024102688A1 PCT/US2023/078886 US2023078886W WO2024102688A1 WO 2024102688 A1 WO2024102688 A1 WO 2024102688A1 US 2023078886 W US2023078886 W US 2023078886W WO 2024102688 A1 WO2024102688 A1 WO 2024102688A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- seq
- sequence
- combinations
- rankl
- pharmaceutically acceptable
- Prior art date
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 80
- 206010039073 rheumatoid arthritis Diseases 0.000 title claims abstract description 60
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 title claims abstract description 26
- 208000035475 disorder Diseases 0.000 title description 6
- 230000001105 regulatory effect Effects 0.000 claims abstract description 41
- 210000000988 bone and bone Anatomy 0.000 claims abstract description 33
- 201000010099 disease Diseases 0.000 claims abstract description 20
- 206010023203 Joint destruction Diseases 0.000 claims abstract description 12
- 208000001132 Osteoporosis Diseases 0.000 claims abstract description 12
- 230000001771 impaired effect Effects 0.000 claims abstract description 11
- 108091034117 Oligonucleotide Proteins 0.000 claims description 104
- 238000012230 antisense oligonucleotides Methods 0.000 claims description 95
- 239000000074 antisense oligonucleotide Substances 0.000 claims description 84
- 239000002773 nucleotide Substances 0.000 claims description 74
- 125000003729 nucleotide group Chemical group 0.000 claims description 74
- 150000003839 salts Chemical class 0.000 claims description 58
- 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 compound 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 claims description 51
- 150000007523 nucleic acids Chemical class 0.000 claims description 50
- 150000002632 lipids Chemical class 0.000 claims description 46
- 102000039446 nucleic acids Human genes 0.000 claims description 43
- 108020004707 nucleic acids Proteins 0.000 claims description 43
- 238000000034 method Methods 0.000 claims description 35
- -1 carrier Substances 0.000 claims description 28
- 108020004459 Small interfering RNA Proteins 0.000 claims description 22
- 239000002105 nanoparticle Substances 0.000 claims description 19
- 230000009368 gene silencing by RNA Effects 0.000 claims description 18
- 108091030071 RNAI Proteins 0.000 claims description 17
- 239000013603 viral vector Substances 0.000 claims description 15
- 238000011282 treatment Methods 0.000 claims description 13
- 239000003981 vehicle Substances 0.000 claims description 13
- 239000002502 liposome Substances 0.000 claims description 10
- 239000003814 drug Substances 0.000 claims description 9
- 239000003085 diluting agent Substances 0.000 claims description 5
- 210000000349 chromosome Anatomy 0.000 claims description 3
- 230000027455 binding Effects 0.000 abstract description 20
- 238000009739 binding Methods 0.000 abstract description 19
- 210000002997 osteoclast Anatomy 0.000 description 134
- 108010025832 RANK Ligand Proteins 0.000 description 120
- 102100024568 Tumor necrosis factor ligand superfamily member 11 Human genes 0.000 description 120
- 210000004027 cell Anatomy 0.000 description 96
- 239000002777 nucleoside Substances 0.000 description 71
- 230000014509 gene expression Effects 0.000 description 70
- 108090000623 proteins and genes Proteins 0.000 description 68
- 235000002639 sodium chloride Nutrition 0.000 description 58
- 239000003623 enhancer Substances 0.000 description 55
- 125000003835 nucleoside group Chemical group 0.000 description 48
- 102100022970 Basic leucine zipper transcriptional factor ATF-like Human genes 0.000 description 34
- 101000903742 Homo sapiens Basic leucine zipper transcriptional factor ATF-like Proteins 0.000 description 34
- 102000053602 DNA Human genes 0.000 description 30
- 108020004414 DNA Proteins 0.000 description 30
- 239000003795 chemical substances by application Substances 0.000 description 28
- 102000040945 Transcription factor Human genes 0.000 description 26
- 108091023040 Transcription factor Proteins 0.000 description 26
- 108010077544 Chromatin Proteins 0.000 description 24
- 210000003483 chromatin Anatomy 0.000 description 24
- 102000040430 polynucleotide Human genes 0.000 description 24
- 108091033319 polynucleotide Proteins 0.000 description 24
- 239000002157 polynucleotide Substances 0.000 description 24
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 22
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 22
- 230000004069 differentiation Effects 0.000 description 22
- 238000004458 analytical method Methods 0.000 description 21
- 101000946889 Homo sapiens Monocyte differentiation antigen CD14 Proteins 0.000 description 20
- 230000001965 increasing effect Effects 0.000 description 20
- 210000001744 T-lymphocyte Anatomy 0.000 description 18
- 150000003833 nucleoside derivatives Chemical class 0.000 description 17
- 239000004055 small Interfering RNA Substances 0.000 description 17
- 108020005065 3' Flanking Region Proteins 0.000 description 16
- 102000007591 Tartrate-Resistant Acid Phosphatase Human genes 0.000 description 16
- 108010032050 Tartrate-Resistant Acid Phosphatase Proteins 0.000 description 16
- 102100035877 Monocyte differentiation antigen CD14 Human genes 0.000 description 15
- 230000000295 complement effect Effects 0.000 description 15
- 210000000963 osteoblast Anatomy 0.000 description 15
- 108020005029 5' Flanking Region Proteins 0.000 description 14
- 239000013598 vector Substances 0.000 description 14
- 108010018525 NFATC Transcription Factors Proteins 0.000 description 13
- 102000002673 NFATC Transcription Factors Human genes 0.000 description 13
- 241000702421 Dependoparvovirus Species 0.000 description 12
- 102100031573 Hematopoietic progenitor cell antigen CD34 Human genes 0.000 description 12
- 101000777663 Homo sapiens Hematopoietic progenitor cell antigen CD34 Proteins 0.000 description 12
- 125000002091 cationic group Chemical group 0.000 description 12
- 230000000694 effects Effects 0.000 description 12
- 102000004169 proteins and genes Human genes 0.000 description 12
- 238000012163 sequencing technique Methods 0.000 description 12
- 239000011780 sodium chloride Substances 0.000 description 12
- 102000007651 Macrophage Colony-Stimulating Factor Human genes 0.000 description 11
- 108010046938 Macrophage Colony-Stimulating Factor Proteins 0.000 description 11
- 102100038895 Myc proto-oncogene protein Human genes 0.000 description 11
- 230000015572 biosynthetic process Effects 0.000 description 11
- 239000000872 buffer Substances 0.000 description 11
- 230000006870 function Effects 0.000 description 11
- 230000002177 osteoclastogenic effect Effects 0.000 description 11
- 241000699666 Mus <mouse, genus> Species 0.000 description 10
- 108010018242 Transcription Factor AP-1 Proteins 0.000 description 10
- 102100023132 Transcription factor Jun Human genes 0.000 description 10
- 150000001875 compounds Chemical class 0.000 description 10
- 210000004940 nucleus Anatomy 0.000 description 10
- 102100024894 PR domain zinc finger protein 1 Human genes 0.000 description 9
- 238000011529 RT qPCR Methods 0.000 description 9
- 230000003247 decreasing effect Effects 0.000 description 9
- 108020004999 messenger RNA Proteins 0.000 description 9
- 229920000642 polymer Polymers 0.000 description 9
- 230000000638 stimulation Effects 0.000 description 9
- 238000012546 transfer Methods 0.000 description 9
- 108091052242 Bromo- and Extra-Terminal domain (BET) family Proteins 0.000 description 8
- 238000012313 Kruskal-Wallis test Methods 0.000 description 8
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 8
- 108091028043 Nucleic acid sequence Proteins 0.000 description 8
- 102000009572 RNA Polymerase II Human genes 0.000 description 8
- 108010009460 RNA Polymerase II Proteins 0.000 description 8
- 238000003559 RNA-seq method Methods 0.000 description 8
- 101150006308 botA gene Proteins 0.000 description 8
- 238000012937 correction Methods 0.000 description 8
- 238000003197 gene knockdown Methods 0.000 description 8
- 230000001575 pathological effect Effects 0.000 description 8
- 238000013518 transcription Methods 0.000 description 8
- 230000035897 transcription Effects 0.000 description 8
- VUVUVNZRUGEAHB-CYBMUJFWSA-N 7-(3,5-dimethyl-4-isoxazolyl)-8-methoxy-1-[(1R)-1-(2-pyridinyl)ethyl]-3H-imidazo[4,5-c]quinolin-2-one Chemical compound C1([C@@H](C)N2C3=C4C=C(C(=CC4=NC=C3NC2=O)C2=C(ON=C2C)C)OC)=CC=CC=N1 VUVUVNZRUGEAHB-CYBMUJFWSA-N 0.000 description 7
- 108010009975 Positive Regulatory Domain I-Binding Factor 1 Proteins 0.000 description 7
- 239000002253 acid Substances 0.000 description 7
- 239000002585 base Substances 0.000 description 7
- 230000000875 corresponding effect Effects 0.000 description 7
- 230000006378 damage Effects 0.000 description 7
- 238000009826 distribution Methods 0.000 description 7
- 210000001616 monocyte Anatomy 0.000 description 7
- 238000003762 quantitative reverse transcription PCR Methods 0.000 description 7
- 241000894007 species Species 0.000 description 7
- RYYWUUFWQRZTIU-UHFFFAOYSA-K thiophosphate Chemical compound [O-]P([O-])([O-])=S RYYWUUFWQRZTIU-UHFFFAOYSA-K 0.000 description 7
- 102100023013 Basic leucine zipper transcriptional factor ATF-like 3 Human genes 0.000 description 6
- 206010051728 Bone erosion Diseases 0.000 description 6
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 6
- 101000903609 Homo sapiens Basic leucine zipper transcriptional factor ATF-like 3 Proteins 0.000 description 6
- 102100038069 Interferon regulatory factor 8 Human genes 0.000 description 6
- 108010057466 NF-kappa B Proteins 0.000 description 6
- 102000003945 NF-kappa B Human genes 0.000 description 6
- 229920002873 Polyethylenimine Polymers 0.000 description 6
- 238000003556 assay Methods 0.000 description 6
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol Chemical group C1C=C2C[C@@H](O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H]([C@H](C)CCCC(C)C)[C@@]1(C)CC2 HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 6
- 210000002540 macrophage Anatomy 0.000 description 6
- 230000001404 mediated effect Effects 0.000 description 6
- 108091027963 non-coding RNA Proteins 0.000 description 6
- 102000042567 non-coding RNA Human genes 0.000 description 6
- 230000008685 targeting Effects 0.000 description 6
- 230000001225 therapeutic effect Effects 0.000 description 6
- 238000002560 therapeutic procedure Methods 0.000 description 6
- 208000006386 Bone Resorption Diseases 0.000 description 5
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 5
- 108010033040 Histones Proteins 0.000 description 5
- 101001046677 Homo sapiens Integrin alpha-V Proteins 0.000 description 5
- 101001032345 Homo sapiens Interferon regulatory factor 8 Proteins 0.000 description 5
- 102100022337 Integrin alpha-V Human genes 0.000 description 5
- 108700011259 MicroRNAs Proteins 0.000 description 5
- 239000007864 aqueous solution Substances 0.000 description 5
- 230000024279 bone resorption Effects 0.000 description 5
- 238000004422 calculation algorithm Methods 0.000 description 5
- 230000008859 change Effects 0.000 description 5
- 238000009472 formulation Methods 0.000 description 5
- 102000046699 human CD14 Human genes 0.000 description 5
- 239000002679 microRNA Substances 0.000 description 5
- 210000001179 synovial fluid Anatomy 0.000 description 5
- 238000003786 synthesis reaction Methods 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 4
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 4
- 108010085371 Activating Transcription Factor 3 Proteins 0.000 description 4
- 208000020084 Bone disease Diseases 0.000 description 4
- 102100029895 Bromodomain-containing protein 4 Human genes 0.000 description 4
- 101710126815 Bromodomain-containing protein 4 Proteins 0.000 description 4
- 108091026890 Coding region Proteins 0.000 description 4
- 102100023032 Cyclic AMP-dependent transcription factor ATF-3 Human genes 0.000 description 4
- 108010010803 Gelatin Proteins 0.000 description 4
- 102000010029 Homer Scaffolding Proteins Human genes 0.000 description 4
- 108010077223 Homer Scaffolding Proteins Proteins 0.000 description 4
- 102100034343 Integrase Human genes 0.000 description 4
- 101710203526 Integrase Proteins 0.000 description 4
- 102100020675 Krueppel-like factor 2 Human genes 0.000 description 4
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 4
- 239000004472 Lysine Substances 0.000 description 4
- 108020003217 Nuclear RNA Proteins 0.000 description 4
- 102000043141 Nuclear RNA Human genes 0.000 description 4
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M Potassium chloride Chemical compound [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 4
- MEFKEPWMEQBLKI-AIRLBKTGSA-N S-adenosyl-L-methioninate Chemical compound O[C@@H]1[C@H](O)[C@@H](C[S+](CC[C@H](N)C([O-])=O)C)O[C@H]1N1C2=NC=NC(N)=C2N=C1 MEFKEPWMEQBLKI-AIRLBKTGSA-N 0.000 description 4
- 101100174184 Serratia marcescens fosA gene Proteins 0.000 description 4
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 4
- ISAKRJDGNUQOIC-UHFFFAOYSA-N Uracil Chemical compound O=C1C=CNC(=O)N1 ISAKRJDGNUQOIC-UHFFFAOYSA-N 0.000 description 4
- 229960001570 ademetionine Drugs 0.000 description 4
- 150000001413 amino acids Chemical class 0.000 description 4
- 239000011324 bead Substances 0.000 description 4
- 210000000234 capsid Anatomy 0.000 description 4
- 238000002487 chromatin immunoprecipitation Methods 0.000 description 4
- 230000002596 correlated effect Effects 0.000 description 4
- OPTASPLRGRRNAP-UHFFFAOYSA-N cytosine Chemical compound NC=1C=CNC(=O)N=1 OPTASPLRGRRNAP-UHFFFAOYSA-N 0.000 description 4
- 230000001973 epigenetic effect Effects 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- 239000012091 fetal bovine serum Substances 0.000 description 4
- 101150078861 fos gene Proteins 0.000 description 4
- 239000008273 gelatin Substances 0.000 description 4
- 229920000159 gelatin Polymers 0.000 description 4
- 235000019322 gelatine Nutrition 0.000 description 4
- 235000011852 gelatine desserts Nutrition 0.000 description 4
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 4
- 238000009396 hybridization Methods 0.000 description 4
- 230000005764 inhibitory process Effects 0.000 description 4
- KWGKDLIKAYFUFQ-UHFFFAOYSA-M lithium chloride Chemical compound [Li+].[Cl-] KWGKDLIKAYFUFQ-UHFFFAOYSA-M 0.000 description 4
- 229910001629 magnesium chloride Inorganic materials 0.000 description 4
- 239000002245 particle Substances 0.000 description 4
- 229920001983 poloxamer Polymers 0.000 description 4
- 108090000765 processed proteins & peptides Proteins 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 239000000523 sample Substances 0.000 description 4
- 238000000527 sonication Methods 0.000 description 4
- 210000000130 stem cell Anatomy 0.000 description 4
- LRSASMSXMSNRBT-UHFFFAOYSA-N 5-methylcytosine Chemical compound CC1=CNC(=O)N=C1N LRSASMSXMSNRBT-UHFFFAOYSA-N 0.000 description 3
- 102100027211 Albumin Human genes 0.000 description 3
- 108010088751 Albumins Proteins 0.000 description 3
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 3
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 3
- 102100024812 DNA (cytosine-5)-methyltransferase 3A Human genes 0.000 description 3
- 108050002829 DNA (cytosine-5)-methyltransferase 3A Proteins 0.000 description 3
- 229920002307 Dextran Polymers 0.000 description 3
- 101000979205 Homo sapiens Transcription factor MafA Proteins 0.000 description 3
- 102100037765 Periostin Human genes 0.000 description 3
- 101710199268 Periostin Proteins 0.000 description 3
- 102100025368 Runt-related transcription factor 2 Human genes 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 102100023237 Transcription factor MafA Human genes 0.000 description 3
- 102100027654 Transcription factor PU.1 Human genes 0.000 description 3
- 230000021736 acetylation Effects 0.000 description 3
- 238000006640 acetylation reaction Methods 0.000 description 3
- 150000007513 acids Chemical class 0.000 description 3
- 230000002917 arthritic effect Effects 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 239000007975 buffered saline Substances 0.000 description 3
- 239000001110 calcium chloride Substances 0.000 description 3
- 229910001628 calcium chloride Inorganic materials 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 235000012000 cholesterol Nutrition 0.000 description 3
- 230000001276 controlling effect Effects 0.000 description 3
- 230000009089 cytolysis Effects 0.000 description 3
- 238000013461 design Methods 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 238000005516 engineering process Methods 0.000 description 3
- 230000002708 enhancing effect Effects 0.000 description 3
- 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 3
- 239000000284 extract Substances 0.000 description 3
- 125000003843 furanosyl group Chemical group 0.000 description 3
- 230000036541 health Effects 0.000 description 3
- 238000012165 high-throughput sequencing Methods 0.000 description 3
- 210000005260 human cell Anatomy 0.000 description 3
- 238000003119 immunoblot Methods 0.000 description 3
- 230000002757 inflammatory effect Effects 0.000 description 3
- 239000003112 inhibitor Substances 0.000 description 3
- 230000002452 interceptive effect Effects 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 238000001543 one-way ANOVA Methods 0.000 description 3
- 230000008506 pathogenesis Effects 0.000 description 3
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 3
- 239000002953 phosphate buffered saline Substances 0.000 description 3
- 229920001184 polypeptide Polymers 0.000 description 3
- 239000002243 precursor Substances 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 102000004196 processed proteins & peptides Human genes 0.000 description 3
- 108010008929 proto-oncogene protein Spi-1 Proteins 0.000 description 3
- 230000007115 recruitment Effects 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 230000011664 signaling Effects 0.000 description 3
- 239000000243 solution Substances 0.000 description 3
- 238000007619 statistical method Methods 0.000 description 3
- 239000000126 substance Substances 0.000 description 3
- 210000001519 tissue Anatomy 0.000 description 3
- TZCPCKNHXULUIY-RGULYWFUSA-N 1,2-distearoyl-sn-glycero-3-phosphoserine Chemical compound CCCCCCCCCCCCCCCCCC(=O)OC[C@H](COP(O)(=O)OC[C@H](N)C(O)=O)OC(=O)CCCCCCCCCCCCCCCCC TZCPCKNHXULUIY-RGULYWFUSA-N 0.000 description 2
- GZCWLCBFPRFLKL-UHFFFAOYSA-N 1-prop-2-ynoxypropan-2-ol Chemical compound CC(O)COCC#C GZCWLCBFPRFLKL-UHFFFAOYSA-N 0.000 description 2
- KSXTUUUQYQYKCR-LQDDAWAPSA-M 2,3-bis[[(z)-octadec-9-enoyl]oxy]propyl-trimethylazanium;chloride Chemical compound [Cl-].CCCCCCCC\C=C/CCCCCCCC(=O)OCC(C[N+](C)(C)C)OC(=O)CCCCCCC\C=C/CCCCCCCC KSXTUUUQYQYKCR-LQDDAWAPSA-M 0.000 description 2
- ZISVTYVLWSZJAL-UHFFFAOYSA-N 3,6-bis[4-[bis(2-hydroxydodecyl)amino]butyl]piperazine-2,5-dione Chemical compound CCCCCCCCCCC(O)CN(CC(O)CCCCCCCCCC)CCCCC1NC(=O)C(CCCCN(CC(O)CCCCCCCCCC)CC(O)CCCCCCCCCC)NC1=O ZISVTYVLWSZJAL-UHFFFAOYSA-N 0.000 description 2
- FHVDTGUDJYJELY-UHFFFAOYSA-N 6-{[2-carboxy-4,5-dihydroxy-6-(phosphanyloxy)oxan-3-yl]oxy}-4,5-dihydroxy-3-phosphanyloxane-2-carboxylic acid Chemical compound O1C(C(O)=O)C(P)C(O)C(O)C1OC1C(C(O)=O)OC(OP)C(O)C1O FHVDTGUDJYJELY-UHFFFAOYSA-N 0.000 description 2
- 102000013563 Acid Phosphatase Human genes 0.000 description 2
- 108010051457 Acid Phosphatase Proteins 0.000 description 2
- 102000007469 Actins Human genes 0.000 description 2
- 108010085238 Actins Proteins 0.000 description 2
- 229930024421 Adenine Natural products 0.000 description 2
- GFFGJBXGBJISGV-UHFFFAOYSA-N Adenine Chemical compound NC1=NC=NC2=C1N=CN2 GFFGJBXGBJISGV-UHFFFAOYSA-N 0.000 description 2
- 102100023006 Basic leucine zipper transcriptional factor ATF-like 2 Human genes 0.000 description 2
- 102000001805 Bromodomains Human genes 0.000 description 2
- 108050009021 Bromodomains Proteins 0.000 description 2
- 229920001661 Chitosan Polymers 0.000 description 2
- 102000008186 Collagen Human genes 0.000 description 2
- 108010035532 Collagen Proteins 0.000 description 2
- 229920000858 Cyclodextrin Polymers 0.000 description 2
- 108010024491 DNA Methyltransferase 3A Proteins 0.000 description 2
- 238000009007 Diagnostic Kit Methods 0.000 description 2
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 2
- JZNWSCPGTDBMEW-UHFFFAOYSA-N Glycerophosphorylethanolamin Natural products NCCOP(O)(=O)OCC(O)CO JZNWSCPGTDBMEW-UHFFFAOYSA-N 0.000 description 2
- ZWZWYGMENQVNFU-UHFFFAOYSA-N Glycerophosphorylserin Natural products OC(=O)C(N)COP(O)(=O)OCC(O)CO ZWZWYGMENQVNFU-UHFFFAOYSA-N 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- AEMRFAOFKBGASW-UHFFFAOYSA-N Glycolic acid Polymers OCC(O)=O AEMRFAOFKBGASW-UHFFFAOYSA-N 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101000903615 Homo sapiens Basic leucine zipper transcriptional factor ATF-like 2 Proteins 0.000 description 2
- 101001139146 Homo sapiens Krueppel-like factor 2 Proteins 0.000 description 2
- 101000990902 Homo sapiens Matrix metalloproteinase-9 Proteins 0.000 description 2
- 101001030211 Homo sapiens Myc proto-oncogene protein Proteins 0.000 description 2
- 101000687344 Homo sapiens PR domain zinc finger protein 1 Proteins 0.000 description 2
- 101000801232 Homo sapiens Tumor necrosis factor receptor superfamily member 1B Proteins 0.000 description 2
- 241000713666 Lentivirus Species 0.000 description 2
- 239000000232 Lipid Bilayer Substances 0.000 description 2
- 102100030412 Matrix metalloproteinase-9 Human genes 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- KPKZJLCSROULON-QKGLWVMZSA-N Phalloidin Chemical compound N1C(=O)[C@@H]([C@@H](O)C)NC(=O)[C@H](C)NC(=O)[C@H](C[C@@](C)(O)CO)NC(=O)[C@H](C2)NC(=O)[C@H](C)NC(=O)[C@@H]3C[C@H](O)CN3C(=O)[C@@H]1CSC1=C2C2=CC=CC=C2N1 KPKZJLCSROULON-QKGLWVMZSA-N 0.000 description 2
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 2
- 229920001212 Poly(beta amino esters) Polymers 0.000 description 2
- 229920000954 Polyglycolide Polymers 0.000 description 2
- ZTHYODDOHIVTJV-UHFFFAOYSA-N Propyl gallate Chemical compound CCCOC(=O)C1=CC(O)=C(O)C(O)=C1 ZTHYODDOHIVTJV-UHFFFAOYSA-N 0.000 description 2
- 241000125945 Protoparvovirus Species 0.000 description 2
- 238000011530 RNeasy Mini Kit Methods 0.000 description 2
- 238000010240 RT-PCR analysis Methods 0.000 description 2
- 238000012300 Sequence Analysis Methods 0.000 description 2
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 2
- RAHZWNYVWXNFOC-UHFFFAOYSA-N Sulphur dioxide Chemical compound O=S=O RAHZWNYVWXNFOC-UHFFFAOYSA-N 0.000 description 2
- 102100040296 TATA-box-binding protein Human genes 0.000 description 2
- 108700019146 Transgenes Proteins 0.000 description 2
- 238000010162 Tukey test Methods 0.000 description 2
- 102100028787 Tumor necrosis factor receptor superfamily member 11A Human genes 0.000 description 2
- 102100033733 Tumor necrosis factor receptor superfamily member 1B Human genes 0.000 description 2
- ATBOMIWRCZXYSZ-XZBBILGWSA-N [1-[2,3-dihydroxypropoxy(hydroxy)phosphoryl]oxy-3-hexadecanoyloxypropan-2-yl] (9e,12e)-octadeca-9,12-dienoate Chemical compound CCCCCCCCCCCCCCCC(=O)OCC(COP(O)(=O)OCC(O)CO)OC(=O)CCCCCCC\C=C\C\C=C\CCCCC ATBOMIWRCZXYSZ-XZBBILGWSA-N 0.000 description 2
- 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 2
- 230000002378 acidificating effect Effects 0.000 description 2
- 229960000643 adenine Drugs 0.000 description 2
- 229940072056 alginate Drugs 0.000 description 2
- 235000010443 alginic acid Nutrition 0.000 description 2
- 229920000615 alginic acid Polymers 0.000 description 2
- AWUCVROLDVIAJX-UHFFFAOYSA-N alpha-glycerophosphate Natural products OCC(O)COP(O)(O)=O AWUCVROLDVIAJX-UHFFFAOYSA-N 0.000 description 2
- 125000000539 amino acid group Chemical group 0.000 description 2
- 230000000692 anti-sense effect Effects 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 206010003246 arthritis Diseases 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 235000011148 calcium chloride Nutrition 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 230000024245 cell differentiation Effects 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 229930183167 cerebroside Natural products 0.000 description 2
- 150000001784 cerebrosides Chemical class 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 230000001684 chronic effect Effects 0.000 description 2
- 238000003776 cleavage reaction Methods 0.000 description 2
- 229920001436 collagen Polymers 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 229940097362 cyclodextrins Drugs 0.000 description 2
- 229940104302 cytosine Drugs 0.000 description 2
- 230000007423 decrease Effects 0.000 description 2
- 230000007812 deficiency Effects 0.000 description 2
- 239000000412 dendrimer Substances 0.000 description 2
- 229920000736 dendritic polymer Polymers 0.000 description 2
- 229960003964 deoxycholic acid Drugs 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 230000003292 diminished effect Effects 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 230000007608 epigenetic mechanism Effects 0.000 description 2
- 230000006718 epigenetic regulation Effects 0.000 description 2
- 150000002148 esters Chemical class 0.000 description 2
- CBOQJANXLMLOSS-UHFFFAOYSA-N ethyl vanillin Chemical compound CCOC1=CC(C=O)=CC=C1O CBOQJANXLMLOSS-UHFFFAOYSA-N 0.000 description 2
- 230000001747 exhibiting effect Effects 0.000 description 2
- 238000010195 expression analysis Methods 0.000 description 2
- 230000001605 fetal effect Effects 0.000 description 2
- 238000000684 flow cytometry Methods 0.000 description 2
- 238000002509 fluorescent in situ hybridization Methods 0.000 description 2
- 239000012634 fragment Substances 0.000 description 2
- 150000002270 gangliosides Chemical class 0.000 description 2
- 230000002068 genetic effect Effects 0.000 description 2
- 235000011187 glycerol Nutrition 0.000 description 2
- 230000013632 homeostatic process Effects 0.000 description 2
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 2
- 238000007901 in situ hybridization Methods 0.000 description 2
- 238000011534 incubation Methods 0.000 description 2
- 230000001939 inductive effect Effects 0.000 description 2
- 239000012442 inert solvent Substances 0.000 description 2
- 230000002401 inhibitory effect Effects 0.000 description 2
- 210000000265 leukocyte Anatomy 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 238000011068 loading method Methods 0.000 description 2
- 239000012139 lysis buffer Substances 0.000 description 2
- 235000011147 magnesium chloride Nutrition 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 230000007935 neutral effect Effects 0.000 description 2
- 231100000252 nontoxic Toxicity 0.000 description 2
- 230000003000 nontoxic effect Effects 0.000 description 2
- 238000009806 oophorectomy Methods 0.000 description 2
- 150000007524 organic acids Chemical class 0.000 description 2
- 235000005985 organic acids Nutrition 0.000 description 2
- 230000037361 pathway Effects 0.000 description 2
- 125000001095 phosphatidyl group Chemical group 0.000 description 2
- WTJKGGKOPKCXLL-RRHRGVEJSA-N phosphatidylcholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCCCCCCC=CCCCCCCCC WTJKGGKOPKCXLL-RRHRGVEJSA-N 0.000 description 2
- 150000008104 phosphatidylethanolamines Chemical class 0.000 description 2
- 229920000747 poly(lactic acid) Polymers 0.000 description 2
- 229920000058 polyacrylate Polymers 0.000 description 2
- 229920001610 polycaprolactone Polymers 0.000 description 2
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 2
- 230000029279 positive regulation of transcription, DNA-dependent Effects 0.000 description 2
- 239000001103 potassium chloride Substances 0.000 description 2
- 235000011164 potassium chloride Nutrition 0.000 description 2
- 239000003755 preservative agent Substances 0.000 description 2
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 description 2
- XKMLYUALXHKNFT-UHFFFAOYSA-N rGTP Natural products C1=2NC(N)=NC(=O)C=2N=CN1C1OC(COP(O)(=O)OP(O)(=O)OP(O)(O)=O)C(O)C1O XKMLYUALXHKNFT-UHFFFAOYSA-N 0.000 description 2
- 238000003753 real-time PCR Methods 0.000 description 2
- 230000008844 regulatory mechanism Effects 0.000 description 2
- 238000012552 review Methods 0.000 description 2
- 238000007363 ring formation reaction Methods 0.000 description 2
- 230000007017 scission Effects 0.000 description 2
- 230000035945 sensitivity Effects 0.000 description 2
- 230000019491 signal transduction Effects 0.000 description 2
- 150000003384 small molecules Chemical class 0.000 description 2
- 235000017557 sodium bicarbonate Nutrition 0.000 description 2
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 2
- FHHPUSMSKHSNKW-SMOYURAASA-M sodium deoxycholate Chemical compound [Na+].C([C@H]1CC2)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC([O-])=O)C)[C@@]2(C)[C@@H](O)C1 FHHPUSMSKHSNKW-SMOYURAASA-M 0.000 description 2
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 2
- 239000007790 solid phase Substances 0.000 description 2
- 239000002904 solvent Substances 0.000 description 2
- 150000003408 sphingolipids Chemical class 0.000 description 2
- 239000003381 stabilizer Substances 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 230000001629 suppression Effects 0.000 description 2
- 229940124597 therapeutic agent Drugs 0.000 description 2
- RWQNBRDOKXIBIV-UHFFFAOYSA-N thymine Chemical compound CC1=CNC(=O)NC1=O RWQNBRDOKXIBIV-UHFFFAOYSA-N 0.000 description 2
- 230000002103 transcriptional effect Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 230000017105 transposition Effects 0.000 description 2
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 2
- 229940035893 uracil Drugs 0.000 description 2
- 230000003612 virological effect Effects 0.000 description 2
- 239000011534 wash buffer Substances 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- RHCSKNNOAZULRK-APZFVMQVSA-N 2,2-dideuterio-2-(3,4,5-trimethoxyphenyl)ethanamine Chemical compound NCC([2H])([2H])C1=CC(OC)=C(OC)C(OC)=C1 RHCSKNNOAZULRK-APZFVMQVSA-N 0.000 description 1
- CHHHXKFHOYLYRE-UHFFFAOYSA-M 2,4-Hexadienoic acid, potassium salt (1:1), (2E,4E)- Chemical compound [K+].CC=CC=CC([O-])=O CHHHXKFHOYLYRE-UHFFFAOYSA-M 0.000 description 1
- AAAQFGUYHFJNHI-SFHVURJKSA-N 2-[(4S)-6-(4-chlorophenyl)-8-methoxy-1-methyl-4H-[1,2,4]triazolo[4,3-a][1,4]benzodiazepin-4-yl]-N-ethylacetamide Chemical compound N([C@H](C1=NN=C(C)N1C1=CC=C(OC)C=C11)CC(=O)NCC)=C1C1=CC=C(Cl)C=C1 AAAQFGUYHFJNHI-SFHVURJKSA-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
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 1
- WXNZTHHGJRFXKQ-UHFFFAOYSA-N 4-chlorophenol Chemical compound OC1=CC=C(Cl)C=C1 WXNZTHHGJRFXKQ-UHFFFAOYSA-N 0.000 description 1
- JYCQQPHGFMYQCF-UHFFFAOYSA-N 4-tert-Octylphenol monoethoxylate Chemical compound CC(C)(C)CC(C)(C)C1=CC=C(OCCO)C=C1 JYCQQPHGFMYQCF-UHFFFAOYSA-N 0.000 description 1
- 241000702423 Adeno-associated virus - 2 Species 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- 229920000936 Agarose Polymers 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
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 206010073478 Anaplastic large-cell lymphoma Diseases 0.000 description 1
- 108020005544 Antisense RNA Proteins 0.000 description 1
- 101100001210 Arabidopsis thaliana AGT3 gene Proteins 0.000 description 1
- 208000023275 Autoimmune disease Diseases 0.000 description 1
- 108091032955 Bacterial small RNA Proteins 0.000 description 1
- 206010065687 Bone loss Diseases 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- 238000010356 CRISPR-Cas9 genome editing Methods 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 108010024682 Core Binding Factor Alpha 1 Subunit Proteins 0.000 description 1
- 102000004127 Cytokines Human genes 0.000 description 1
- 108090000695 Cytokines Proteins 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
- 125000000824 D-ribofuranosyl group Chemical group [H]OC([H])([H])[C@@]1([H])OC([H])(*)[C@]([H])(O[H])[C@]1([H])O[H] 0.000 description 1
- 230000007067 DNA methylation Effects 0.000 description 1
- 101710177611 DNA polymerase II large subunit Proteins 0.000 description 1
- 101710184669 DNA polymerase II small subunit Proteins 0.000 description 1
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 1
- 102000007260 Deoxyribonuclease I Human genes 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 101100310856 Drosophila melanogaster spri gene Proteins 0.000 description 1
- 102100039247 ETS-related transcription factor Elf-4 Human genes 0.000 description 1
- 108010067770 Endopeptidase K Proteins 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 229920001917 Ficoll Polymers 0.000 description 1
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 1
- 102100023372 Fos-related antigen 1 Human genes 0.000 description 1
- 108090000123 Fos-related antigen 1 Proteins 0.000 description 1
- 108700039691 Genetic Promoter Regions Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 102100039869 Histone H2B type F-S Human genes 0.000 description 1
- 101000813135 Homo sapiens ETS-related transcription factor Elf-4 Proteins 0.000 description 1
- 101001035372 Homo sapiens Histone H2B type F-S Proteins 0.000 description 1
- 101000876829 Homo sapiens Protein C-ets-1 Proteins 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 229930010555 Inosine Natural products 0.000 description 1
- UGQMRVRMYYASKQ-KQYNXXCUSA-N Inosine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC(O)=C2N=C1 UGQMRVRMYYASKQ-KQYNXXCUSA-N 0.000 description 1
- 238000012351 Integrated analysis Methods 0.000 description 1
- 108091029795 Intergenic region Proteins 0.000 description 1
- 101710186679 Kruppel-like factor 2 Proteins 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- 229930182816 L-glutamine Natural products 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
- 102000006835 Lamins Human genes 0.000 description 1
- 108010047294 Lamins Proteins 0.000 description 1
- 208000032004 Large-Cell Anaplastic Lymphoma Diseases 0.000 description 1
- 102000043136 MAP kinase family Human genes 0.000 description 1
- 108091054455 MAP kinase family Proteins 0.000 description 1
- 102100028123 Macrophage colony-stimulating factor 1 Human genes 0.000 description 1
- 101710127797 Macrophage colony-stimulating factor 1 Proteins 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 102000001776 Matrix metalloproteinase-9 Human genes 0.000 description 1
- 108010015302 Matrix metalloproteinase-9 Proteins 0.000 description 1
- 108010050345 Microphthalmia-Associated Transcription Factor Proteins 0.000 description 1
- 102100030157 Microphthalmia-associated transcription factor Human genes 0.000 description 1
- 102000007474 Multiprotein Complexes Human genes 0.000 description 1
- 108010085220 Multiprotein Complexes Proteins 0.000 description 1
- 206010031149 Osteitis Diseases 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 108010016731 PPAR gamma Proteins 0.000 description 1
- 239000002033 PVDF binder Substances 0.000 description 1
- 108020002230 Pancreatic Ribonuclease Proteins 0.000 description 1
- 102000005891 Pancreatic ribonuclease Human genes 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 102100038825 Peroxisome proliferator-activated receptor gamma Human genes 0.000 description 1
- 102000018042 Peroxisome proliferator-activated receptor gamma coactivator 1-beta Human genes 0.000 description 1
- 108050007135 Peroxisome proliferator-activated receptor gamma coactivator 1-beta Proteins 0.000 description 1
- 108010009711 Phalloidine Proteins 0.000 description 1
- RVGRUAULSDPKGF-UHFFFAOYSA-N Poloxamer Chemical compound C1CO1.CC1CO1 RVGRUAULSDPKGF-UHFFFAOYSA-N 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 102100035251 Protein C-ets-1 Human genes 0.000 description 1
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 description 1
- 101710086015 RNA ligase Proteins 0.000 description 1
- 108091028733 RNTP Proteins 0.000 description 1
- 108010038036 Receptor Activator of Nuclear Factor-kappa B Proteins 0.000 description 1
- 108091081062 Repeated sequence (DNA) Proteins 0.000 description 1
- 101710148027 Ribulose bisphosphate carboxylase/oxygenase activase 1, chloroplastic Proteins 0.000 description 1
- 101710201629 Ribulose bisphosphate carboxylase/oxygenase activase 2, chloroplastic Proteins 0.000 description 1
- 101710102802 Runt-related transcription factor 2 Proteins 0.000 description 1
- 238000012311 Shapiro-Wilk normality test Methods 0.000 description 1
- 238000012167 Small RNA sequencing Methods 0.000 description 1
- ABBQHOQBGMUPJH-UHFFFAOYSA-M Sodium salicylate Chemical compound [Na+].OC1=CC=CC=C1C([O-])=O ABBQHOQBGMUPJH-UHFFFAOYSA-M 0.000 description 1
- 108010090804 Streptavidin Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- RYYWUUFWQRZTIU-UHFFFAOYSA-N Thiophosphoric acid Chemical class OP(O)(S)=O RYYWUUFWQRZTIU-UHFFFAOYSA-N 0.000 description 1
- 108700029229 Transcriptional Regulatory Elements Proteins 0.000 description 1
- 102000008579 Transposases Human genes 0.000 description 1
- 108010020764 Transposases Proteins 0.000 description 1
- 239000013504 Triton X-100 Substances 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 1
- 101710178436 Tumor necrosis factor receptor superfamily member 11A Proteins 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 239000003070 absorption delaying agent Substances 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 239000012190 activator Substances 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 235000010419 agar Nutrition 0.000 description 1
- 125000001931 aliphatic group Chemical group 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 230000000844 anti-bacterial effect Effects 0.000 description 1
- 229940121375 antifungal agent Drugs 0.000 description 1
- 239000003429 antifungal agent Substances 0.000 description 1
- 239000002787 antisense oligonuctleotide Substances 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- WPYMKLBDIGXBTP-UHFFFAOYSA-N benzoic acid group Chemical group C(C1=CC=CC=C1)(=O)O WPYMKLBDIGXBTP-UHFFFAOYSA-N 0.000 description 1
- 238000007622 bioinformatic analysis Methods 0.000 description 1
- 238000003766 bioinformatics method Methods 0.000 description 1
- 230000008827 biological function Effects 0.000 description 1
- 230000031018 biological processes and functions Effects 0.000 description 1
- 208000018339 bone inflammation disease Diseases 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 229940125763 bromodomain inhibitor Drugs 0.000 description 1
- 230000003139 buffering effect Effects 0.000 description 1
- 210000004899 c-terminal region Anatomy 0.000 description 1
- 238000010805 cDNA synthesis kit Methods 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000004700 cellular uptake Effects 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- 230000003081 coactivator Effects 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 239000000084 colloidal system Substances 0.000 description 1
- 239000003184 complementary RNA Substances 0.000 description 1
- 238000004590 computer program Methods 0.000 description 1
- 239000012141 concentrate Substances 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 230000002079 cooperative effect Effects 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- 230000007547 defect Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 238000000432 density-gradient centrifugation Methods 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 229940042399 direct acting antivirals protease inhibitors Drugs 0.000 description 1
- 239000002612 dispersion medium Substances 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 239000012149 elution buffer Substances 0.000 description 1
- 238000010201 enrichment analysis Methods 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 229940073505 ethyl vanillin Drugs 0.000 description 1
- 239000013604 expression vector Substances 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 239000013020 final formulation Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 239000008098 formaldehyde solution Substances 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 238000010230 functional analysis Methods 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000010362 genome editing Methods 0.000 description 1
- 229960005150 glycerol Drugs 0.000 description 1
- 125000003827 glycol group Chemical group 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 210000003917 human chromosome Anatomy 0.000 description 1
- 125000004435 hydrogen atom Chemical group [H]* 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 229960003786 inosine Drugs 0.000 description 1
- 108010051621 interferon regulatory factor-8 Proteins 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 210000001503 joint Anatomy 0.000 description 1
- 208000018937 joint inflammation Diseases 0.000 description 1
- 238000011813 knockout mouse model Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 210000005053 lamin Anatomy 0.000 description 1
- 239000006194 liquid suspension Substances 0.000 description 1
- 239000008176 lyophilized powder Substances 0.000 description 1
- 238000010801 machine learning Methods 0.000 description 1
- 159000000003 magnesium salts Chemical class 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 238000013507 mapping Methods 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 238000002483 medication Methods 0.000 description 1
- 239000002609 medium Substances 0.000 description 1
- 239000012528 membrane Substances 0.000 description 1
- WSFSSNUMVMOOMR-NJFSPNSNSA-N methanone Chemical compound O=[14CH2] WSFSSNUMVMOOMR-NJFSPNSNSA-N 0.000 description 1
- DWCZIOOZPIDHAB-UHFFFAOYSA-L methyl green Chemical compound [Cl-].[Cl-].C1=CC(N(C)C)=CC=C1C(C=1C=CC(=CC=1)[N+](C)(C)C)=C1C=CC(=[N+](C)C)C=C1 DWCZIOOZPIDHAB-UHFFFAOYSA-L 0.000 description 1
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 108091070501 miRNA Proteins 0.000 description 1
- 239000011859 microparticle Substances 0.000 description 1
- 239000004005 microsphere Substances 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 210000005087 mononuclear cell Anatomy 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 210000005088 multinucleated cell Anatomy 0.000 description 1
- 238000002887 multiple sequence alignment Methods 0.000 description 1
- 210000000066 myeloid cell Anatomy 0.000 description 1
- 239000002088 nanocapsule Substances 0.000 description 1
- 239000002077 nanosphere Substances 0.000 description 1
- 238000007481 next generation sequencing Methods 0.000 description 1
- 238000001151 non-parametric statistical test Methods 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 230000004072 osteoblast differentiation Effects 0.000 description 1
- 208000002865 osteopetrosis Diseases 0.000 description 1
- 230000001009 osteoporotic effect Effects 0.000 description 1
- 210000004663 osteoprogenitor cell Anatomy 0.000 description 1
- 229940090668 parachlorophenol Drugs 0.000 description 1
- 238000001769 parametric statistical test Methods 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 239000001814 pectin Substances 0.000 description 1
- 235000010987 pectin Nutrition 0.000 description 1
- 229920001277 pectin Polymers 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 239000000546 pharmaceutical excipient Substances 0.000 description 1
- 239000012071 phase Substances 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- OJMIONKXNSYLSR-UHFFFAOYSA-N phosphorous acid Chemical class OP(O)O OJMIONKXNSYLSR-UHFFFAOYSA-N 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 229960000502 poloxamer Drugs 0.000 description 1
- 229920000768 polyamine Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 230000001323 posttranslational effect Effects 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 235000010241 potassium sorbate Nutrition 0.000 description 1
- 239000004302 potassium sorbate Substances 0.000 description 1
- 229940069338 potassium sorbate Drugs 0.000 description 1
- GUUBJKMBDULZTE-UHFFFAOYSA-M potassium;2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid;hydroxide Chemical compound [OH-].[K+].OCCN1CCN(CCS(O)(=O)=O)CC1 GUUBJKMBDULZTE-UHFFFAOYSA-M 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 210000001978 pro-t lymphocyte Anatomy 0.000 description 1
- 239000000473 propyl gallate Substances 0.000 description 1
- 235000010388 propyl gallate Nutrition 0.000 description 1
- 229940075579 propyl gallate Drugs 0.000 description 1
- 238000003908 quality control method Methods 0.000 description 1
- 238000010791 quenching Methods 0.000 description 1
- 230000000171 quenching effect Effects 0.000 description 1
- 239000013608 rAAV vector Substances 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000004985 regulation of immune system process Effects 0.000 description 1
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 1
- 238000009256 replacement therapy Methods 0.000 description 1
- 230000008672 reprogramming Effects 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 239000003161 ribonuclease inhibitor Substances 0.000 description 1
- 239000012723 sample buffer Substances 0.000 description 1
- 229940016590 sarkosyl Drugs 0.000 description 1
- 108700004121 sarkosyl Proteins 0.000 description 1
- 238000002864 sequence alignment Methods 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 229960005480 sodium caprylate Drugs 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- OABYVIYXWMZFFJ-ZUHYDKSRSA-M sodium glycocholate Chemical compound [Na+].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)NCC([O-])=O)C)[C@@]2(C)[C@@H](O)C1 OABYVIYXWMZFFJ-ZUHYDKSRSA-M 0.000 description 1
- KSAVQLQVUXSOCR-UHFFFAOYSA-M sodium lauroyl sarcosinate Chemical compound [Na+].CCCCCCCCCCCC(=O)N(C)CC([O-])=O KSAVQLQVUXSOCR-UHFFFAOYSA-M 0.000 description 1
- BYKRNSHANADUFY-UHFFFAOYSA-M sodium octanoate Chemical compound [Na+].CCCCCCCC([O-])=O BYKRNSHANADUFY-UHFFFAOYSA-M 0.000 description 1
- 239000001488 sodium phosphate Substances 0.000 description 1
- 229910000162 sodium phosphate Inorganic materials 0.000 description 1
- 235000011008 sodium phosphates Nutrition 0.000 description 1
- 229960004025 sodium salicylate Drugs 0.000 description 1
- JAJWGJBVLPIOOH-IZYKLYLVSA-M sodium taurocholate Chemical compound [Na+].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)[C@@H](O)C1 JAJWGJBVLPIOOH-IZYKLYLVSA-M 0.000 description 1
- FIWQZURFGYXCEO-UHFFFAOYSA-M sodium;decanoate Chemical compound [Na+].CCCCCCCCCC([O-])=O FIWQZURFGYXCEO-UHFFFAOYSA-M 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 125000001424 substituent group Chemical group 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- CCEKAJIANROZEO-UHFFFAOYSA-N sulfluramid Chemical group CCNS(=O)(=O)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)F CCEKAJIANROZEO-UHFFFAOYSA-N 0.000 description 1
- 229940044609 sulfur dioxide Drugs 0.000 description 1
- 235000010269 sulphur dioxide Nutrition 0.000 description 1
- 230000008093 supporting effect Effects 0.000 description 1
- 238000001847 surface plasmon resonance imaging Methods 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000011285 therapeutic regimen Methods 0.000 description 1
- 229940113082 thymine Drugs 0.000 description 1
- 230000036962 time dependent Effects 0.000 description 1
- 230000014616 translation Effects 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
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 1
- 125000002948 undecyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 238000012800 visualization Methods 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
Classifications
-
- 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
- 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
- A61K31/7125—Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
-
- 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
-
- 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.
-
- 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
- C12N2310/30—Chemical structure
- C12N2310/32—Chemical structure of the sugar
- C12N2310/323—Chemical structure of the sugar modified ring structure
- C12N2310/3231—Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
-
- 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
- C12N2310/30—Chemical structure
- C12N2310/34—Spatial arrangement of the modifications
- C12N2310/341—Gapmers, i.e. of the type ===---===
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/185—Osteoprotegerin; Osteoclast differentiation factor (ODF, RANKL)
Definitions
- Osteoclasts are large, multinucleated cells derived from myeloid lineage cells and are the primary cells responsible for bone resorption and bone homeostasis 1 ' 3 . Dysregulated osteoclast differentiation and function lead to bone destruction in pathological conditions, including rheumatoid arthritis (RA) 4 ' 6 .
- Macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-B ligand (RANKL) are key regulators of osteoclast differentiation, survival, and activity of osteoclasts.
- RANKL binds to its receptor, RANK 2 .
- RANKL activates the canonical osteoclastogenic signaling pathways and induces osteoclastogenic transcriptional and epigenetic program during osteoclastogenesis 7 ' 11 .
- RANKL- or RANK-deficient mice exhibit severe osteopetrosis with impaired osteoclast activity 12 ' 14 .
- the mechanisms by which RANKL induces osteoclast differentiation have been extensively studied, and the central transcription factors that are involved in initiating and maintaining the osteoclastogenic program are well defined.
- Nuclear Factor of Activated T cells, cl NFATcl
- NFATcl nuclear Factor of Activated T cells
- RANKL-induced S-adenosylmethionine (SAM) production mediates DNA methylation by de novo DNA methyltransferase 3a (Dnmt3a) during osteoclastogenesis and that inhibiting Dnmt3a regulates osteoclastogenesis and ovariectomy-induced bone loss 21 .
- SAM S-adenosylmethionine
- BET bromodomain and extraterminal
- targeting epigenetic regulators broadly affects many cell types since most cells utilize similar chromatin regulators to regulate epigenetic mechanisms. Thus, unraveling the osteoclast-specific regulatory mechanism is important for understanding the epigenetic network mediating lineage control of osteoclast formation and activity and for identifying a safe therapeutic target.
- Enhancers are cis regulatory elements and contain transcription factor-binding motifs 24 . Enhancers derive cell-type specific transcriptional activation or suppression independent of orientation or distance 24 .
- Super-enhancers SEs are clusters of enhancers and underlie cell identity and lineage commitment by driving high-level expression of genes encoding key regulators of cell identity and cell fate 25 ' 29 . SEs serve as hubs that recruit large protein complexes including the transcriptional apparatus, transcription factors, co-activators and chromatin regulators, and promote the expression of cell-fate decision genes 30 .
- Histone 3 acetylation at lysine 27 (H3K27ac), an active enhancer mark, is highly enriched in SEs 31 .
- SEs are also sensitive to the dynamic changes of pioneer master regulators during differentiation 32 .
- a recent study identified SEs in mouse osteoclasts using ChlP-seq of PU.l, a myeloid pioneer transcription factor 33 .
- human osteoclast-specific SEs have not yet been defined.
- enhancers produce non-coding RNA species, called enhancer RNAs (eRNAs) 34 ' 37 .
- eRNAs are transcribed from active tissue-specific enhancers including super-enhancers, and their biological function has been discovered in many different cells 38 ' 51 .
- the function and mechanism of superenhancers as well as SE-associated eRNAs in RANKL-stimulated human osteoclasts are yet to be firmly established.
- the inventor has identified a novel region within the regulatory region for a Nfatcl gene which is osteoclast- specific. The identification of this region has enabled the design of various osteoclast-specific therapies, including antisense oligonucleotide (ASO) therapy. Other suitable agents may be additionally or alternatively targeted to this region.
- ASO antisense oligonucleotide
- the invention provides a composition comprising: a pharmaceutically acceptable delivery vehicle, carrier, and/or diluent, and at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
- TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- ATTCAGCTCCACTTA SEQ ID NO: 2
- SEQ ID NO: 2 ATTCAGCTCCACTTA
- the composition comprises at least one of these antisense oligonucleotides comprising 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO: 1.
- the at least one antisense oligonucleotide comprises 18 to 30 nucleotides in length which comprise at least 15 consecutive nucleotides of SEQ ID NO: 2.
- the composition comprises an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2.
- the composition comprises an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2. In certain embodiments, the composition comprises an antisense oligonucleotide having at least one modified internucleoside linkage, sugar moiety, or nucleobase. In certain embodiments, the composition comprises a non- viral vector comprising the oligo is a lipid nanoparticle, lipidoid, or liposome.
- a method for treating an osteoclastogenesis- related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis, said method comprising delivering an agent which specifically binding to a human osteoclast-specific NFATcl regulatory region in chromosome 18.
- the agent is an anti-sense oligonucleotide, an RNAi, siRNA, or combinations thereof.
- the agent is delivered via a lipid nanoparticle, lipidoid, or liposome.
- the composition is delivered intravenously, via sitc-spccific administration to a joint, via targeted delivery to the bone, or intrathccally.
- a composition which is suitable for treatment of an osteoclastogenesis-related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis which comprises an agent which modulates the regulatory region of the osteoclast- specific Nfatcl gene.
- FIGs. 1A-1F provide the identification of RANKL-sensitive super enhancers (SEs) in human osteoclasts.
- SEs RANKL-sensitive super enhancers
- FIG 1A Schematic showing the differentiation of osteoclasts.
- FIGG IB Distribution of H3K27ac ChlP-seq enrichment scores in the indicated conditions.
- Enhancer regions are plotted in an increasing order based on their input-normalized H3K27ac ChlP- seq signal. SEs are defined as the population of enhancers above the inflection point of the curve.
- RANKL-sensitive SE was defined as a > 1.5-fold change in H3K27ac read density by RANKL (left panel).
- Disease ontology for RANKL-sensitive SE-associated genes with corresponding q-value (right panel) (FIG ID; control in black, R) Read density plot of H3K27ac ChlP-seq across typical enhancers (TEs) and SEs domains.
- FIG IE Graphs showing the differential H3K27ac ChlP-seq signals between RANKL and CTRL across enhancer domains. Increased (left) or decreased (right) H3K27ac ChlP-seq signals in RANKL are shown. Dark dots indicate the signals from SE regions and gray dots indicate the signals from TE regions. Boxplots of the change in H3K27ac ChlP-seq signals in the indicated conditions. :/? ⁇ ().() I by Kruskal-Wallis test with the Bonferroni correction. Representative SEs are highlighted along with their associated genes, (f) Representative tracks of H3K27ac ChlP-seq at NFATcl, PRDM1, MYC. IRF8, and KLF2 loci in the indicated conditions. Gray or black bar indicates up-regulated or down-regulated SEs, respectively.
- FIGs. 2A-2D show RANKL-sensitive SEs exhibit osteoclast-specificity, (a) H3K27ac ChlP-scq distribution for osteoclast, osteoblast, CD34+ progenitor and CD4+ T cell within RANKL-induced osteoclast SEs. Each color indicates a different cell type, (b) Box plots showing H3K27ac ChlP-seq signals across SE domains in the indicated cell type. ***; /? ⁇ ().001 , n.s.: not significant by Kruskal-Wallis test with the Bonferroni correction, (c) Chow-Ruskey diagram of SE- or TE-associated genes in osteoclast, osteoblast, CD34+ progenitor and CD4+ T cell.
- FIGs. 3A-3F illustrate dynamic chromatin accessibility at RANKL-sensitive SEs.
- FIGs. 4A-4I show sensitivity to iBET differentially regulates SE-eRNA expression.
- FIG4A is a Box plot displaying read densities of H3K27ac ChlP-seq in RANKL-sensitive SE domains in presence or absence of LBET151 (500 nM).
- the black line represents the control group (CTRL) and the gray line represents the RANKL-treated group
- CTR control group
- BATF1/3 knock-down efficiency in human OCPs was measured by RT-qPCR.
- Graph shows the percentage of TRAP-positive multinuclear cells (MNCs: more than three nuclei) per control, normalized relative to the number of osteoclasts formed in control siRNA conditions. ***; p ⁇ 0.001, **; p ⁇ 0.01, or *; p ⁇ 0.05 by Kruskal- Wallis test with the Bonferroni correction (c) or by student t- test (h,i).
- FIGs. 5A-5H demonstrate active RNA Pol II recruitment and transcription within RANKL-sensitive super-enhancers in human osteoclasts
- (a) Heatmaps show the enrichment of RNA Pol II ChlP-seq signals in RANKL-sensitive enhancer regions
- FIGs. 6A-6G show identification of dREG peaks in human osteoclasts, (a) Representative tracks of pro-seq at NFATcl -associated SE domain in the indicated conditions (CTRL: controls and RANKL: RANKL treated conditions). Up-regulated dREG peaks of pro-seq are shown, (b) A pie chart showing the genomic location of RANKL- inducible dREG peaks from pro-seq data.
- a bar graph is depicted to number of dREG peaks at TE and SE domains, (c) Aggregate plots showing mean Pro-seq, H3K27ac, Pol II ChlP- seq, and ATAC-seq signals centered on up-regulated dREG peaks in the indicated conditions, (d) Box plots showing Pro-seq, H3K27ac, Pol II ChlP-seq, and ATAC-seq signals centered on up-regulated dREG peaks in the indicated conditions.
- FIGs. 7A-7K show RANKL-induced SE-cRNAs arc enriched in synovial CD 14+ cells from RA patients,
- FISH fluorescence in situ hybridization
- NFATcl mRNA were measured by RT-qPCR in the indicated conditions
- a-tubulin was served as a loading control, (i) Box plots showing pro-seq signals within RANKL-induced SE derived from 2 different RA synovial CD14+ cells (RA-1, RA-2) and CD14+ cells from healthy control (CTRL), (j) Representative tracks for nuclear seq of RA synovial CD14+ cells in vicinity of NFATcl (RA: RA synovial OCPs, CTRL: disease control), (k) SE-eRNA:NFATcl was measured using RT-qPCR (see Methods). The data are shown as means ⁇ SEM.
- FIGs. 8A - 8B provides a mapping of RANKL-sensitive SEs in human osteoclasts.
- FIG. 8 A is a Table showing the correlation between biological replicates for H3K27ac, Pol II ChlP-seq, ATAC-seq experiments. The Pearson correlation coefficient was estimated based on the average read densities in 4kb regions around the TSS of RefSeq.
- FIG 8B shows gene ontology for downregulated SEs with corresponding adjusted p-value.
- FIGs. 9A - 9B provides Cell-type specific SEs in osteoblast and CD4+ T cell. Gene ontology for osteoblast-specific (a) and CD4+ T cell-specific (b) SEs with corresponding adjusted p-value.
- FIGs. 10A-10C provide Transcription Factors at RANKL-sensitive SEs.
- FIG 10A mRNA expression of NFATcl, PPARGC1B, and ITGAV obtained from publicly available data SRP047069.
- (b) Overall sequencing coverage of H3K27ac and ATAC-scq around Fos and NFkB binding motifs. Dotted line, dashed line, and solid lines represent coverage around motifs genome-wide, motifs within TEs, and motifs within SEs, respectively.
- the black line represents the control group (CTRL) and the gray line represents the RANKL-treated group.
- the solid line represents super-enhancer and the dotted line represents typical enhancer (TE).
- FIG. 11 provides RANKL-induced SE-eRNA expression in human osteoclasts. Representative tracks of pro-seq (upper tracks) and RNA-seq (bottom tracks, RA: RA synovial OCPs, CTRL: disease control) at PRDM-associated (a), MYC-associated (b) SE domain in the indicated conditions. Up-regulated dREG peaks of pro-seq (brown) are shown.
- FIGs. 12A-F demonstrate the role of SE-eRNA:NFATcl in human osteoclasts.
- M-CSF 20ng/ml
- RANKL 40ng/ml
- Osteoclastogenesis assay Left panel shows representative images of TRAP-stained cells. Right panel shows the percentage of TRAP positive multinuclear cells (MNCs: more than three nuclei) per control.
- NFATcl mRNA were measured by RT-qPCR in the indicated conditions after RANKL stimulation for 24 h.
- d Immunoblot analysis of NFATcl expression in the indicated conditions after RANKL stimulation for 24 h. a-tubulin served as a loading control. Representative results from three donors are shown,
- e SE-eRNA:NFATcl was measured using real-time PCR in presence or absence of I-BET151 (500 nM) after RANKL stimulation for 24 h.
- f Representative tracks for nuclear seq of RA synovial CD 14+ cells in vicinity of PRDM1 and MYC (RA: RA synovial OCPs, CTRL: disease control).
- compositions provided herein are useful in therapies for treating rheumatoid arthritis, osteoporosis, bone and joint destruction associated disorders, and other disorders associated with osteoclastogenesis.
- SEs SE-rcsponsivc human ostcoclast- specific super enhancers
- SE-eRNAs SE-associated enhancer RNAs
- RANKL induced 200 SEs, large clusters of enhancers, while suppressing 148 SEs compared to macrophages.
- RANKL-responsive SEs are strongly correlated with genes in the osteoclastogenic program and are increased selectively in human osteoclasts but are marginally presented in osteoblasts, CD4+ T cells, or CD34+ cells.
- BATE binding motifs were found to be highly enriched in RANKL-responsive SEs.
- increased chromatin accessibility was found within SE regions, where RNA polymerase II was significantly recruited to induce extragenic transcription of SE-eRNAs in human osteoclasts.
- Knockdown of SE-eRNAs in the vicinity of NFATcl gene diminished expression of NFATcl, a major regulator of osteoclasts, and osteoclastogenesis.
- Inhibiting BET proteins suppressed the formation of RANKL-responsive SEs and NFATcl-associated SEs and the expression of SE-eRNA:NFATcl.
- SE- eRNA:NFATcl was highly expressed in synovial macrophages of rheumatoid arthritis patients exhibiting high-osteoclastogenic potential.
- the genome-wide analysis described herein identified RANKL-inducible SE-eRNAs as an osteoclast-specific signature, permitting the development of osteoclast-specific therapeutic intervention.
- the DNA sense (positive (+)) strand or its complementary strand (-), or a transcript thereof (an RNA), may be targeted by an agent as provided herein which interferes with binding to the osteoclast-specific Nuclear Factor of Activated T cells, cl (NFATcl) regulatory region.
- osteoclast-specific NFATcl refers to a sequence which, when targeted, modulates the expression of NFATcl-associated SEs, the expression of SE-eRNA:NFATcl, and/or RANKL-inducible SE-eRNAs in osteoclasts.
- the knockdown of the targeted region defined herein avoids undesirable off-target modulation of one or more of osteoblasts, CD4+ T cells and/or CD34+ cells.
- the osteoclast-specific NFATcl regulatory region in human chromosome 18 (e.g., 18q23) is targeted by an agent which interferences with its function.
- the target sequence is located in one or more of: chrl8: position 8966290 to 9255580; chrl8 position 59836471 to 60067649; and/or chrl8 position 77322750 to 77406946, or a transcript thereof.
- the agent is an ASO, RNAi, small interfering RNA (siRNA), microRNA (miRNA), or another interfering sequence which targets the osteoclastspecific regulatory region.
- the ASOs are designed as gapmer ASO’s.
- “Gapmer” means an ASO comprising an internal region having a plurality of nucleosides that support RNase H cleavage positioned between external regions having one or more nucleosides, wherein the nucleosides comprising the internal region are chemically distinct from the nucleoside or nucleosides comprising the external regions.
- the internal region may be referred to as the “gap” and the external regions may be referred to as the “wings.”
- at least one antisense oligonucleotide in a composition of the invention is a gapmer.
- the ASOs and other therapeutic agents are targeted to sequences which interfere the osteoclast-specific Nuclear Factor of Activated T cells, cl (NFATcl) regulatory region.
- an ASO is selected which has a sequence (5’ to 3’) of at least 12 consecutive nucleotides of SEQ ID NO: 1 or 2, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof.
- an ASO is selected which has a sequence (5’ to 3') of at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof.
- an ASO is selected which has a sequence (5’ to 3’) of at least 18 consecutive nucleotides of SEQ ID NO: 1, or comprising at least 12 or 15 consecutive nucleotides of SEQ ID NO: 2, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof.
- an ASO is selected which has a sequence (5’ to 3’) of a nucleic acid sequence of at least 15, 20, 25, 30, 35, 40, 45, 50, consecutive nucleotides of SEQ ID NO: 1 or values therebetween, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof.
- an ASO is selected which has a sequence (5’ to 3’) of a nucleic acid sequence of at least 15, 20, 25, 30, 35, 40, 45, 50, consecutive nucleotides of SEQ ID NO: 2 or values therebetween, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof.
- an ASO is selected which has a sequence (5’ to 3’) of a nucleic acid sequence comprising at least 18 to 50, or about 20, about 25, about 30, about 35, about 40, about 45, or up to 50, consecutive nucleotides comprising SEQ ID NO: 1, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof.
- an ASO is selected which has a sequence (5’ to 3’) of a nucleic acid sequence comprising at least 18 to 50, or about 20, about 25, about 30, about 35, about 40, about 45, or up to 50, consecutive nucleotides comprising SEQ ID NO: 2, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof.
- combinations of two or more different ASOs targeted to one, two or all three of the positions identified above provided.
- the at least one agent comprises an ASO of 15 to 30 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising a sequence having at least 95% complementarity to SEQ ID NO: 1 or 2, or a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof.
- a composition and/or a therapeutic regimen comprises an ASO of 15 to 30 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising: combinations of a sequence having at least 95% complementarity to SEQ ID NO: 1 or 2, or a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof.
- a composition comprises a combination of an ASO having 100% complementarity to SEQ ID NO: 1 and an ASO having 100% complementarity to SEQ ID NO: 2.
- a composition comprises combinations of ASOs, combinations of one or more different ASOs with another agent having therapeutic effect, and/or combinations of the ASOs or another interfering agent as provided herein with gene replacement therapy and/or other therapies useful for treating osteoclastogenesis disorders, osteoporosis, joint or bone destruction, rheumatoid arthritis, or other disorders.
- an ASO or another moiety may be in the form of a pharmaceutically acceptable salt.
- pharmaceutically acceptable salts includes salts of the active compounds (agents, e.g., ASOs) that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
- base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
- pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
- acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
- Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p- tolyl- sulfonic, citric, tartaric, methanesulfonic, and the like.
- inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic or phosphorous acids and
- salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, e.g., Berge et al., Journal of Pharmaceutical Science 66: 1 - 19 (1977)).
- Certain specific compounds contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
- Other pharmaceutically acceptable carriers known to those of skill in the art are suitable. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms. In other cases, the preparation may be a lyophilized powder that is combined with buffer prior to use.
- the agent may be any suitable genetic element or chemical moiety including, e.g., an anti-sense oligonucleotide (ASO), an RNAi, or combinations thereof.
- the agent is engineered to be delivered via a viral vector or another genetic element.
- Suitable viral vectors may include, e.g., selected from a recombinant parvovirus, a recombinant lentivirus, or non-viral vector.
- a non-viral vector may be selected which comprises one or more agent(s).
- the non- viral vector is a lipid nanoparticle, lipidoid, or liposome.
- an “antisense oligonucleotide” or “ASO” means an oligonucleotide having a nucleobase sequence that is complementary to a target nucleic acid or region or segment thereof.
- An antisense oligonucleotide is specifically hybridizable to a target nucleic acid or region or segment thereof, the hybridization of which results in RNase H mediated cleavage of the target nucleic acid.
- enhancer RNAs are relatively long (about 500 bp to about 2000 bp in length) non-coding RNA (ncRNA) molecules transcribed from the DNA sequence of enhancer regions. These eRNA may function independently of orientation.
- SE refers to super enhancers.
- SE-eRNAs refers to super enhancer- associated eRNAs.
- Contiguous in the context of an oligonucleotide refers to nucleosides, nucleobases, sugar moieties, or internucleoside linkages that are immediately adjacent to each other.
- contiguous nucleobases means nucleobases that are immediately adjacent to each other in a sequence.
- “Portion” refers to a defined number of contiguous (i.e. , linked) nucleobases of a nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of a target nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of an oligomeric compound.
- nucleotide bases and/or polynucleotides that are capable of hybridizing to one another, e.g., the nucleotide sequence of such polynucleotides or one or more regions thereof matches the nucleotide sequence of another polynucleotide or one or more regions thereof when the two nucleotide sequences are aligned in opposing directions.
- Nucleobase matches or complementary nucleobases include the following pairs: adenine (A) with thymine (T), adenine (A) with uracil (U), cytosine (C) with guanine (G), and 5-methyl cytosine ( m C) with guanine (G).
- Complementary polynucleotides and/or nucleic acids need not have nucleobase complementarity at each nucleoside and may include one or more nucleobase mismatches. Accordingly, the present disclosure also includes isolated polynucleotides that are complementary to sequences as disclosed or used herein as well as those substantially similar nucleic acid sequences.
- a polynucleotide has 70%, at least 70%, 75%. at least 75%, 80%, at least 80%, 85%, at least 85%, 90%, at least 90%, 95%, at least 95%, 97%, at least 97%, 98%, at least 98%, 99%, or at least 99% or 100% complementarity with another polynucleotide or a target nucleic acid provided herein.
- polynucleotides or a polynucleotide and a target nucleic acid are “fully complementary” or “100% complementary,” such polynucleotides have nuclcobasc matches at each nucleoside without any nucleobase mismatches. Unless otherwise indicated, percent complementarity is the percent of the nucleobases of the shorter sequence that are complementary to the longer sequence.
- An ASO or RNA agent may contain one or more mismatches to the target sequence.
- the sequence as described herein contains no more than 3 mismatches. If the sequence contains mismatches to a target sequence, in some aspects, the area of mismatch is not located in the center of the region of complementarity. If the oligonucleotide contains mismatches to the target sequence, in some embodiments, the mismatch should be restricted to be within the last 5 nucleotides from either the 5'- or 3 '-end of the region of complementarity .
- Specifically hybridizable refers to a polynucleotide having a sufficient degree of complementarity between the polynucleotide and a target nucleic acid to induce a desired effect, while exhibiting minimal or no effects on non-target nucleic acids. In certain embodiments, specific hybridization occurs under physiological conditions.
- Specifically interfering refers to an agent which blocks binding of a protein to its native target, while having minimal or no effect on non-target nucleic acids.
- mismatch or “non-complementary” means a nucleobase of a first polynucleotide that is not complementary to the corresponding nucleobase of a second polynucleotide or target nucleic acid when the first and second polynucleotides are aligned.
- nucleobases including but not limited to a universal nucleobase, inosine, and hypoxanthine, are capable of hybridizing with at least one nucleobase but are still mismatched or non- complementary with respect to nucleobase to which it hybridized.
- a nucleobase of a first polynucleotide that is not capable of hybridizing to the corresponding nucleobase of a second polynucleotide or target nucleic acid when the first and second polynucleotides are aligned is a mismatch or non-complementary nucleobase.
- the agent comprises is an antisense oligonucleotide having at least one modified internucleoside linkage, sugar moiety, or nucleobase.
- one or more ASO is a chimeric oligonucleotide having a gap segment positioned between 5' and 3' wing segments.
- the gap segment of the chimeric oligonucleotide is comprised of 2'-deoxynucleotides and the wing segments are comprised of nucleotides having modified sugar moieties.
- the gap segment of the chimeric oligonucleotide consists of ten 2'- dcoxynuclcotidcs and each wing segment consists of five 2'-O-mcthoxycthyl-modificd nucleotides.
- one or more ASO comprises a modified sugar moiety is 2'- OMe or a bicyclic nucleic acid.
- An oligonucleotide, or pharmaceutically acceptable salt thereof can be chemically synthesized.
- An oligonucleotide, or pharmaceutically acceptable salt thereof can be synthesized by standard methods known in the art as further discussed below, e.g., by use of an automated DNA synthesizer, such as are commercially available from, for example, Biosearch, Applied Biosystems, Inc.
- An oligonucleotide, or pharmaceutically acceptable salt thereof, compound can be prepared using solution-phase or solid-phase organic synthesis or both.
- Organic synthesis offers the advantage that the oligonucleotide, or pharmaceutically acceptable salt thereof, comprising unnatural or alternative nucleotides can be easily prepared.
- a single-stranded oligonucleotide, or pharmaceutically acceptable salt thereof, can be prepared using solutionphase or solid-phase organic synthesis or both.
- the oligonucleotide, or contiguous nucleotide region thereof has a gapmer design or structure also referred herein merely as “gapmer.”
- a gapmer structure the oligonucleotide comprises at least three distinct structural regions a 5 '-flanking sequence (also known as a 5'-wing), a DNA core sequence (also known as a gap) and a 3'- flanking sequence (also known as a 3'-wing), in ‘5->3’ orientation.
- the 5' and 3' flanking sequences comprise at least one alternative nucleoside which is adjacent to a DNA core sequence, and can, in some aspects, comprise a contiguous stretch of 2 to 7 alternative nucleosides, or a contiguous stretch of alternative and DNA nucleosides (mixed flanking sequences comprising both alternative and DNA nucleosides).
- the length of the 5 '-flanking sequence region can be at least two nucleosides in length (e.g., at least at least 2, at least 3, at least 4, at least 5, at least 6, or more nucleosides in length).
- the length of the 3'-flanking sequence region can be at least two nucleosides in length (e.g., at least 2, at least 3, at least at least 4, at least 5, at least 6, or more nucleosides in length).
- the 5' and 3' flanking sequences can be symmetrical or asymmetrical with respect to the number of nucleosides they comprise.
- the DNA core sequence comprises about 10 nucleosides flanked by a 5' and a 3' flanking sequence each comprising about 5 nucleosides.
- the DNA core sequence comprises about 11 nucleosides flanked by a 5' and a 3' flanking sequence each comprising about 5 or about 6 nucleosides. In some aspects, the DNA core sequence comprises about 12 nucleosides flanked by a 5' sequence comprising about 5 nucleosides, and a 3' flanking sequence comprising about 6 nucleosides. In some aspects, the DNA core sequence comprises about 12 nucleosides flanked by a 5' sequence comprising about 6 nucleosides, and a 3' flanking sequence comprising about 5 nucleosides. In some aspects, the DNA core sequence comprises about 12 nucleosides flanked by a 5' and a 3' flanking sequence each comprising about 6 nucleosides.
- “2'-deoxyfuranosyl sugar- moiety” or “2'-dcoxy furanosyl sugar” means a furanosyl sugar moiety having two hydrogens at the 2'-position. 2'-deoxyfuranosyl sugar moieties may be unmodified or modified and may be substituted at positions other than the 2'-position or unsubstituted.
- a 0-D-2'-deoxyribosyl sugar moiety in the context of an oligonucleotide is an unsubstituted, unmodified 2'-deoxyfuranosyl and is found in naturally occurring deoxyribonucleic acids (DNA).
- 2'-deoxynucleoside means a nucleoside comprising 2'-H(H) furanosyl sugar moiety, as found in naturally occurring deoxyribonucleic acids (DNA).
- a 2'-deoxynucleoside may comprise a modified nucleobase or may comprise an RNA nucleobase (uracil).
- 2'-O-methoxyethyl refers to a 2'-O(CH2)2 — OCH3) in the place of the 2' — OH group of a ribosyl ring.
- a 2'-O-methoxyethyl modified sugar is a modified sugar-.
- 2'-M0E nucleoside (also 2'-O-methoxyethyl nucleoside) means a nucleoside comprising a 2'-M0E modified sugar- moiety.
- “2'-substituted nucleoside” or “2 -modified nucleoside” means a nucleoside comprising a 2'-substituted or 2'-modified sugar moiety.
- “2'-substituted” or “2 -modified” in reference to a sugar moiety means a sugar moiety comprising at least one 2'- substituent group other than H or OH.
- 5-methylcytosine means a cytosine with a methyl group attached to the 5 position.
- a 5-methyl cytosine is a modified nucleobase.
- “Overhanging nucleosides” refers to unpaired nucleotides at either or both ends of a duplex formed by hybridization of an antisense RNAi oligonucleotide and a sense RNAi oligonucleotide.
- the 5' and 3' flanking sequences, flanking the 5' and 3' ends of an ASO core sequence can comprise one or more affinity enhancing alternative nucleosides.
- the 5' and/or 3' flanking sequence comprises at least one 2'-O-methoxyethyl (MOE) nucleoside.
- the 5' and/or 3' flanking sequences contain at least two MOE nucleosides.
- the 5' flanking sequence comprises at least one, at least two, at least three, at least four, at least five, or at least six or more MOE nucleosides.
- the 5' flanking sequence comprises at least one, at least two, at least three, at least four, at least five, or at least six or more MOE nucleosides. In some aspects, both the 5' and 3' flanking sequence comprise a MOE nucleoside. In some aspects, all the nucleosides in the flanking sequences are MOE nucleosides.
- flanking sequence can comprise both MOE nucleosides and other nucleosides (mixed flanking sequence), such as DNA nucleosides and/or non-MOE alternative nucleosides, such as bicyclic nucleosides (BNAs) (e.g., LNA nucleosides (e.g., A-LNA, 5mC L-NA, G-LNA, T-LNA) or cET nucleosides), or other 2' substituted nucleosides.
- BNAs bicyclic nucleosides
- LNA nucleosides e.g., A-LNA, 5mC L-NA, G-LNA, T-LNA
- cET nucleosides e.g., cET nucleosides
- the DNA core sequence is defined as a contiguous sequence of at least 5 RNase H recruiting nucleosides (such as 5 to 16 DNA nucleosides or gapmers) flanked at the 5' and 3' end by an affinity enhancing alternative nucleoside, such as an MOE nucleoside.
- the 5' and/or 3' flanking sequence comprises at least one BNA (e.g., at least one LNA nucleoside (e.g., A-LNA, 5mC L-NA, G-LNA, T-LNA) or cET nucleoside).
- BNA e.g., at least one LNA nucleoside (e.g., A-LNA, 5mC L-NA, G-LNA, T-LNA) or cET nucleoside).
- 5' and/or 3' flanking sequence comprises at least 2 bicyclic nucleosides.
- the 5' flanking sequence comprises at least one BNA.
- both the 5' and 3' flanking sequence comprise a BNA.
- all the nucleosides in the flanking sequences are BNAs.
- flanking sequence can comprise both BNAs and other nucleosides (mixed flanking sequences), such as DNA nucleosides and/or non-BNA alternative nucleosides, such as 2' substituted nucleosides.
- DNA core sequence is defined as a contiguous sequence of at least five RNase H recruiting nucleosides (such as 5-16 DNA nucleosides) flanked at the 5' and 3' end by an affinity enhancing alternative nucleoside, such as a BNA, such as an LNA, such as beta-D-oxy-LNA.
- the 5' flank attached to the 5' end of the DNA core sequence comprises, contains, or consists of at least one alternative sugar moiety (e.g., at least three, at least four, at least five, at least six, at least seven, or more alternative sugar moieties).
- the flanking sequence comprises or consists of from 1 to 7 alternative nucleobases, such as from 2 to 6 alternative nucleobases, such as from 2 to 5 alternative nucleobases, such as from 2 to 4 alternative nucleobases, such as from 1 to 3 alternative nucleobases, such as one, two, three or four alternative nucleobases.
- the flanking sequence comprises or consists of at least one alternative internucleoside linkage (e.g., at least three, at least four, at least five, at least six, at least seven, or more alternative internucleoside linkages).
- the 3' flank attached to the 3' end of the DNA core sequence comprises, contains, or consists of at least one alternative sugar- moiety (e.g., at least three, at least four, at least five, at least six, at least seven, or more alternative sugar moieties).
- the flanking sequence comprises or consists of from 1 to 7 alternative nucleobases, such as from 2 to 6 alternative nucleobases, such as from 2 to 5 alternative nucleobases, such as from 2 to 4 alternative nucleobases, such as from 1 to 3 alternative nucleobases, such as one, two, three, or four alternative nucleobases.
- the flanking sequence comprises or consists of at least one alternative internucleoside linkage (e.g., at least three, at least four, at least five, at least six, at least seven, or more alternative internucleoside linkages).
- one or more or all of the alternative sugar moieties in the flanking sequence are 2' alternative sugar moieties.
- one or more of the 2' alternative sugar moieties in the wing regions are selected from 2'-O-alkyl-sugar moieties, 2'-O-methyl-sugar moieties, 2'-amino- sugar moieties, 2'-fluoro-sugar moieties, 2'-alkoxy-sugar moieties, MOE sugar moieties, LNA sugar moieties, arabino nucleic acid (ANA) sugar moieties, and 2'-fluoro-ANA sugar moieties.
- all the alternative nucleosides in the flanking sequences are bicyclic nucleosides.
- the bicyclic nucleosides in the flanking sequences are independently selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA, cET, and/or ENA, in either the beta-D or alpha-L configurations or combinations thereof.
- the one or more alternative internucleoside linkages in the flanking sequences are phosphorothioate internucleoside linkages.
- the phosphorothioate linkages are stereochemically pure phosphorothioate linkages.
- the phosphorothioate linkages are Sp phosphorothioate linkages.
- the phosphorothioate linkages are Rp phosphorothioate linkages.
- the alternative internucleoside linkages are 2'-alkoxy internucleoside linkages.
- the alternative internucleoside linkages are alkyl phosphate internucleoside linkages.
- ASOs or RNA are chemically linked or encapsulated in one or more ligands, moieties, or conjugates that enhance the activity, cellular distribution, or cellular uptake of the oligonucleotide.
- moieties include but arc not limited to lipid moieties such as a cholesterol moiety, an aliphatic chain, e.g., dodecandiol or undecyl residues, a polyamine or a polyethylene glycol chain, or the like.
- oligonucleotides used in the conjugates can be conveniently and routinely made through the well-known technique of solid-phase synthesis.
- Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.). Any other means for such synthesis known in the ait can additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides, such as the phosphorothioates and alkylated derivatives.
- compositions provided herein comprise polynucleotides (e.g., ASOs or RNA transcripts, e.g., siRNA, RNAi, or miRNA) linked or encapsulated in a lipid or lipid-like particle, polymer, or other non-viral delivery system.
- polynucleotides e.g., ASOs or RNA transcripts, e.g., siRNA, RNAi, or miRNA
- sequences may be encapsulated in a lipid nanoparticle (LNP).
- LNP lipid nanoparticle
- the phrase “lipid nanoparticle” refers to a transfer vehicle comprising one or more lipids (e.g., cationic lipids, non- cationic lipids, and PEG-modified lipids).
- the lipid nanoparticles are formulated to deliver one or more ASOs and/or siRNA or RNAi to one or more target cells.
- suitable lipids include, for example, the phosphatidyl compounds (e.g., phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides).
- phosphatidyl compounds e.g., phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides.
- polymers as transfer vehicles, whether alone or in combination with other transfer vehicles.
- Suitable polymers may include, for example, polyacrylates, polyalkycyanoacrylates, polylactide, polylactide- polyglycolide copolymers, polycaprolactones, dextran, albumin, gelatin, alginate, collagen, chitosan, cyclodextrins, dendrimers and polyethylenimine.
- the transfer vehicle is selected based upon its ability to facilitate the transfection of a target cell.
- siRNA siRNA
- cationic lipid refers to any of a number of lipid species that carry a net positive charge at a selected pH, such as physiological pH.
- the contemplated lipid nanoparticles may be prepared by including multi-component lipid mixtures of varying ratios employing one or more cationic lipids, non-cationic lipids and PEG- modified lipids.
- Several cationic lipids have been described in the literature, many of which are commercially available. See, e.g., WO2014/089486, US 2018/0353616A1, and US 8,853,377B2, which are incorporated by reference.
- LNP formulation is performed using routine procedures comprising cholesterol, ionizable lipid, helper lipid, PEG-lipid and polymer forming a lipid bilayer around encapsulated mRNA (Kowalski et al., 2019, Mol. Ther. 27(4):710-728).
- LNP comprises a cationic lipids (i.e. N-[l-(2,3- dioleoyloxy)propyl]-N.N,N-trimethylammonium chloride (DOTMA), or l,2-dioleoyl-3- trimethylammonium-propane (DOTAP)) with helper lipid DOPE.
- DOTMA N-[l-(2,3- dioleoyloxy)propyl]-N.N,N-trimethylammonium chloride
- DOTAP l,2-dioleoyl-3- trimethylammonium-propane
- LNP comprises an ionizable lipid Dlin-MC3-DMA ionizable lipids, or diketopiperazine- based ionizable lipids (cKK-E12).
- polymer comprises a polyethyleneimine (PEI), or a poly(0-amino)esters (PBAEs). See, e.g., WO2014/089486, US 2018/0353616A1, US2013/0037977A1, W02015/074085A1, US9670152B2, and US 8,853,377B2, which are incorporated by reference.
- a non-viral vector is used for delivery of ASO.
- the ASO and/or RNAi is delivered at an amount greater than about 0.5 mg/kg (e.g., greater than about 1.0 mg/kg, 1.5 mg/kg, 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg, 4.0 mg/kg, 5.0 mg/kg, 6.0 mg/kg, 7.0 mg/kg, 8.0 mg/kg, 9.0 mg/kg, or 10.0 mg/kg) body weight per dose.
- the ASO and/or RNAi is delivered at an amount ranging from about 0.1-100 mg/kg (e.g., about 0.1-90 mg/kg, 0.1-80 mg/kg, 0.1-70 mg/kg, 0.1-60 mg/kg, 0.1-50 mg/kg, 0.1-40 mg/kg, 0.1-30 mg/kg, 0.1-20 mg/kg, 0.1-10 mg/kg) body weight per dose.
- the ASO and/or RNAi is delivered at an amount of or greater than about 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 450 mg, or 500 mg per dose.
- ASOs and/or RNAi are encapsulated in a lipid nanoparticle (LNP).
- LNP lipid nanoparticle
- the phrase “lipid nanoparticle” refers to a transfer vehicle comprising one or more lipids (e.g., cationic lipids, non- cationic lipids, and PEG-modified lipids).
- the lipid nanoparticles are formulated to deliver one or more miRNA to one or more target cells (e.g., osteoclasts or other cell types identified above in the bone, joint, CNS).
- lipids include, for example, the phosphatidyl compounds (e.g., phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides). Also contemplated is the use of polymers as transfer vehicles, whether alone or in combination with other transfer vehicles.
- phosphatidyl compounds e.g., phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides.
- polymers as transfer vehicles, whether alone or in combination with other transfer vehicles.
- Suitable polymers may include, for example, polyacrylates, polyalkycyanoacrylates, polylactide, polylactide- polyglycolide copolymers, polycaprolactones, dextran, albumin, gelatin, alginate, collagen, chitosan, cyclodextrins, dendrimers and polyethylenimine.
- the transfer vehicle is selected based upon its ability to facilitate the transfection of an agent (e.g., ASO, miRNA, and/or siRNA) to a target cell.
- agent e.g., ASO, miRNA, and/or siRNA
- Useful lipid nanoparticles for and agent comprise a cationic lipid to encapsulate and/or enhance the delivery of the agent into the target cell that will act as a depot for protein production.
- cationic lipid refers to any of a number of lipid species that carry a net positive charge at a selected pH, such as physiological pH.
- the contemplated lipid nanoparticles may be prepared by including multi-component lipid mixtures of varying ratios employing one or more cationic lipids, non-cationic lipids and PEG- modified lipids.
- Several cationic lipids have been described in the literature, many of which are commercially available. See, e.g., WO2014/089486, US 2018/0353616A1, and US 8,853,377B2, which are incorporated by reference.
- LNP formulation is performed using routine procedures comprising cholesterol, ionizable lipid, helper lipid, PEG-lipid and polymer forming a lipid bilayer around encapsulated mRNA (Kowalski et al., 2019, Mol. Ther. 27(4):710-728).
- LNP comprises a cationic lipids (i.e. N-[l-(2,3- dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), or l,2-dioleoyl-3- trimethylammonium-propane (DOTAP)) with helper lipid DOPE.
- DOTMA N-[l-(2,3- dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
- DOTAP l,2-dioleoyl-3- trimethylammonium-propane
- LNP comprises an ionizable lipid Dlin-MC3-DMA ionizable lipids, or diketopiperazine- based ionizable lipids (cKK-E12).
- polymer comprises a polyethyleneimine (PEI), or a poly(P-amino)esters (PBAEs). See, e.g., WO2014/089486, US 2018/0353616A1, US2013/0037977A1, W02015/074085A1, US9670152B2, and US 8,853,377B2, which are incorporated by reference.
- one or more nucleic acid sequences provided herein may be delivered via a recombinant, replication-defective viral vector (e.g, a lentivirus, adenovirus, adeno-associated virus).
- a viral vector may further comprise one or more additional sequences for delivery to a subject.
- an agent e.g., an ASO or RNAi
- a “vector genome” refers to the nucleic acid sequence packaged inside a parvovirus (e.g., rAAV) capsid which forms a viral particle.
- a nucleic acid sequence contains AAV inverted terminal repeat sequences (ITRs).
- ITRs AAV inverted terminal repeat sequences
- a vector genome contains, at a minimum, from 5’ to 3’, an AAV 5’ ITR, coding sequence(s), and an AAV 3’ ITR.
- ITRs from AAV2, a different source AAV than the capsid, or other than full-length ITRs may be selected.
- the ITRs are from the same AAV source as the AAV which provides the rep function during production or a transcomplementing AAV. Further, other ITRs, e.g., self- complementary (scAAV) ITRs, may be used.
- the vector genome contains regulatory sequences which direct expression of the gene products. Suitable components of a vector genome are discussed in more detail herein.
- vectors for expression of polynucleotides can be accomplished using conventional techniques.
- regulatory sequences that control the expression of the polynucleotide.
- These regulatory sequences include, e.g., at least one promoter, a poly A signal, and various other vector elements (e.g., one or more of each, an enhancer, an intron, a post-translational regulatory element) and are influenced by specific cellular factors that interact with these sequences, and are well known in the art.
- an “expression cassette” refers to a nucleic acid molecule which comprises a coding sequence, promoter, and may include other regulatory sequences therefor.
- a vector genome may contain two or more expression cassettes.
- the term “transgene” may be used interchangeably with “expression cassette”.
- a therapeutically effective human dosage of viral vector is generally in the range of from about 25 to about 1000 microliters to about 100 mL of solution containing concentrations of from about 1 x 10 9 to 1 x 10 16 genomes virus vector (to treat an average subject of 70 kg in body weight) including all integers or fractional amounts within the range, and preferably 1.0 x 10 12 GC to 1.0 x 10 14 GC for a human patient.
- the compositions are formulated to contain at least IxlO 9 , 2xl0 9 , 3xl0 9 , 4xl0 9 , 5xlO 9 , 6xl0 9 , 7xl0 9 , 8xlO 9 , or 9xl0 9 GC per dose including all integers or fractional amounts within the range.
- the compositions are formulated to contain at least IxlO 10 , 2xlO 10 , 3xlO 10 , 4xlO 10 , 5xlO 10 , 6xlO 10 , 7xlO 10 , 8xlO 10 , or 9xlO 10 GC per dose including all integers or fractional amounts within the range.
- compositions are formulated to contain at least IxlO 11 , 2xlO n , 3x10", 4x10", 5xl0 n , 6x10", 7xlO n , 8xl0 n , or 9xlO n GC per dose including all integers or fractional amounts within the range.
- compositions are formulated to contain at least IxlO 12 , 2xl0 12 , 3xl0 12 , 4xl0 12 , 5xl0 12 , 6xl0 12 , 7xl0 12 , 8xl0 12 , or 9xl0 12 GC per dose including all integers or fractional amounts within the range.
- compositions are formulated to contain at least IxlO 13 , 2xl0 13 , 3xl0 13 , 4xl0 13 , 5xl0 13 , 6xl0 13 , 7xl0 13 , 8xlO 13 , or 9xl0 13 GC per dose including all integers or fractional amounts within the range.
- the dose can range from IxlO 9 to about IxlO 13 GC per dose including all integers or fractional amounts within the range.
- the vector is administered to a subject in a single dose.
- vector may be delivered via multiple injections (for example 2 doses) if desired.
- the composition for delivery may contain a buffered saline aqueous solution.
- the composition does not comprise sodium bicarbonate.
- suitable buffered saline aqueous solutions comprising one or more of sodium phosphate, sodium chloride, potassium chloride, calcium chloride, magnesium chloride and mixtures thereof, in water, such as a Harvard’s buffer.
- the aqueous solution may further contain Kolliphor® Pl 88, a poloxamer which is commercially available from BASF which was formerly sold under the trade name Lutrol® F68.
- the aqueous solution may have a pH of 7.2 or a pH of 7.4.
- the formulation may contain a buffered saline aqueous solution comprising 1 mM Sodium Phosphate (Na3PO4), 150 mM sodium chloride (NaCl), 3mM potassium chloride (KC1), 1.4 mM calcium chloride (CaC12), 0.8 mM magnesium chloride (MgC12), and 0.001% Kolliphor® 188. See, e.g., harvardapparatus.com/harvard-apparatus-perfusion-fluid.html. In certain embodiments, Harvard’s buffer is preferred. In other embodiments, the formulation may contain one or more permeation enhancers.
- Suitable permeation enhancers may include, e.g., mannitol, sodium glycocholate, sodium taurocholate, sodium deoxycholate, sodium salicylate, sodium caprylate, sodium caprate, sodium lauryl sulfate, polyoxyethylene-9-laurel ether, or EDTA.
- the composition includes a carrier, diluent, excipient and/or adjuvant.
- Suitable carriers may be readily selected by one of skill in the art in view of the indication for which the transfer vims is directed.
- one suitable carrier includes saline, which may be formulated with a variety of buffering solutions (c.g., phosphate buffered saline).
- Other exemplary carriers include sterile saline, lactose, sucrose, calcium phosphate, gelatin, dextran, agar, pectin, peanut oil, sesame oil, and water.
- the buffer/carrier should include a component that prevents the rAAV, from sticking to the infusion tubing but does not interfere with the rAAV binding activity in vivo.
- compositions may contain, in addition to the vector and carrier(s), other conventional pharmaceutical ingredients, such as preservatives, or chemical stabilizers.
- preservatives include chlorobutanol, potassium sorbate, sorbic acid, sulfur dioxide, propyl gallate, the parabens, ethyl vanillin, glycerin, phenol, and parachlorophenol.
- chemical stabilizers include gelatin and albumin.
- carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
- carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
- Supplementary active ingredients can also be incorporated into the compositions.
- pharmaceutically-acceptable refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a host.
- Delivery vehicles such as liposomes, nanocapsules, microparticles, microspheres, lipid particles, vesicles, and the like, may be used for the introduction of the compositions of the present invention into suitable host cells.
- the rAAV vector delivered transgenes may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like.
- a composition in one embodiment, includes a final formulation suitable for delivery to a subject, e.g., is an aqueous liquid suspension buffered to a physiologically compatible pH and salt concentration.
- one or more surfactants are present in the formulation.
- the composition may be transported as a concentrate which is diluted for administration to a subject.
- the composition may be lyophilized and reconstituted at the time of administration.
- a method for treating a human with an osteoclastogenesis- related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis is provided.
- use of a composition as described herein in preparing a medicament is provided.
- a composition suitable for treating an osteoclastogenesis-related disease, bone or joint destruction, bonc-rclatcd impaired mobility, osteoporosis, and/or rheumatoid arthritis is provided.
- the composition(s) comprises use of at least one agent which specifically binding to a human osteoclast-specific NFATcl regulatory region (e.g., the target sequence is located in one or more of: chrl8: position 8966290 to 9255580; chrl8 position 59836471 to 60067649; and/or chr!8 position 77322750 to 77406946, or a transcript thereof).
- the at least one agent is at least one anti-sense oligonucleotide, at least one RNAi, at least one siRNA, or combinations thereof.
- the agent is delivered via a viral vector (e.g., AAV) or a non-viral vector or other carrier, e.g., a lipid nanoparticle, lipidoid, or liposome.
- the at least one agent is at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising: (a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof; (b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof; (c) a sequence comprising a sequence having at least 95% complementarity to SEQ ID NO: 1 or 2, or a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof
- the method comprises use of at least one antisense oligonucleotide comprises 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO: 1. In certain embodiments, the method comprises use of at least one antisense oligonucleotide comprises 18 to 30 nucleotides in length which comprises SEQ ID NO: 2. In certain embodiments, the method comprises use of an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2. In certain embodiments, method comprises use of an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2.
- composition as provided herein is delivered intravenously, via site-specific administration to a joint, via targeted delivery to the bone, or intrathecally.
- operably linked refers to both expression control sequences that arc contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.
- heterologous when used with reference to a protein or a nucleic acid indicates that the protein or the nucleic acid comprises two or more sequences or subsequences which are not found in the same relationship to each other in nature.
- the nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged to make a new functional nucleic acid.
- the nucleic acid has a promoter from one gene arranged to direct the expression of a coding sequence from a different gene.
- the promoter is heterologous.
- Identity or similarity with respect to a sequence is defined herein as the percentage of amino acid residues in the candidate sequence that are identical (i.e., same residue) or similar (i.e. , amino acid residue from the same group based on common side-chain properties, see below) with the peptide and polypeptide regions provided herein, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity.
- Percent (%) identity is a measure of the relationship between two polynucleotides or two polypeptides, as determined by comparing their nucleotide or amino acid sequences, respectively. In general, the two sequences to be compared are aligned to give a maximum correlation between the sequences.
- the alignment of the two sequences is examined and the number of positions giving an exact amino acid or nucleotide correspondence between the two sequences determined, divided by the total length of the alignment and multiplied by 100 to give a % identity figure.
- This % identity figure may be determined over the whole length of the sequences to be compared, which is particularly suitable for sequences of the same or very similar length and which are highly homologous, or over shorter defined lengths, which is more suitable for sequences of unequal length or which have a lower level of homology.
- algorithms, and computer programs based thereon which are available to be used the literature and/or publicly or commercially available for performing alignments and percent identity. The selection of the algorithm or program is not a limitation.
- suitable alignment programs including, e.g., the software CLUSTALW under Unix and then be imported into the Bioedit program (Hall, T. A. 1999, BioEdit: a user- friendly biological sequence alignment editor and analysis program for Windows 95/98/NT. Nucl. Acids. Symp. Scr. 41:95-98); the Clustal Omega available from EMBL-EBI (Sievers, Fabian, et al. “Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega.” Molecular systems biology 7.1 (2011): 539 and Goujon, Mickael, et al. “A new bioinformatics analysis tools framework at EMBL-EBI.” Nucleic acids research 38.
- a composition comprising: a pharmaceutically acceptable delivery vehicle, carrier, and/or diluent, and at least one nucleic acid of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
- TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- ATTCAGCTCCACTTA SEQ ID NO: 2
- SEQ ID NO: 2 ATTCAGCTCCACTTA, a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- composition according to embodiment 1, wherein at least one nucleic acid comprises 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO:
- composition according to embodiment 1, wherein at least one nucleic acid comprises 18 to 30 nucleotides in length which comprise at least 15 consecutive nucleotides of SEQ ID NO: 2.
- composition of embodiment 1 or 2 wherein said composition comprises an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2.
- composition of embodiment 1 or 2 wherein said composition comprises an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2.
- composition of any one of embodiments 1 to 5 wherein the nucleic acid is an antisense oligonucleotide having at least one modified internucleoside linkage, sugar moiety, or nucleobase.
- composition according to any one of embodiments 1 to 8, wherein the composition comprises a non-viral vector comprising the nucleic acid which is a lipid nanoparticle, lipidoid, or liposome.
- a method for treating an osteoclastogenesis-related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis comprising delivering a nucleic acid that specifically binds to a human osteoclastspecific NFATcl regulatory region in chromosome 18.
- nucleic acid is an anti-sense oligonucleotide, an RNAi, siRNA, or combinations thereof.
- nucleic acid is delivered via a lipid nanoparticle, lipidoid, or liposome.
- nucleic acid comprises at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
- TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- ATTCAGCTCCACTTA SEQ ID NO: 2
- SEQ ID NO: 2 ATTCAGCTCCACTTA, a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- At least one antisense oligonucleotide comprises 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO:
- At least one antisense oligonucleotide comprises 18 to 30 nucleotides in length which comprises SEQ ID NO: 2.
- composition comprises an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2.
- composition comprises an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2.
- composition is delivered intravenously, via site-specific administration to a joint, via targeted delivery to the bone, or intrathecally.
- ASO antisense oligonucleotide
- TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- ATTCAGCTCCACTTA SEQ ID NO: 2
- SEQ ID NO: 2 ATTCAGCTCCACTTA, a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- a composition suitable for treatment of an osteoclastogenesis-related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis comprising at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
- TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- ATTCAGCTCCACTTA SEQ ID NO: 2
- SEQ ID NO: 2 ATTCAGCTCCACTTA, a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
- the following examples investigated the super enhancer (SE) landscape during human osteoclastogenesis and defined RANKL-responsive SEs and SE-associated eRNAs in human osteoclasts.
- SE super enhancer
- BATE Basic Leucine Zipper ATF-Like Transcription Factor
- BATF1/3 new positive regulators of osteoclasts.
- SE-eRNAs eRNAs transcribed from superenhancers
- RANKL-responsive SE- eRNAs were found in RA synovial macrophages that are prone to form osteoclasts.
- PPARGC1B Peroxisome proliferator-activated receptor gamma coactivator 1-beta PRDM1 PR/SET Domain 1 Pro-seq Precision nuclear run-on and sequencing RA Rheumatoid arthritis
- PBMCs Peripheral blood mononuclear cells
- CD14 + cells were obtained by isolation using anti-CD14 magnetic beads, as recommended by the manufacturer (Miltenyi Biotec, CA). Human CD14 + cells were cultured in a-MEM medium (Invitrogen) supplemented with 10 % fetal bovine scrum (FBS, Hyclonc; SH30070.03) and 1% L-glutamin with 20 ng/ml of M-CSF for 24 hours to generate osteoclast precursor cells (OCPs) The purity of monocytes was >97%, as verified by flow cytometric analysis 22 .
- FBS fetal bovine scrum
- OCPs osteoclast precursor cells
- RNA free RNA was obtained using the RNeasy Mini Kit from QIAGEN with DNase treatment, and 0.5 mg of total RNA was reverse transcribed using a First Strand cDNA Synthesis kit (Fermentas, Hanover, MD).
- Real-time PCR was performed in triplicate using the iCycler iQ thermal cycler and detection system (Applied Biosystems, Carlsbad, CA) following the manufacturer’s protocols. Expression of the tested gene was normalized relative to the levels of TBP.
- the primer sequences are listed in the following Table.
- TRAP staining Cells were fixed and stained for TRAP using the Acid Phosphatase Leukocyte diagnostic kit (Sigma) as recommended by the manufacturer. Nuclei was stained with methyl green. Multinucleated (more than three nuclei), TRAP-positive osteoclasts were counted in triplicate wells.
- RNA Interference 0.2 nmol of antisense LNAs (LNATM GapmeR) or siRNAs (BATF1 and BATF3) were designed and synthesized by Qiagen (Exiqon) and Ambion, respectively. Knock down of human Se-RNA; NF ATcl or control siRNA were transfected into primary human CD14 + monocytes with the Amaxa Nucleofector device set to program Y-001 using the Human Monocyte Nucleofector kit (Amaxa), as previously described 54 .
- Fluorescence in situ hybridization Fluorescence-conjugated Se- RNA:NFATcl probes for RNA-FISH were generated according to protocols of Biosearch Technologies. Hybridization was carried according to the protocol of Biosearch Technologies. The stained cells were imaged using a Zeiss Axioplan microscope (Zeiss) with an attached Leica DC 200 digital camera (Leica).
- ChlP-sequencing Cells were crosslinked for 5 min at room temperature by the addition of one tenth of the volume of 11% formaldehyde solution (11% formaldehyde, 50 mM HEPES, pH 7.5, 100 mM NaCl, 1 mM EDTA, pH 8.0, and 0.5 mM EGTA, pH 8.0) to the growth medium, followed by 5 min of quenching with 100 mM glycine. Cells were pelleted at 4 °C and washed with ice-cold PBS.
- 11% formaldehyde solution 11% formaldehyde, 50 mM HEPES, pH 7.5, 100 mM NaCl, 1 mM EDTA, pH 8.0, and 0.5 mM EGTA, pH 8.0
- the crosslinked cells were lysed with lysis buffer (50 mM HEPES-KOH, pH 7.5, 140 mM NaCl, 1 mM EDTA, 10% glycerol, 0.5% NP-40 and 0.25% Triton X-100) with protease inhibitors on ice for 10 min and were washed with washing buffer (10 mM Tris-HCl, pH 8.0, 200 mM NaCl, 1 mM EDTA and 0.5 mM EGTA) for 10 min.
- lysis buffer 50 mM HEPES-KOH, pH 7.5, 140 mM NaCl, 1 mM EDTA, 10% glycerol, 0.5% NP-40 and 0.25% Triton X-100
- the lysis samples were resuspended and sonicated in sonication buffer (10 mM Tris-HCl, pH 8.0, 100 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 0.1% sodium deoxy cholate and 0.5% N-lauroylsarcosinc) using a Bioruptor (Diagcnodc) with 30 s on and 30 s off on a high-power output for 12 cycles. After sonication, samples were centrifuged at 12,000 r.p.m. for 10 min at 4 °C, and 1% of sonicated cell extracts was saved as input.
- sonication buffer 10 mM Tris-HCl, pH 8.0, 100 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 0.1% sodium deoxy cholate and 0.5% N-lauroylsarcosinc
- Precleared extracts were then incubated with 5 mg of H3K27ac antibody (ab4729) and RNA polymerase II (pol II, MMS-126R) from abeam overnight at 4 °C. After overnight incubation, antibody-bound agarose beads were washed twice with sonication buffer, once with sonication buffer with 500 mM NaCl, once with LiCl wash buffer (10 mM Tris- HC1, pH 8.0, 1 mM EDTA, 250 mM LiCl and 1% NP-40) and once with TE with 50 mM NaCl. After washing, DNA was eluted in freshly prepared elution buffer (1% SDS and 0.1 M NaHCO3).
- RNA and protein were digested using RNase A and proteinase K, respectively, and DNA was purified with ChIP DNA Clean & Concentrator (Zymo Research). 10 ng of purified immunoprecipitated DNA per sample was ligated with adaptors, and 100-300 bp DNA fragments were purified to prepare DNA libraries using an Illumina TruSeq ChIP Library Prep Kit following the manufacturer’s instructions. ChIP libraries were sequenced (50 bp single end reads) using an Illumina HiSeq 2500 Sequencer at the Weill Cornell Medicine Epigenomic Core Facility per the manufacturer’s recommended protocol.
- ATAC-sequencing ATAC-seq was performed as previously described. To prepare nuclei, we spun 50,000 cells at 500g for 5 min, which was followed by a wash using 50 mL of cold lx PBS and centrifugation at 500g for 5 min. Cells were lysed using cold lysis buffer (10 mM Tris-HCl, pH 7.4, 10 mM NaCl, 3 mM MgC12 and 0.1% IGEPAL CA-630). Immediately after lysis, nuclei were spun at 500g for 10 min in a refrigerated centrifuge. Immediately following the nuclei prep, the pellet was resuspended in the transposase reaction mix (Illumina).
- the transposition reaction was carried out for 30 min at 37 °C. Directly following transposition, the sample was purified using a Qiagen MinElute kit. Then, we amplified library fragments using lx NEB next PCR master mix and custom Nextera PCR primers as previously described, using the following PCR conditions: 72 °C for 5 min; 98 °C for 30 s; and thermocycling at 98 °C for 10s, 63 °C for 30s and 72 °C for Imin. The libraries were purified using a Qiagen PCR cleanup kit yielding a final library concentration of ⁇ 30 nM in 20 ml. Libraries were amplified for a total of 10-13 cycles and were subjected to high-throughput sequencing using the Illumina HiSeq 2500 Sequencer (single end).
- ChlP-seq and ATAC-seq analysis Quality of reads was assessed with FastQC (vO.11.9). Low-quality reads were filtered with skewer (v.0.2.2). Reads were then mapped to reference genome GRCh37 (hgl9) with bowtic2 (v2.3.5.1) with “—very-sensitive” parameters. Ambiguous alignments were filtered out with samtools (vl.10) “view -q 10", then the alignments were sorted according to the coordinates with samtools sort. Definition of super- or typical enhancers was performed with HOMER “findPeaks -style super - typical”.
- HOMER annotatePeaks.pl was run with “-hist -size given -d” options.
- the alignment files were processed to bedGraph files with bedtools (v2.27.1) genomecov, then converted to bigWig format with bedSort and bedGraphToBigWig (UCSC tools), then uploaded to UCSC genome browser.
- HOMER annotatePeaks.pl was used to assign each interval to its nearest gene, whose list is then presented as a Chow-Ruskey plot with R “vennerable” package. ChlP-seq read count at enhancer regions is measured with bedtools multicov, then normalized to RPKM with sequencing depth and interval length. Enhancers' “sensitivity to IBET” was determined with HOMER getDifferentialPeaks with -F 1.5 option.
- PRO-sequencing Cells were incubated in the nuclear’ run-on reaction condition (5 mM Tris-HCl pH 8.0, 2.5 mM MgCh, 0.5 mM DTT, 150 mM KC1, 0.5% Sarkosyl, 0.4 units / pl of RNase inhibitor) with biotin-NTPs and rNTPs supplied (18.75 pM rATP, 18.75 pM rGTP, 1.875 pM biotin- 11 -CTP, 1.875 pM biotin- 11 -UTP for uPRO; 18.75 pM rATP, 18.75 pM rGTP, 18.75 pM rUTP, 0.75 pM CTP, 7.5 pM biotin-11-CTP for pChRO) for 5 min at 37°C.
- Run-On RNA was extracted using TRIzol, and fragmented under 0.2 N NaOH for 15 min on ice. Fragmented RNA was neutralized, and buffer exchanged by passing through P- 30 columns (Biorad). 3' RNA adaptor is ligated at 5 pM concentration for 1 hours at room temperature using T4 RNA ligase (NEB), followed by 2 consecutive streptavidin bead bindings and extractions. Extracted RNA is converted to cDNA using template switch reverse transcription with 1 pM RPl-short RT primer, 3.75 pM RTP-Template Switch Oligo, lx Template Switch Enzyme and Buffer (NEB) at 42°C for 30 min.
- NEB RNA ligase
- the cDNA is PCR amplified up to 20 cycles using primers compatible with Illumina Small RNA sequencing.
- Low quality reads along with Illumina small RNA 3’ adapter were filtered.
- PRO-seq reads were aligned to hgl9 genome with STAR aligner with default options.
- 5 ’-end positions of the alignments upon each strand were summarized to bedGraph tracks with bedtools genomecov -5 -strand + (or -), then converted to bigWig formatted files.
- the bigWig files, along with pre-trained support vector matrix (SVM) that is provided with the program, were run as inputs to the dREG program.
- SVM support vector matrix
- EXAMPLE 2 RANKL regulates SEs in the vicinity of master osteoclast regulators in human osteoclast
- RANKL is a key driver to induce human osteoclast differentiation 22 .
- Osteoclasts are defined as TRAP-positive multinuclear cells (> 3 nuclei). Phalloidin staining showed actin ring formation of osteoclasts.
- SEs superenhancers
- SEs are strongly associated with the expression of master transcription factors (TFs) that control cell identity and state 25,27 .
- TFs master transcription factors
- RANKL- induced SEs were located in the vicinity of NFATC1, a master regulator of ostcoclastogcncsis 16 , and key regulators of osteoclast differentiation such as MYC 55 , PR/SET Domain 1 (PRDM1) 56 , and matrix metalloproteinase-9 (MMP9) 57 (FIGs.lE-F ).
- H3K27ac read density near negative regulators such as interferon regulatory factor 8 (IRF8) 58 , Kruppel Like factor 2 (KLF2) 59 , and TNF receptor superfamily member IB (TNFRSF1B) 60 was decreased in response to RANKL (FIGs.le-f).
- IRF8 interferon regulatory factor 8
- KLF2 Kruppel Like factor 2
- TNFRSF1B TNF receptor superfamily member IB
- EXAMPLE 3 RANKL-induced SEs are osteoclast-specific and mark osteoclast identity genes
- osteoblast-specific SEs were associated with the genes encoding runt-related transcription factor 2 (RUNX2) and periostin (POSTN), which are required for osteoblast differentiation (FIG. 2d).
- RUNX2 runt-related transcription factor 2
- POSTN periostin
- FIG. 9 Gene ontology analysis using the set of genes associated with SEs in osteoblasts and CD4+ T cells showed that significant biological process terms were strongly descriptive of cell-type specific function (FIG. 9). This result suggests that the genes associated with RANKL-induced SEs are highly cell-type specific relative to TE-associated genes.
- Active enhancers are cis-regulatory elements that modulate the quality and quantity of gene expression 24 . These regions are characterized primarily by specific combinations of posttranslational modifications of histone proteins (H3K27ac and H3K4mel) and “open” chromatins where major TFs can bind 31 .
- Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) was performed to investigate chromatin changes during osteoclast differentiation. The ATAC-Seq peaks were binned according to their respective fold changes in response to RANKL (FIG. 3A). Increased chromatin accessibility showed both higher fold change (FIG.
- TF binding motifs were investigated in open chromatin within RANKL-regulated SEs.
- Open chromatin within RANKL-induced SEs were significantly enriched for TF motifs bound by BATF, activator protein-1 (AP-1), NFAT:AP-1, NF AT, and NF-kB, which has shown to be an important TF for osteoclast differentiation 2 (FIG. 3E).
- Sequence motifs bound by TFs regulating osteoclast differentiation such as PU.1 :IRF8, MAF BZIP transcription factor A (MAFA), and IRF8, were enriched in down-regulated ATAC-seq peaks within SEs (FIG. 3F).
- BET protein inhibition differentially regulates SE-eRNA expression BET protein inhibition effectively suppresses osteoclastogenesis and arthritic bone erosion 22 .
- Bromodomain-containing protein 4 (BRD4) occupancy at SEs in other cell types is highly sensitive to BET inhibition 26 , 1-BET151 -mediated suppression of osteoclastogenesis was assessed to determine if it results from disturbing SE formation which drives target gene expression in human osteoclasts.
- H3K27ac ChlP-seq was performed in the presence of I- BET151 and examined intensity of H3K27ac signals in pre-defined osteoclast-specific SEs. I-BET treatment significantly decreased H3K27ac signals in RANKL-induced SEs (FIG.4a).
- I-BET 151 treatment was found to not cause a decrease of H3K27ac in all RANKL-induced SEs, whereas significant changes in 49 out of 200 SEs contributed to an overall decrease of H3K27ac read densities in RANKL-induced SEs (FIGs.4b-c).
- SEs in the vicinity of NFATC1 and peroxisome proliferator-activated receptor gamma coactivator 1-bcta (PPARGC1B) genes were highly sensitive to I-BET151, while SEs near integrin subunit alpha V (ITGAV) were relatively resistant to inhibitors (FIG.4d).
- IBET151 Concomitantly, the expression of NFATcl and PPARGC1B were suppressed by I-BET151 treatment while I-BET151 had a minimal effect on ITGAV (FIG.10a). Motif analysis further showed that IBET-sensitive H3K27ac peaks within RANKL-induced SEs were enriched for TF motifs bound by AP-1, activating transcription factor 3 (ATF3), and BATF (FIG.4e), which are similar to the results of motif enrichment derived from chromatin accessibility (FIG.3g). IBET-insensitive H3K27ac peaks were also enriched for PU.l family motifs (FIG.4e).
- BATF belongs to the AP-1 family and consists of three members: BATF, BATF2, and BATF3 62 M .
- BATF1 was diminished initially and increased later, while BATF3 was induced by RANKL during osteoclastogenesis (FIG.10c). However, BATF2 was not detected (data not shown). It has been shown that BATF1 and BATF3 compensate each other 64 .
- both BATF1 and BATF3 were knocked-down using small interfering RNAs (siRNAs) in CD14+ cells. KD cells were cultured with M-CSF and RANKL to differentiate into osteoclasts.
- siRNAs small interfering RNAs
- ChlP-seq and RNA-seq were performed to investigate how Pol II recruitment and eRNA expression are associated with SEs and TEs across the human osteoclast genome.
- Pol II was found to be enriched in overall enhancers in human osteoclasts (FIG.5a).
- RANKL-induccd SEs relative to TEs, were highly occupied by a large portion of Pol II (FIGs.5b-c).
- the evidence that the high level of Pol II is occupied within RANKL-induced SEs suggests that these large m-clcmcnt domains may promote transcription of eRNA which drive high-level expression of SE-associated genes.
- eRNA expression in osteoclasts nuclear RNA and examined nascent transcripts were isolated by Nuclear RNA (Nuc)-seq, as previously described 67 .
- Expression of eRNA was increased in 83 - 84 % RANKL-induced SEs, while 85 - 88 % down-regulated SEs showed decreased eRNA expression (FIG.5e).
- Correlated expression of SE and eRNAs included a set of gene encoding major TFs important for osteoclasts such as NFATC1 and MYC (FIG.5f).
- EXAMPLE 7 Global identification and characteristics of SE-eRNA in human osteoclast
- nascent transcripts were found to be highly enriched at NFATC1 -associated SE as well as PRDM1- and MYC-associated SE (FIG.6a, FIG. 11).
- dREG discriminative regulatory-element detection from gro-seq
- eRNA loci derived from dREG analysis were highly occupied by several marks of active enhancer including H3K27ac, open chromatin (ATAC-seq), and Pol II (FIGs.6c-d).
- RANKL stimulation dramatically increased signaling intensities of H3K27ac, Pol II ChlP-seq, and ATAC-seq, indicating that dREG analysis successfully predicted eRNA loci in both SE and TE (FIGs.6c-d).
- dREG was also found within RANKL-induccd enhancers were significantly enriched for TF motifs bound by AP-1 family such as ATF3, Fos, Fral, and BATF (FIG.6e).
- RANKL significantly increased the expression of eRNAs in both I-BET-sensitive and -insensitive SEs.
- the abundance of RANKL-induced eRNAs in I-BET-sensitive SEs was comparatively higher than in LBET insensitive SEs (FIG.6g). This result suggests that BET proteins may associate with SE- eRNA transcription by interacting with SEs during osteoclastogenesis. Taken together, these data suggest that RANKL-regulated signaling input is associated with SE-eRNA expression.
- EXAMPLE 8 SE-eRNA regulates expression of SE-associated TF in human osteoclast
- NFATcl is a master regulator of osteoclasts 15 .
- RANKL rheumatoid arthritis
- FIG. 7b Freshly isolated synovial CD14+ cells from RA patients have a higher osteoclastogenic potential and express NFATcl protein without RANKL stimulation 70 .
- knockdown of SE-eRNA:NFATCl was observed to be mediated by antisense oligonucleotides (ASO-LNA) suppressed the differentiation of human OCPs into multi-nucleated TRAP-positive cells (FIGs. 7e-f, FIGs. 12a-b, see Example 1 for details of experiment).
- ASO-LNA antisense oligonucleotides
- knockdown of SE- eRNA:NFATCl abrogated RANKL-induced expression of NFATcl mRNA (FIG. 7g, FIG. 12c) and protein (FIG. 7h, FIG. 12d) in human OCPs.
- RANKL-responsive SEs were enriched by BATF-motifs and BET proteins, which are important regulators of osteoclasts. Targeting SE-eRNAs in the proximity of the NFATcl gene suppressed NFATcl expression and osteoclastogenesis. Moreover, RANKL-responsive SE-eRNAs were found to be present in synovial macrophages from RA patients. This study provides better understanding of the cell-specific programs driving osteoclastogenesis and insights into the control of pathological bone resorption and suggests that manipulating SE-eRNAs offers a novel, osteoclast-specific therapeutic strategy for treating osteoclast-mediated bone diseases.
- CD34+ progenitor cells may share common features in the histone code with other cells, which can be retained in subsequently differentiated cells.
- the importance of shared features between progenitors and differentiated cells remains unknown.
- Human and mouse osteoclasts share key transcription factors that drive osteoclastogenesis including NFATcl 2 .
- RANKL transduces the same signaling pathways, including the NF-kB and MAPK pathways, in both human and mouse osteoclasts.
- genes proximal to SEs are important for tissue or cell identity, SEs among different species do not exhibit sequence conservation 75 , and the overall sequence similarity in non-coding areas between different species is widely variable 76,77 .
- NFATcl, AP-1, BATF and NF-kB binding motifs are top-ranked motifs in RANKL- responsive SEs.
- NFATcl, AP-1, and NF-kB the crucial contribution of NFATcl, AP-1, and NF-kB to osteoclasts is well known.
- Carey et al. showed that PU.1 and MITF coordinated the expression of essential transcription factors for mouse bone marrow derived osteoclasts and the PU.1-Eomes-MITF complex is enriched in two intronic enhancer of NFATcl in mouse osteoclasts 10,33 .
- PU.l motif was not in the top-ranked TF motifs in RANKL- responsive SEs.
- PU.l -binding motifs were found to be pre-existing in both induced and repressed RANKL-rcsponsivc SEs in control conditions (data not shown), which is consistent with the previous report showing that PU.l can reshape the chromatin landscape by occupying all open regulatory regions in multipotent myeloid precursor cells, including closed sites 78 ' 80 .
- PU.l is able to activate or repress transcription by cooperative action with other TFs. Izawa et al.
- Non-coding RNAs contribute significantly to cellular functions and differentiation 84,85 .
- the expression and role of non-coding RNAs such as micro-RNAs, long non-coding (line) RNAs, and intronic eRNAs in osteoclasts and in osteoporosis have been documented 86 ' 88 .
- non-coding RNA species have been identified in mouse and human osteoclasts, SE-eRNAs and their function in osteoclastogenesis and bone resorption are largely unexplored in human cells.
- eRNAs were found to be transcribed from SEs in human osteoclasts. Sakaguchi et al.
- eRNAs are nascent RNA transcripts which can be easily targetable.
- Rheumatoid arthritis is a chronic autoimmune disease that primarily affects the joints, along with various body systems 92 .
- Bone erosion is one of the key clinical features of RA 6 , leading to joint destruction and impaired mobility 93 ' 96 .
- Osteoclasts are the major cell type responsible for bone erosion of RA and are found at sites of structural bone damage in inflammatory arthritis 97 . Increased number and activity of osteoclasts play crucial roles in rapid bone erosion in RA 98,99 .
- Recent advances in genome wide sequencing and highdimensional molecular profiling provide insights into RA pathogenesis and the contribution of cell types and pathways to its pathogenesis.
- RA synovial CD 14+ cells also have high osteoclastogenic potential and high levels of NFATcl expression 70 .
- this study is the first study in human osteoclasts to define SEs and SE-eRNAs and will provide a better understanding of human osteoclast biology and open new therapeutic avenues for human pathological bone destruction.
- B-ATF a novel human bZIP protein that associates with members of the AP-1 transcription factor family. Oncogene 11, 2255-2265 (1995).
- Echlin, D. R., Tae, H. J., Mitin, N. & Taparowsky, E. J. B-ATF functions as a negative regulator of AP-1 mediated transcription and blocks cellular transformation by Ras and Fos. Oncogene 19, 1752-1763, doi:10.1038/sj.onc.l203491 (2000).
- TNF activates calcium-nuclear factor of activated T cells (NFAT)cl signaling pathways in human macrophages. Proceedings of the National Academy of Sciences of the United States of America 108, 1573-1578, doi: 10.1073/pnas.1010030108 (2011).
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Physical Education & Sports Medicine (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Biomedical Technology (AREA)
- Medicinal Chemistry (AREA)
- Biochemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Rheumatology (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Biophysics (AREA)
- Plant Pathology (AREA)
- Physics & Mathematics (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Compositions and uses thereof are provided for treating an osteoclastogenesis-related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis. The compositions comprise an agent which specifically binding to a human ostcoclast-spccific NFATcl regulatory region.
Description
COMPOSITIONS FOR TREATING OSTEOCLASTOGENESIS DISORDERS AND/OR RHEUMATOID ARTHRITIS
REFERENCE TO AN ELECTRONIC SEQUENCE LISTING
The contents of the electronic sequence listing (HSS-2022-009.PCT_Seq- Listing.xml; Size: 13,481 bytes; and Date of Creation: November 6, 2023) is hereby incoiporatcd by reference in its entirety.
BACKGROUND OF THE INVENTION
Osteoclasts are large, multinucleated cells derived from myeloid lineage cells and are the primary cells responsible for bone resorption and bone homeostasis1'3. Dysregulated osteoclast differentiation and function lead to bone destruction in pathological conditions, including rheumatoid arthritis (RA)4'6. Macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-B ligand (RANKL) are key regulators of osteoclast differentiation, survival, and activity of osteoclasts. RANKL binds to its receptor, RANK2. RANKL activates the canonical osteoclastogenic signaling pathways and induces osteoclastogenic transcriptional and epigenetic program during osteoclastogenesis7'11. RANKL- or RANK-deficient mice exhibit severe osteopetrosis with impaired osteoclast activity12'14. The mechanisms by which RANKL induces osteoclast differentiation have been extensively studied, and the central transcription factors that are involved in initiating and maintaining the osteoclastogenic program are well defined. Among them, Nuclear Factor of Activated T cells, cl (NFATcl) is known as a master regulator of osteoclastogenesis15, and NFATcl deficiency also leads to an osteoporotic phenotype in mice16'18. However, NFATcl and other key factors driving osteoclastogenesis also express and function in other cell types, and the cell-specific regulatory mechanisms controlling the expression of such factors in osteoclasts remain poorly characterized.
Environmental stimuli trigger immediate and/or chronic changes in gene transcription, leading to the differentiation of cells into various subsets with a distinct function and phenotype. Epigenetic regulation plays a critical role in cell-type specific gene expression that establishes physiological phenotypes of cells but also contributes to human diseases11 19,20. There have been considerable efforts to understand epigenetic regulation in
osteoclasts7'11,21. Nishikawa et al. demonstrated that RANKL-induced S-adenosylmethionine (SAM) production mediates DNA methylation by de novo DNA methyltransferase 3a (Dnmt3a) during osteoclastogenesis and that inhibiting Dnmt3a regulates osteoclastogenesis and ovariectomy-induced bone loss21. Recent studies have also shown that targeting bromodomain and extraterminal (BET) proteins, a chromatin reader, using small molecule inhibitors suppresses osteoclastogenesis and bone destruction in inflammatory arthritis22,23. However, targeting epigenetic regulators broadly affects many cell types since most cells utilize similar chromatin regulators to regulate epigenetic mechanisms. Thus, unraveling the osteoclast-specific regulatory mechanism is important for understanding the epigenetic network mediating lineage control of osteoclast formation and activity and for identifying a safe therapeutic target.
Enhancers are cis regulatory elements and contain transcription factor-binding motifs24. Enhancers derive cell-type specific transcriptional activation or suppression independent of orientation or distance24. Super-enhancers (SEs) are clusters of enhancers and underlie cell identity and lineage commitment by driving high-level expression of genes encoding key regulators of cell identity and cell fate25'29. SEs serve as hubs that recruit large protein complexes including the transcriptional apparatus, transcription factors, co-activators and chromatin regulators, and promote the expression of cell-fate decision genes30. Histone 3 acetylation at lysine 27 (H3K27ac), an active enhancer mark, is highly enriched in SEs31. SEs are also sensitive to the dynamic changes of pioneer master regulators during differentiation32. A recent study identified SEs in mouse osteoclasts using ChlP-seq of PU.l, a myeloid pioneer transcription factor33. However, human osteoclast-specific SEs have not yet been defined. Moreover, recent data has revealed that enhancers produce non-coding RNA species, called enhancer RNAs (eRNAs)34'37. eRNAs are transcribed from active tissue-specific enhancers including super-enhancers, and their biological function has been discovered in many different cells38'51. However, the function and mechanism of superenhancers as well as SE-associated eRNAs in RANKL-stimulated human osteoclasts are yet to be firmly established.
There remains a need for osteoclast-specific specific therapies for treatment of bone and joint destruction and/or rheumatoid arthritis.
SUMMARY OF THE INVENTION
The inventor has identified a novel region within the regulatory region for a Nfatcl gene which is osteoclast- specific. The identification of this region has enabled the design of various osteoclast-specific therapies, including antisense oligonucleotide (ASO) therapy. Other suitable agents may be additionally or alternatively targeted to this region.
In one embodiment, the invention provides a composition comprising: a pharmaceutically acceptable delivery vehicle, carrier, and/or diluent, and at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising a sequence having at least 95% complementarity to SEQ ID NO: 1 or 2, or a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or (d) combinations of (a), (b), or (c).
The composition comprises at least one of these antisense oligonucleotides comprising 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO: 1. In certain embodiments, the at least one antisense oligonucleotide comprises 18 to 30 nucleotides in length which comprise at least 15 consecutive nucleotides of SEQ ID NO: 2. In certain embodiments, the composition comprises an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2. In certain embodiments, the composition comprises an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2. In certain embodiments, the composition comprises an antisense oligonucleotide having at least one modified internucleoside linkage, sugar moiety, or nucleobase. In certain embodiments, the composition comprises a non- viral vector comprising the oligo is a lipid nanoparticle, lipidoid, or liposome.
In certain embodiments, a method is provided for treating an osteoclastogenesis- related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis,
and/or rheumatoid arthritis, said method comprising delivering an agent which specifically binding to a human osteoclast-specific NFATcl regulatory region in chromosome 18. In certain embodiments, the agent is an anti-sense oligonucleotide, an RNAi, siRNA, or combinations thereof. In certain embodiments, the agent is delivered via a lipid nanoparticle, lipidoid, or liposome. In certain embodiments, the composition is delivered intravenously, via sitc-spccific administration to a joint, via targeted delivery to the bone, or intrathccally.
In certain embodiments, use of at least one antisense oligonucleotide (ASO) of 15 to 50 nucleotides in length in preparing a medicament is provided. In certain embodiments, a composition is provided which is suitable for treatment of an osteoclastogenesis-related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis which comprises an agent which modulates the regulatory region of the osteoclast- specific Nfatcl gene.
These and other aspects and embodiments of the invention will be apparent from the specification.
BRIEF DESCRIPTION OF THE FIGURES
FIGs. 1A-1F provide the identification of RANKL-sensitive super enhancers (SEs) in human osteoclasts. Human CD14+ monocytes cultured overnight with M-CSF (20 ng ml-1) and then were treated with M-CSF (CTRL) or M-CSF with RANKL (40 ng ml-1, RANKL) for three days (FIG 1A) or one day (FIGS 1B-1F). FIG 1A: Schematic showing the differentiation of osteoclasts. FIGs IBb-F H3K27ac ChlP-seq was performed. (FIG IB) Distribution of H3K27ac ChlP-seq enrichment scores in the indicated conditions. Enhancer regions are plotted in an increasing order based on their input-normalized H3K27ac ChlP- seq signal. SEs are defined as the population of enhancers above the inflection point of the curve. (FIG 1C) RANKL-sensitive SE was defined as a > 1.5-fold change in H3K27ac read density by RANKL (left panel). Disease ontology for RANKL-sensitive SE-associated genes with corresponding q-value (right panel) (FIG ID; control in black, R) Read density plot of H3K27ac ChlP-seq across typical enhancers (TEs) and SEs domains. (FIG IE) Graphs showing the differential H3K27ac ChlP-seq signals between RANKL and CTRL across enhancer domains. Increased (left) or decreased (right) H3K27ac ChlP-seq signals in RANKL are shown. Dark dots indicate the signals from SE regions and gray dots indicate the signals from TE regions. Boxplots of the change in H3K27ac ChlP-seq signals in the indicated conditions. :/?<().() I by Kruskal-Wallis test with the Bonferroni correction.
Representative SEs are highlighted along with their associated genes, (f) Representative tracks of H3K27ac ChlP-seq at NFATcl, PRDM1, MYC. IRF8, and KLF2 loci in the indicated conditions. Gray or black bar indicates up-regulated or down-regulated SEs, respectively.
FIGs. 2A-2D show RANKL-sensitive SEs exhibit osteoclast-specificity, (a) H3K27ac ChlP-scq distribution for osteoclast, osteoblast, CD34+ progenitor and CD4+ T cell within RANKL-induced osteoclast SEs. Each color indicates a different cell type, (b) Box plots showing H3K27ac ChlP-seq signals across SE domains in the indicated cell type. ***; /?<().001 , n.s.: not significant by Kruskal-Wallis test with the Bonferroni correction, (c) Chow-Ruskey diagram of SE- or TE-associated genes in osteoclast, osteoblast, CD34+ progenitor and CD4+ T cell. Lighter shades represent the overlap of fewer cell types. Area of each intersection is proportional to number of genes within the intersection, (d) Representative tracks for H3K27ac ChlP-seq of the indicated cell types in vicinity of SEs of osteoclast, osteoblast, CD34+ progenitor or CD4+ T cells.
FIGs. 3A-3F illustrate dynamic chromatin accessibility at RANKL-sensitive SEs. (a) Heatmaps of differential chromatin accessibility (ATAC-seq) upon RANKL (40 ng mF1) treatment; left panel: increased ATAC-seq peaks in RANKL (a black box indicates highly inducible ATAC-seq peaks), right panel: decreased ATAC-seq peaks in RANKL. (b) Read density plot of ATAC-seq across TE and SEs domains in the indicated conditions, (c) Box plots of ATAC-seq read density at TEs and SEs in the indicated conditions. ***; ><0.01 by Kruskal- Wallis test with the Bonferroni correction, (d) Representative tracks of ATAC-seq in the vicinity of NFATcl, PRDM1 and MYC loci. Gray boxes indicate the RANKL- induced SE regions (left). Enlarged ATAC-seq tracks and peaks at SE domains are shown (right), (e, f) Motif analysis of RANKL-regulated ATAC-seq peaks within SE regions.
FIGs. 4A-4I show sensitivity to iBET differentially regulates SE-eRNA expression. FIG4A is a Box plot displaying read densities of H3K27ac ChlP-seq in RANKL-sensitive SE domains in presence or absence of LBET151 (500 nM). (b) Distribution of H3K27ac ChlP-seq signals within all RANKL-induced SEs (left, n=200) and I-BET15Lsensitive SEs that underwent 1.5-fold H3K27ac enrichment changes from 200 RANKL-sensitive SEs (right, n=49). (c) Box plots showing H3K27ac ChlP-seq signals across SE domains in the indicated conditions, (d) Representative tracks of IBET-sensitive or -insensitive SEs in the indicated conditions. Normalized read counts of H3K27ac ChlP-seq are shown, (e, f) Motif enrichment analysis of I-BET151-sensitive or insensitive SEs. (g) Overall sequencing
coverage of H3K27ac and ATAC-seq around BATF binding motifs. Dotted line, dashed line, and solid lines represent coverage around motifs genome-wide, motifs within TEs, and motifs within SEs, respectively. The black line represents the control group (CTRL) and the gray line represents the RANKL-treated group, (h, i) Osteoclastogenesis assay for human OCPs transfected with control or BATFl/3-specific siRNA and treated with RANKL (40 ng ml-1) (n=3). Scale bar: 200 pm. (h) BATF1/3 knock-down efficiency in human OCPs was measured by RT-qPCR. (i) Graph shows the percentage of TRAP-positive multinuclear cells (MNCs: more than three nuclei) per control, normalized relative to the number of osteoclasts formed in control siRNA conditions. ***; p<0.001, **; p< 0.01, or *; p< 0.05 by Kruskal- Wallis test with the Bonferroni correction (c) or by student t- test (h,i).
FIGs. 5A-5H demonstrate active RNA Pol II recruitment and transcription within RANKL-sensitive super-enhancers in human osteoclasts, (a) Heatmaps show the enrichment of RNA Pol II ChlP-seq signals in RANKL-sensitive enhancer regions, (b) Distribution of RNA Pol II ChlP-seq signals across RANKL-sensitive TEs and SE domains in the indicated conditions, (c) The box plots depict quantitation of normalized tag counts of RNA pol II ChlP-seq signals in TE and SE domains in the indicated conditions, (d) The box plots depict the quantitation of nascent transcripts of nuclear RNA-seq signals in TE and SE domains in the indicated conditions, (e) Scatter plot for transcript levels of the genes associated with RANKL-sensitive SEs depending on whether those are upregulated or downregulated. Dark dots indicate the genes related to RANKL-induced SEs, while blue dots indicate the genes related to RANKL-suppressed SEs. (g) Volcano plot of RNA-seq analysis of differentially expressed SE-associated genes. Light dots show genes associated with up-regulated SEs and dark dots show genes associated with down-regulated SEs. (g, h) Representative tracks of RNA Pol II ChlP-seq (g) and RNA-seq (h) in the vicinity of NFATcl, PRDM1, and MYC loci in the indicated conditions. Gray boxes depict the RANKL-sensitive SE region. ***; cO.OOl , n.s.: not significant by Kruskal-Wallis test with the Bonferroni correction (c, d).
FIGs. 6A-6G show identification of dREG peaks in human osteoclasts, (a) Representative tracks of pro-seq at NFATcl -associated SE domain in the indicated conditions (CTRL: controls and RANKL: RANKL treated conditions). Up-regulated dREG peaks of pro-seq are shown, (b) A pie chart showing the genomic location of RANKL- inducible dREG peaks from pro-seq data. A bar graph is depicted to number of dREG peaks at TE and SE domains, (c) Aggregate plots showing mean Pro-seq, H3K27ac, Pol II ChlP- seq, and ATAC-seq signals centered on up-regulated dREG peaks in the indicated
conditions, (d) Box plots showing Pro-seq, H3K27ac, Pol II ChlP-seq, and ATAC-seq signals centered on up-regulated dREG peaks in the indicated conditions. ***; <0.001 by Kruskal-Wallis test with the Bonferroni correction, (e, f) Motif analysis of RANKL-induced (e) and RANKL-suppressed (f) dREG peaks, (g) Box plots showing Pro-seq signals across RANKL-induced SE domains in the indicated conditions.
FIGs. 7A-7K show RANKL-induced SE-cRNAs arc enriched in synovial CD 14+ cells from RA patients, (a) Disease ontology for dREG peak-associated genes with corresponding adjusted p-value. (b, c) RT-qPCR analysis of NFATcl during osteoclastogenesis (b) or RA synovial CD 14+ cells (c), normalized by TBP mRNA. (d) Applications of fluorescence in situ hybridization (FISH) in human osteoclasts. DAPI and signals for SE-eRNA:NFATcl are shown. Scale bar: 50 pm. (e-h) The expression of SE- eRNA:NFATcl is knocked down by electroporation with antisense-LNA GapmeR in human OCPs. (n=3) (e) The expression of SE-eRNA:NFATcl was measured by RT-qPCR in the indicated conditions, (f) Osteoclastogenesis assay. Cells were subsequently cultured with M- CSF and RANKL for three days. Left panel shows representative images of TRAP-stained cells. Right panel shows the percentage of TRAP-positive multinuclear cells (MNCs: more than three nuclei) per control. Scale bar: 200 pm. (g) NFATcl mRNA were measured by RT-qPCR in the indicated conditions, (h) Immunoblot analysis of NFATcl expression by human CD14+ cells transfected with control or SE-eRNA:NFATcl -specific siRNA and treated for 24 h with RANKL. a-tubulin was served as a loading control, (i) Box plots showing pro-seq signals within RANKL-induced SE derived from 2 different RA synovial CD14+ cells (RA-1, RA-2) and CD14+ cells from healthy control (CTRL), (j) Representative tracks for nuclear seq of RA synovial CD14+ cells in vicinity of NFATcl (RA: RA synovial OCPs, CTRL: disease control), (k) SE-eRNA:NFATcl was measured using RT-qPCR (see Methods). The data are shown as means ± SEM. *: p< 0.05, **; p< 0.01, ****; p< 0.0001, n.s.: not significant by one way ANOVA (b, e, g) or by student /-test (c, f, k) or by Kruskal-Wallis test with the Bonferroni correction (i).
FIGs. 8A - 8B provides a mapping of RANKL-sensitive SEs in human osteoclasts. FIG. 8 A is a Table showing the correlation between biological replicates for H3K27ac, Pol II ChlP-seq, ATAC-seq experiments. The Pearson correlation coefficient was estimated based on the average read densities in 4kb regions around the TSS of RefSeq. FIG 8B shows gene ontology for downregulated SEs with corresponding adjusted p-value.
FIGs. 9A - 9B provides Cell-type specific SEs in osteoblast and CD4+ T cell. Gene ontology for osteoblast-specific (a) and CD4+ T cell-specific (b) SEs with corresponding adjusted p-value.
FIGs. 10A-10C provide Transcription Factors at RANKL-sensitive SEs. FIG 10A mRNA expression of NFATcl, PPARGC1B, and ITGAV obtained from publicly available data SRP047069. (b) Overall sequencing coverage of H3K27ac and ATAC-scq around Fos and NFkB binding motifs. Dotted line, dashed line, and solid lines represent coverage around motifs genome-wide, motifs within TEs, and motifs within SEs, respectively. The black line represents the control group (CTRL) and the gray line represents the RANKL-treated group. The solid line represents super-enhancer and the dotted line represents typical enhancer (TE). (c) Human BATF1 or 3 expressions are measured by real time-qPCR. Cells were cultured with RANKL for the indicated times. *: p< 0.05, **: p< 0.01, ***: p< 0.001, n.s.: not significant by Student’s /-test (a) or one way ANOVA with post-tukey test (c).
FIG. 11 provides RANKL-induced SE-eRNA expression in human osteoclasts. Representative tracks of pro-seq (upper tracks) and RNA-seq (bottom tracks, RA: RA synovial OCPs, CTRL: disease control) at PRDM-associated (a), MYC-associated (b) SE domain in the indicated conditions. Up-regulated dREG peaks of pro-seq (brown) are shown.
FIGs. 12A-F demonstrate the role of SE-eRNA:NFATcl in human osteoclasts. The expression of SE-eRNA:NFATcl was knocked down by electroporation with ASO-LNA in human OCPs. Cells were subsequently cultured with M-CSF (20ng/ml) and RANKL (40ng/ml), n=4. (a) The expression of SE-eRNA:NFATcl was measured by RT-qPCR in the indicated conditions after RANKL stimulation for 24 h. (b) Osteoclastogenesis assay. Left panel shows representative images of TRAP-stained cells. Right panel shows the percentage of TRAP positive multinuclear cells (MNCs: more than three nuclei) per control. Scale bar: 200 pm. (c) NFATcl mRNA were measured by RT-qPCR in the indicated conditions after RANKL stimulation for 24 h. (d) Immunoblot analysis of NFATcl expression in the indicated conditions after RANKL stimulation for 24 h. a-tubulin served as a loading control. Representative results from three donors are shown, (e) SE-eRNA:NFATcl was measured using real-time PCR in presence or absence of I-BET151 (500 nM) after RANKL stimulation for 24 h. (f) Representative tracks for nuclear seq of RA synovial CD 14+ cells in vicinity of PRDM1 and MYC (RA: RA synovial OCPs, CTRL: disease control). *: p< 0.05, **p< 0.01, ***: p< 0.001, ****: p< 0.0001 by one way ANOVA with post-tukey test (a,c,e) or by Student’s /-test (b).
DETAILED DESCRIPTION OF THE INVENTION
The compositions provided herein are useful in therapies for treating rheumatoid arthritis, osteoporosis, bone and joint destruction associated disorders, and other disorders associated with osteoclastogenesis.
As described in the examples below, novel RANKL-rcsponsivc human ostcoclast- specific super enhancers (SEs) and SE-associated enhancer RNAs (SE-eRNAs) have been identified using an integrated analysis of ChlP-seq, ATAC-seq, nuclear RNA-seq and Pro- seq. RANKL induced 200 SEs, large clusters of enhancers, while suppressing 148 SEs compared to macrophages. RANKL-responsive SEs are strongly correlated with genes in the osteoclastogenic program and are increased selectively in human osteoclasts but are marginally presented in osteoblasts, CD4+ T cells, or CD34+ cells. In addition to the known major transcription factors in osteoclasts, BATE binding motifs were found to be highly enriched in RANKL-responsive SEs. The depletion of BATF1/3 inhibited RANKL-induced osteoclast differentiation. Furthermore, increased chromatin accessibility was found within SE regions, where RNA polymerase II was significantly recruited to induce extragenic transcription of SE-eRNAs in human osteoclasts. Knockdown of SE-eRNAs in the vicinity of NFATcl gene diminished expression of NFATcl, a major regulator of osteoclasts, and osteoclastogenesis. Inhibiting BET proteins suppressed the formation of RANKL-responsive SEs and NFATcl-associated SEs and the expression of SE-eRNA:NFATcl. Moreover, SE- eRNA:NFATcl was highly expressed in synovial macrophages of rheumatoid arthritis patients exhibiting high-osteoclastogenic potential. The genome-wide analysis described herein identified RANKL-inducible SE-eRNAs as an osteoclast-specific signature, permitting the development of osteoclast-specific therapeutic intervention.
In certain embodiments, the DNA sense (positive (+)) strand or its complementary strand (-), or a transcript thereof (an RNA), may be targeted by an agent as provided herein which interferes with binding to the osteoclast-specific Nuclear Factor of Activated T cells, cl (NFATcl) regulatory region. In certain embodiments, osteoclast-specific NFATcl refers to a sequence which, when targeted, modulates the expression of NFATcl-associated SEs, the expression of SE-eRNA:NFATcl, and/or RANKL-inducible SE-eRNAs in osteoclasts. Suitably, the knockdown of the targeted region defined herein avoids undesirable off-target modulation of one or more of osteoblasts, CD4+ T cells and/or CD34+ cells.
In certain embodiments, the osteoclast-specific NFATcl regulatory region in human chromosome 18 (e.g., 18q23) is targeted by an agent which interferences with its function. In certain embodiments, the target sequence is located in one or more of: chrl8: position 8966290 to 9255580; chrl8 position 59836471 to 60067649; and/or chrl8 position 77322750 to 77406946, or a transcript thereof.
In certain embodiments, the agent is an ASO, RNAi, small interfering RNA (siRNA), microRNA (miRNA), or another interfering sequence which targets the osteoclastspecific regulatory region. In certain embodiments, the ASOs are designed as gapmer ASO’s.
“Gapmer” means an ASO comprising an internal region having a plurality of nucleosides that support RNase H cleavage positioned between external regions having one or more nucleosides, wherein the nucleosides comprising the internal region are chemically distinct from the nucleoside or nucleosides comprising the external regions. The internal region may be referred to as the “gap” and the external regions may be referred to as the “wings.” In certain embodiments, at least one antisense oligonucleotide in a composition of the invention is a gapmer.
In certain embodiments, the ASOs and other therapeutic agents (e.g., RNAi) described herein are targeted to sequences which interfere the osteoclast-specific Nuclear Factor of Activated T cells, cl (NFATcl) regulatory region. In certain embodiments, an ASO is selected which has a sequence (5’ to 3’) of at least 12 consecutive nucleotides of SEQ ID NO: 1 or 2, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof. In certain embodiments, an ASO is selected which has a sequence (5’ to 3') of at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof. In certain embodiments, an ASO is selected which has a sequence (5’ to 3’) of at least 18 consecutive nucleotides of SEQ ID NO: 1, or comprising at least 12 or 15 consecutive nucleotides of SEQ ID NO: 2, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof. In certain embodiments, an ASO is selected which has a sequence (5’ to 3’) of a nucleic acid sequence of at least 15, 20, 25, 30, 35, 40, 45, 50, consecutive nucleotides of SEQ ID NO: 1 or values therebetween, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof. In certain
embodiments, an ASO is selected which has a sequence (5’ to 3’) of a nucleic acid sequence of at least 15, 20, 25, 30, 35, 40, 45, 50, consecutive nucleotides of SEQ ID NO: 2 or values therebetween, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof. In certain embodiments, an ASO is selected which has a sequence (5’ to 3’) of a nucleic acid sequence comprising at least 18 to 50, or about 20, about 25, about 30, about 35, about 40, about 45, or up to 50, consecutive nucleotides comprising SEQ ID NO: 1, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof. In certain embodiments, an ASO is selected which has a sequence (5’ to 3’) of a nucleic acid sequence comprising at least 18 to 50, or about 20, about 25, about 30, about 35, about 40, about 45, or up to 50, consecutive nucleotides comprising SEQ ID NO: 2, a sequence at least 99% identical thereto, a sequence having at least 95% complementarity thereto, or a pharmaceutically acceptable salt thereof.
In certain embodiments, combinations of two or more different ASOs targeted to one, two or all three of the positions identified above provided.
In certain embodiments, the at least one agent comprises an ASO of 15 to 30 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising a sequence having at least 95% complementarity to SEQ ID NO: 1 or 2, or a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof.
In certain embodiments, a composition and/or a therapeutic regimen comprises an ASO of 15 to 30 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising: combinations of a sequence having at least 95% complementarity to SEQ ID NO: 1 or 2, or a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof.
In certain embodiments, a composition comprises a combination of an ASO having 100% complementarity to SEQ ID NO: 1 and an ASO having 100% complementarity to SEQ ID NO: 2.
In certain embodiments, a composition comprises combinations of ASOs, combinations of one or more different ASOs with another agent having therapeutic effect, and/or combinations of the ASOs or another interfering agent as provided herein with gene replacement therapy and/or other therapies useful for treating osteoclastogenesis disorders, osteoporosis, joint or bone destruction, rheumatoid arthritis, or other disorders.
As described herein, an ASO or another moiety may be in the form of a pharmaceutically acceptable salt. The term “pharmaceutically acceptable salts” includes salts of the active compounds (agents, e.g., ASOs) that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt. When the compounds contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p- tolyl- sulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, e.g., Berge et al., Journal of Pharmaceutical Science 66: 1 - 19 (1977)). Certain specific compounds contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts. Other pharmaceutically acceptable carriers known to those of skill in the art are suitable. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms. In other cases, the preparation may be a lyophilized powder that is combined with buffer prior to use.
Provided herein are additionally or alternatively, other therapeutic agents. The agent may be any suitable genetic element or chemical moiety including, e.g., an anti-sense oligonucleotide (ASO), an RNAi, or combinations thereof. In certain embodiments, the agent is engineered to be delivered via a viral vector or another genetic element. Suitable viral vectors may include, e.g., selected from a recombinant parvovirus, a recombinant lentivirus, or non-viral vector. Additionally or alternatively, a non-viral vector may be selected which
comprises one or more agent(s). In certain embodiments, the non- viral vector is a lipid nanoparticle, lipidoid, or liposome.
As used herein, an “antisense oligonucleotide” or “ASO” means an oligonucleotide having a nucleobase sequence that is complementary to a target nucleic acid or region or segment thereof. An antisense oligonucleotide is specifically hybridizable to a target nucleic acid or region or segment thereof, the hybridization of which results in RNase H mediated cleavage of the target nucleic acid.
As used herein, enhancer RNAs (eRNAs) are relatively long (about 500 bp to about 2000 bp in length) non-coding RNA (ncRNA) molecules transcribed from the DNA sequence of enhancer regions. These eRNA may function independently of orientation. The term “SE” refers to super enhancers. The term SE-eRNAs refers to super enhancer- associated eRNAs.
“Contiguous” in the context of an oligonucleotide refers to nucleosides, nucleobases, sugar moieties, or internucleoside linkages that are immediately adjacent to each other. For example, “contiguous nucleobases” means nucleobases that are immediately adjacent to each other in a sequence.
“Portion” refers to a defined number of contiguous (i.e. , linked) nucleobases of a nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of a target nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of an oligomeric compound.
The term “complementary” is used to describe the relationship between nucleotide bases and/or polynucleotides that are capable of hybridizing to one another, e.g., the nucleotide sequence of such polynucleotides or one or more regions thereof matches the nucleotide sequence of another polynucleotide or one or more regions thereof when the two nucleotide sequences are aligned in opposing directions. Nucleobase matches or complementary nucleobases, as described herein, include the following pairs: adenine (A) with thymine (T), adenine (A) with uracil (U), cytosine (C) with guanine (G), and 5-methyl cytosine (mC) with guanine (G). Complementary polynucleotides and/or nucleic acids need not have nucleobase complementarity at each nucleoside and may include one or more nucleobase mismatches. Accordingly, the present disclosure also includes isolated polynucleotides that are complementary to sequences as disclosed or used herein as well as those substantially similar nucleic acid sequences. The degree to which two polynucleotides have matching nucleobases can be expressed in terms of “percent complementarity” or
“percent complementary.” In some embodiments, a polynucleotide has 70%, at least 70%, 75%. at least 75%, 80%, at least 80%, 85%, at least 85%, 90%, at least 90%, 95%, at least 95%, 97%, at least 97%, 98%, at least 98%, 99%, or at least 99% or 100% complementarity with another polynucleotide or a target nucleic acid provided herein. In embodiments wherein two polynucleotides or a polynucleotide and a target nucleic acid are “fully complementary” or “100% complementary,” such polynucleotides have nuclcobasc matches at each nucleoside without any nucleobase mismatches. Unless otherwise indicated, percent complementarity is the percent of the nucleobases of the shorter sequence that are complementary to the longer sequence.
An ASO or RNA agent may contain one or more mismatches to the target sequence. In one aspect, the sequence as described herein contains no more than 3 mismatches. If the sequence contains mismatches to a target sequence, in some aspects, the area of mismatch is not located in the center of the region of complementarity. If the oligonucleotide contains mismatches to the target sequence, in some embodiments, the mismatch should be restricted to be within the last 5 nucleotides from either the 5'- or 3 '-end of the region of complementarity .
“Specifically hybridizable” refers to a polynucleotide having a sufficient degree of complementarity between the polynucleotide and a target nucleic acid to induce a desired effect, while exhibiting minimal or no effects on non-target nucleic acids. In certain embodiments, specific hybridization occurs under physiological conditions.
“Specifically interfering” refers to an agent which blocks binding of a protein to its native target, while having minimal or no effect on non-target nucleic acids.
“Mismatch” or “non-complementary” means a nucleobase of a first polynucleotide that is not complementary to the corresponding nucleobase of a second polynucleotide or target nucleic acid when the first and second polynucleotides are aligned. For example, nucleobases including but not limited to a universal nucleobase, inosine, and hypoxanthine, are capable of hybridizing with at least one nucleobase but are still mismatched or non- complementary with respect to nucleobase to which it hybridized. As another example, a nucleobase of a first polynucleotide that is not capable of hybridizing to the corresponding nucleobase of a second polynucleotide or target nucleic acid when the first and second polynucleotides are aligned is a mismatch or non-complementary nucleobase.
In certain embodiments, the agent comprises is an antisense oligonucleotide having at least one modified internucleoside linkage, sugar moiety, or nucleobase.
In certain embodiments, one or more ASO is a chimeric oligonucleotide having a gap segment positioned between 5' and 3' wing segments. In certain embodiments, the gap segment of the chimeric oligonucleotide is comprised of 2'-deoxynucleotides and the wing segments are comprised of nucleotides having modified sugar moieties. In certain embodiments, the gap segment of the chimeric oligonucleotide consists of ten 2'- dcoxynuclcotidcs and each wing segment consists of five 2'-O-mcthoxycthyl-modificd nucleotides.
In certain embodiments, one or more ASO comprises a modified sugar moiety is 2'- OMe or a bicyclic nucleic acid.
An oligonucleotide, or pharmaceutically acceptable salt thereof, can be chemically synthesized. An oligonucleotide, or pharmaceutically acceptable salt thereof, can be synthesized by standard methods known in the art as further discussed below, e.g., by use of an automated DNA synthesizer, such as are commercially available from, for example, Biosearch, Applied Biosystems, Inc.
An oligonucleotide, or pharmaceutically acceptable salt thereof, compound can be prepared using solution-phase or solid-phase organic synthesis or both. Organic synthesis offers the advantage that the oligonucleotide, or pharmaceutically acceptable salt thereof, comprising unnatural or alternative nucleotides can be easily prepared. A single-stranded oligonucleotide, or pharmaceutically acceptable salt thereof, can be prepared using solutionphase or solid-phase organic synthesis or both.
In some embodiments, the oligonucleotide, or contiguous nucleotide region thereof, has a gapmer design or structure also referred herein merely as “gapmer.” In a gapmer structure the oligonucleotide comprises at least three distinct structural regions a 5 '-flanking sequence (also known as a 5'-wing), a DNA core sequence (also known as a gap) and a 3'- flanking sequence (also known as a 3'-wing), in ‘5->3’ orientation. In this design, the 5' and 3' flanking sequences comprise at least one alternative nucleoside which is adjacent to a DNA core sequence, and can, in some aspects, comprise a contiguous stretch of 2 to 7 alternative nucleosides, or a contiguous stretch of alternative and DNA nucleosides (mixed flanking sequences comprising both alternative and DNA nucleosides).
The length of the 5 '-flanking sequence region can be at least two nucleosides in length (e.g., at least at least 2, at least 3, at least 4, at least 5, at least 6, or more nucleosides in length). The length of the 3'-flanking sequence region can be at least two nucleosides in length (e.g., at least 2, at least 3, at least at least 4, at least 5, at least 6, or more nucleosides
in length). The 5' and 3' flanking sequences can be symmetrical or asymmetrical with respect to the number of nucleosides they comprise. In some aspects, the DNA core sequence comprises about 10 nucleosides flanked by a 5' and a 3' flanking sequence each comprising about 5 nucleosides. In some aspects, the DNA core sequence comprises about 11 nucleosides flanked by a 5' and a 3' flanking sequence each comprising about 5 or about 6 nucleosides. In some aspects, the DNA core sequence comprises about 12 nucleosides flanked by a 5' sequence comprising about 5 nucleosides, and a 3' flanking sequence comprising about 6 nucleosides. In some aspects, the DNA core sequence comprises about 12 nucleosides flanked by a 5' sequence comprising about 6 nucleosides, and a 3' flanking sequence comprising about 5 nucleosides. In some aspects, the DNA core sequence comprises about 12 nucleosides flanked by a 5' and a 3' flanking sequence each comprising about 6 nucleosides.
“2'-deoxyfuranosyl sugar- moiety” or "2'-dcoxy furanosyl sugar” means a furanosyl sugar moiety having two hydrogens at the 2'-position. 2'-deoxyfuranosyl sugar moieties may be unmodified or modified and may be substituted at positions other than the 2'-position or unsubstituted. A 0-D-2'-deoxyribosyl sugar moiety in the context of an oligonucleotide is an unsubstituted, unmodified 2'-deoxyfuranosyl and is found in naturally occurring deoxyribonucleic acids (DNA).
“2'-deoxynucleoside” means a nucleoside comprising 2'-H(H) furanosyl sugar moiety, as found in naturally occurring deoxyribonucleic acids (DNA). In certain embodiments, a 2'-deoxynucleoside may comprise a modified nucleobase or may comprise an RNA nucleobase (uracil).
“2'-O-methoxyethyl” (also 2'-M0E) refers to a 2'-O(CH2)2 — OCH3) in the place of the 2' — OH group of a ribosyl ring. A 2'-O-methoxyethyl modified sugar is a modified sugar-.
“2'-M0E nucleoside” (also 2'-O-methoxyethyl nucleoside) means a nucleoside comprising a 2'-M0E modified sugar- moiety.
“2'-substituted nucleoside” or “2 -modified nucleoside” means a nucleoside comprising a 2'-substituted or 2'-modified sugar moiety. As used herein, “2'-substituted” or “2 -modified” in reference to a sugar moiety means a sugar moiety comprising at least one 2'- substituent group other than H or OH.
“5-methylcytosine” means a cytosine with a methyl group attached to the 5 position. A 5-methyl cytosine is a modified nucleobase.
“Overhanging nucleosides” refers to unpaired nucleotides at either or both ends of a duplex formed by hybridization of an antisense RNAi oligonucleotide and a sense RNAi oligonucleotide.
The 5' and 3' flanking sequences, flanking the 5' and 3' ends of an ASO core sequence (e.g., SEQ ID NO: 1, 2, 3 or 4), can comprise one or more affinity enhancing alternative nucleosides. In some aspects, the 5' and/or 3' flanking sequence comprises at least one 2'-O-methoxyethyl (MOE) nucleoside. In some aspects, the 5' and/or 3' flanking sequences, contain at least two MOE nucleosides. In some aspects, the 5' flanking sequence comprises at least one, at least two, at least three, at least four, at least five, or at least six or more MOE nucleosides. In some aspects, the 5' flanking sequence comprises at least one, at least two, at least three, at least four, at least five, or at least six or more MOE nucleosides. In some aspects, both the 5' and 3' flanking sequence comprise a MOE nucleoside. In some aspects, all the nucleosides in the flanking sequences are MOE nucleosides. In other aspects, the flanking sequence can comprise both MOE nucleosides and other nucleosides (mixed flanking sequence), such as DNA nucleosides and/or non-MOE alternative nucleosides, such as bicyclic nucleosides (BNAs) (e.g., LNA nucleosides (e.g., A-LNA, 5mC L-NA, G-LNA, T-LNA) or cET nucleosides), or other 2' substituted nucleosides. In this case the DNA core sequence is defined as a contiguous sequence of at least 5 RNase H recruiting nucleosides (such as 5 to 16 DNA nucleosides or gapmers) flanked at the 5' and 3' end by an affinity enhancing alternative nucleoside, such as an MOE nucleoside.
In certain embodiments, the 5' and/or 3' flanking sequence comprises at least one BNA (e.g., at least one LNA nucleoside (e.g., A-LNA, 5mC L-NA, G-LNA, T-LNA) or cET nucleoside). In some embodiments, 5' and/or 3' flanking sequence comprises at least 2 bicyclic nucleosides. In some embodiments, the 5' flanking sequence comprises at least one BNA. In some embodiments, both the 5' and 3' flanking sequence comprise a BNA. In some aspects, all the nucleosides in the flanking sequences are BNAs. In other aspects, the flanking sequence can comprise both BNAs and other nucleosides (mixed flanking sequences), such as DNA nucleosides and/or non-BNA alternative nucleosides, such as 2' substituted nucleosides. In this case the DNA core sequence is defined as a contiguous sequence of at least five RNase H recruiting nucleosides (such as 5-16 DNA nucleosides) flanked at the 5' and 3' end by an affinity enhancing alternative nucleoside, such as a BNA, such as an LNA, such as beta-D-oxy-LNA.
The 5' flank attached to the 5' end of the DNA core sequence comprises, contains, or consists of at least one alternative sugar moiety (e.g., at least three, at least four, at least five, at least six, at least seven, or more alternative sugar moieties). In some aspects, the flanking sequence comprises or consists of from 1 to 7 alternative nucleobases, such as from 2 to 6 alternative nucleobases, such as from 2 to 5 alternative nucleobases, such as from 2 to 4 alternative nucleobases, such as from 1 to 3 alternative nucleobases, such as one, two, three or four alternative nucleobases. In some aspects, the flanking sequence comprises or consists of at least one alternative internucleoside linkage (e.g., at least three, at least four, at least five, at least six, at least seven, or more alternative internucleoside linkages).
The 3' flank attached to the 3' end of the DNA core sequence comprises, contains, or consists of at least one alternative sugar- moiety (e.g., at least three, at least four, at least five, at least six, at least seven, or more alternative sugar moieties). In some aspects, the flanking sequence comprises or consists of from 1 to 7 alternative nucleobases, such as from 2 to 6 alternative nucleobases, such as from 2 to 5 alternative nucleobases, such as from 2 to 4 alternative nucleobases, such as from 1 to 3 alternative nucleobases, such as one, two, three, or four alternative nucleobases. In some aspects, the flanking sequence comprises or consists of at least one alternative internucleoside linkage (e.g., at least three, at least four, at least five, at least six, at least seven, or more alternative internucleoside linkages).
In an aspect, one or more or all of the alternative sugar moieties in the flanking sequence are 2' alternative sugar moieties.
In a further aspect, one or more of the 2' alternative sugar moieties in the wing regions are selected from 2'-O-alkyl-sugar moieties, 2'-O-methyl-sugar moieties, 2'-amino- sugar moieties, 2'-fluoro-sugar moieties, 2'-alkoxy-sugar moieties, MOE sugar moieties, LNA sugar moieties, arabino nucleic acid (ANA) sugar moieties, and 2'-fluoro-ANA sugar moieties.
In one aspect, all the alternative nucleosides in the flanking sequences are bicyclic nucleosides. In a further aspect, the bicyclic nucleosides in the flanking sequences are independently selected from the group consisting of oxy-LNA, thio-LNA, amino-LNA, cET, and/or ENA, in either the beta-D or alpha-L configurations or combinations thereof.
In some aspects, the one or more alternative internucleoside linkages in the flanking sequences are phosphorothioate internucleoside linkages. In some aspects, the phosphorothioate linkages are stereochemically pure phosphorothioate linkages. In some aspects, the phosphorothioate linkages are Sp phosphorothioate linkages. In other aspects,
the phosphorothioate linkages are Rp phosphorothioate linkages. In some aspects, the alternative internucleoside linkages are 2'-alkoxy internucleoside linkages. In other aspects, the alternative internucleoside linkages are alkyl phosphate internucleoside linkages.
In certain embodiments, ASOs or RNA are chemically linked or encapsulated in one or more ligands, moieties, or conjugates that enhance the activity, cellular distribution, or cellular uptake of the oligonucleotide. Such moieties include but arc not limited to lipid moieties such as a cholesterol moiety, an aliphatic chain, e.g., dodecandiol or undecyl residues, a polyamine or a polyethylene glycol chain, or the like.
The oligonucleotides used in the conjugates can be conveniently and routinely made through the well-known technique of solid-phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.). Any other means for such synthesis known in the ait can additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides, such as the phosphorothioates and alkylated derivatives.
In certain embodiments, compositions provided herein comprise polynucleotides (e.g., ASOs or RNA transcripts, e.g., siRNA, RNAi, or miRNA) linked or encapsulated in a lipid or lipid-like particle, polymer, or other non-viral delivery system. For example, sequences may be encapsulated in a lipid nanoparticle (LNP). As used herein, the phrase “lipid nanoparticle” refers to a transfer vehicle comprising one or more lipids (e.g., cationic lipids, non- cationic lipids, and PEG-modified lipids). Preferably, the lipid nanoparticles are formulated to deliver one or more ASOs and/or siRNA or RNAi to one or more target cells. Examples of suitable lipids include, for example, the phosphatidyl compounds (e.g., phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides). Also contemplated is the use of polymers as transfer vehicles, whether alone or in combination with other transfer vehicles. Suitable polymers may include, for example, polyacrylates, polyalkycyanoacrylates, polylactide, polylactide- polyglycolide copolymers, polycaprolactones, dextran, albumin, gelatin, alginate, collagen, chitosan, cyclodextrins, dendrimers and polyethylenimine. In one embodiment, the transfer vehicle is selected based upon its ability to facilitate the transfection of a target cell. Useful lipid nanoparticles for a cationic lipid to encapsulate and/or enhance the delivery of an siRNA (RNAi, or ASO) into the target cell. As used herein, the phrase “cationic lipid” refers to any of a number of lipid species that carry a net positive charge at a selected pH, such as physiological pH. The contemplated lipid
nanoparticles may be prepared by including multi-component lipid mixtures of varying ratios employing one or more cationic lipids, non-cationic lipids and PEG- modified lipids. Several cationic lipids have been described in the literature, many of which are commercially available. See, e.g., WO2014/089486, US 2018/0353616A1, and US 8,853,377B2, which are incorporated by reference. In certain embodiments, LNP formulation is performed using routine procedures comprising cholesterol, ionizable lipid, helper lipid, PEG-lipid and polymer forming a lipid bilayer around encapsulated mRNA (Kowalski et al., 2019, Mol. Ther. 27(4):710-728). In some embodiments, LNP comprises a cationic lipids (i.e. N-[l-(2,3- dioleoyloxy)propyl]-N.N,N-trimethylammonium chloride (DOTMA), or l,2-dioleoyl-3- trimethylammonium-propane (DOTAP)) with helper lipid DOPE. In some embodiments, LNP comprises an ionizable lipid Dlin-MC3-DMA ionizable lipids, or diketopiperazine- based ionizable lipids (cKK-E12). In some embodiments, polymer comprises a polyethyleneimine (PEI), or a poly(0-amino)esters (PBAEs). See, e.g., WO2014/089486, US 2018/0353616A1, US2013/0037977A1, W02015/074085A1, US9670152B2, and US 8,853,377B2, which are incorporated by reference.
In certain embodiment, a non-viral vector is used for delivery of ASO. In some embodiments, the ASO and/or RNAi is delivered at an amount greater than about 0.5 mg/kg (e.g., greater than about 1.0 mg/kg, 1.5 mg/kg, 2.0 mg/kg, 2.5 mg/kg, 3.0 mg/kg, 4.0 mg/kg, 5.0 mg/kg, 6.0 mg/kg, 7.0 mg/kg, 8.0 mg/kg, 9.0 mg/kg, or 10.0 mg/kg) body weight per dose. In some embodiments, the ASO and/or RNAi is delivered at an amount ranging from about 0.1-100 mg/kg (e.g., about 0.1-90 mg/kg, 0.1-80 mg/kg, 0.1-70 mg/kg, 0.1-60 mg/kg, 0.1-50 mg/kg, 0.1-40 mg/kg, 0.1-30 mg/kg, 0.1-20 mg/kg, 0.1-10 mg/kg) body weight per dose. In some embodiments, the ASO and/or RNAi is delivered at an amount of or greater than about 1 mg, 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 450 mg, or 500 mg per dose.
In certain embodiments, ASOs and/or RNAi are encapsulated in a lipid nanoparticle (LNP). As used herein, the phrase “lipid nanoparticle” refers to a transfer vehicle comprising one or more lipids (e.g., cationic lipids, non- cationic lipids, and PEG-modified lipids). Preferably, the lipid nanoparticles are formulated to deliver one or more miRNA to one or more target cells (e.g., osteoclasts or other cell types identified above in the bone, joint, CNS). Examples of suitable lipids include, for example, the phosphatidyl compounds (e.g., phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine,
sphingolipids, cerebrosides, and gangliosides). Also contemplated is the use of polymers as transfer vehicles, whether alone or in combination with other transfer vehicles. Suitable polymers may include, for example, polyacrylates, polyalkycyanoacrylates, polylactide, polylactide- polyglycolide copolymers, polycaprolactones, dextran, albumin, gelatin, alginate, collagen, chitosan, cyclodextrins, dendrimers and polyethylenimine. In one embodiment, the transfer vehicle is selected based upon its ability to facilitate the transfection of an agent (e.g., ASO, miRNA, and/or siRNA) to a target cell. Useful lipid nanoparticles for and agent comprise a cationic lipid to encapsulate and/or enhance the delivery of the agent into the target cell that will act as a depot for protein production. As used herein, the phrase “cationic lipid” refers to any of a number of lipid species that carry a net positive charge at a selected pH, such as physiological pH. The contemplated lipid nanoparticles may be prepared by including multi-component lipid mixtures of varying ratios employing one or more cationic lipids, non-cationic lipids and PEG- modified lipids. Several cationic lipids have been described in the literature, many of which are commercially available. See, e.g., WO2014/089486, US 2018/0353616A1, and US 8,853,377B2, which are incorporated by reference. In certain embodiments, LNP formulation is performed using routine procedures comprising cholesterol, ionizable lipid, helper lipid, PEG-lipid and polymer forming a lipid bilayer around encapsulated mRNA (Kowalski et al., 2019, Mol. Ther. 27(4):710-728). In some embodiments, LNP comprises a cationic lipids (i.e. N-[l-(2,3- dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA), or l,2-dioleoyl-3- trimethylammonium-propane (DOTAP)) with helper lipid DOPE. In some embodiments, LNP comprises an ionizable lipid Dlin-MC3-DMA ionizable lipids, or diketopiperazine- based ionizable lipids (cKK-E12). In some embodiments, polymer comprises a polyethyleneimine (PEI), or a poly(P-amino)esters (PBAEs). See, e.g., WO2014/089486, US 2018/0353616A1, US2013/0037977A1, W02015/074085A1, US9670152B2, and US 8,853,377B2, which are incorporated by reference.
Recombinant Viral Vectors
In some embodiments, one or more nucleic acid sequences provided herein may be delivered via a recombinant, replication-defective viral vector (e.g, a lentivirus, adenovirus, adeno-associated virus). In certain embodiments, a viral vector may further comprise one or more additional sequences for delivery to a subject.
In certain embodiments, an agent (e.g., an ASO or RNAi) is expressed from a recombinant adeno-associated virus, which has an AAV capsid and a vector genome packaged in the AAV capsid. In certain embodiments, a “vector genome” refers to the nucleic acid sequence packaged inside a parvovirus (e.g., rAAV) capsid which forms a viral particle. Such a nucleic acid sequence contains AAV inverted terminal repeat sequences (ITRs). In the examples herein, a vector genome contains, at a minimum, from 5’ to 3’, an AAV 5’ ITR, coding sequence(s), and an AAV 3’ ITR. ITRs from AAV2, a different source AAV than the capsid, or other than full-length ITRs may be selected. In certain embodiments, the ITRs are from the same AAV source as the AAV which provides the rep function during production or a transcomplementing AAV. Further, other ITRs, e.g., self- complementary (scAAV) ITRs, may be used. Further, the vector genome contains regulatory sequences which direct expression of the gene products. Suitable components of a vector genome are discussed in more detail herein.
Construction of vectors for expression of polynucleotides can be accomplished using conventional techniques. For generation of efficient expression vectors, it is necessary to have regulatory sequences that control the expression of the polynucleotide. These regulatory sequences include, e.g., at least one promoter, a poly A signal, and various other vector elements (e.g., one or more of each, an enhancer, an intron, a post-translational regulatory element) and are influenced by specific cellular factors that interact with these sequences, and are well known in the art.
As used herein, an “expression cassette” refers to a nucleic acid molecule which comprises a coding sequence, promoter, and may include other regulatory sequences therefor. In certain embodiments, a vector genome may contain two or more expression cassettes. In other embodiments, the term “transgene” may be used interchangeably with “expression cassette”.
Dosages of the vector will depend primarily on factors such as the condition being treated, the age, weight and health of the patient, and may thus vary among patients. For example, a therapeutically effective human dosage of viral vector is generally in the range of from about 25 to about 1000 microliters to about 100 mL of solution containing concentrations of from about 1 x 109 to 1 x 1016 genomes virus vector (to treat an average subject of 70 kg in body weight) including all integers or fractional amounts within the range, and preferably 1.0 x 1012 GC to 1.0 x 1014 GC for a human patient. In one embodiment, the compositions are formulated to contain at least IxlO9, 2xl09, 3xl09, 4xl09,
5xlO9, 6xl09, 7xl09, 8xlO9, or 9xl09 GC per dose including all integers or fractional amounts within the range. In another embodiment, the compositions are formulated to contain at least IxlO10, 2xlO10, 3xlO10, 4xlO10, 5xlO10, 6xlO10, 7xlO10, 8xlO10, or 9xlO10 GC per dose including all integers or fractional amounts within the range. In another embodiment, the compositions are formulated to contain at least IxlO11, 2xlOn, 3x10", 4x10", 5xl0n, 6x10", 7xlOn, 8xl0n, or 9xlOn GC per dose including all integers or fractional amounts within the range. In another embodiment, the compositions are formulated to contain at least IxlO12, 2xl012, 3xl012, 4xl012, 5xl012, 6xl012, 7xl012, 8xl012, or 9xl012 GC per dose including all integers or fractional amounts within the range. In another embodiment, the compositions are formulated to contain at least IxlO13, 2xl013, 3xl013, 4xl013, 5xl013, 6xl013, 7xl013, 8xlO13, or 9xl013 GC per dose including all integers or fractional amounts within the range. In one embodiment, for human application the dose can range from IxlO9 to about IxlO13 GC per dose including all integers or fractional amounts within the range.
In certain embodiments, the vector is administered to a subject in a single dose. In certain embodiments, vector may be delivered via multiple injections (for example 2 doses) if desired.
The composition for delivery may contain a buffered saline aqueous solution. In certain embodiments, the composition does not comprise sodium bicarbonate. Examples of suitable buffered saline aqueous solutions comprising one or more of sodium phosphate, sodium chloride, potassium chloride, calcium chloride, magnesium chloride and mixtures thereof, in water, such as a Harvard’s buffer. The aqueous solution may further contain Kolliphor® Pl 88, a poloxamer which is commercially available from BASF which was formerly sold under the trade name Lutrol® F68. The aqueous solution may have a pH of 7.2 or a pH of 7.4. In another embodiment, the formulation may contain a buffered saline aqueous solution comprising 1 mM Sodium Phosphate (Na3PO4), 150 mM sodium chloride (NaCl), 3mM potassium chloride (KC1), 1.4 mM calcium chloride (CaC12), 0.8 mM magnesium chloride (MgC12), and 0.001% Kolliphor® 188. See, e.g., harvardapparatus.com/harvard-apparatus-perfusion-fluid.html. In certain embodiments, Harvard’s buffer is preferred. In other embodiments, the formulation may contain one or more permeation enhancers. Examples of suitable permeation enhancers may include, e.g., mannitol, sodium glycocholate, sodium taurocholate, sodium deoxycholate, sodium
salicylate, sodium caprylate, sodium caprate, sodium lauryl sulfate, polyoxyethylene-9-laurel ether, or EDTA.
In another embodiment, the composition includes a carrier, diluent, excipient and/or adjuvant. Suitable carriers may be readily selected by one of skill in the art in view of the indication for which the transfer vims is directed. For example, one suitable carrier includes saline, which may be formulated with a variety of buffering solutions (c.g., phosphate buffered saline). Other exemplary carriers include sterile saline, lactose, sucrose, calcium phosphate, gelatin, dextran, agar, pectin, peanut oil, sesame oil, and water. The buffer/carrier should include a component that prevents the rAAV, from sticking to the infusion tubing but does not interfere with the rAAV binding activity in vivo.
Optionally, the compositions may contain, in addition to the vector and carrier(s), other conventional pharmaceutical ingredients, such as preservatives, or chemical stabilizers. Suitable exemplary preservatives include chlorobutanol, potassium sorbate, sorbic acid, sulfur dioxide, propyl gallate, the parabens, ethyl vanillin, glycerin, phenol, and parachlorophenol. Suitable chemical stabilizers include gelatin and albumin.
As used herein, “carrier” includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Supplementary active ingredients can also be incorporated into the compositions. The phrase “pharmaceutically-acceptable” refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a host. Delivery vehicles such as liposomes, nanocapsules, microparticles, microspheres, lipid particles, vesicles, and the like, may be used for the introduction of the compositions of the present invention into suitable host cells. In particular, the rAAV vector delivered transgenes may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like.
In one embodiment, a composition includes a final formulation suitable for delivery to a subject, e.g., is an aqueous liquid suspension buffered to a physiologically compatible pH and salt concentration. Optionally, one or more surfactants are present in the formulation. In another embodiment, the composition may be transported as a concentrate which is diluted for administration to a subject. In other embodiments, the composition may be lyophilized and reconstituted at the time of administration.
In certain embodiments, a method for treating a human with an osteoclastogenesis- related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis is provided. In certain embodiments, use of a composition as described herein in preparing a medicament is provided. In certain embodiments, a composition suitable for treating an osteoclastogenesis-related disease, bone or joint destruction, bonc-rclatcd impaired mobility, osteoporosis, and/or rheumatoid arthritis is provided.
In certain embodiments, the composition(s) comprises use of at least one agent which specifically binding to a human osteoclast-specific NFATcl regulatory region (e.g., the target sequence is located in one or more of: chrl8: position 8966290 to 9255580; chrl8 position 59836471 to 60067649; and/or chr!8 position 77322750 to 77406946, or a transcript thereof). In certain embodiments, the at least one agent is at least one anti-sense oligonucleotide, at least one RNAi, at least one siRNA, or combinations thereof. In certain embodiments, the agent is delivered via a viral vector (e.g., AAV) or a non-viral vector or other carrier, e.g., a lipid nanoparticle, lipidoid, or liposome.
Various of these agents are described herein. For example, in certain embodiments, the at least one agent is at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising: (a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof; (b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof; (c) a sequence comprising a sequence having at least 95% complementarity to SEQ ID NO: 1 or 2, or a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or (d) combinations of (a), (b), or (c). In certain embodiments, the method comprises use of at least one antisense oligonucleotide comprises 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO: 1. In certain embodiments, the method comprises use of at least one antisense oligonucleotide comprises 18 to 30 nucleotides in length which comprises SEQ ID NO: 2. In certain embodiments, the method comprises use of an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID
NO: 2. In certain embodiments, method comprises use of an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2.
In certain embodiments, a composition as provided herein is delivered intravenously, via site-specific administration to a joint, via targeted delivery to the bone, or intrathecally.
As used herein, the term “operably linked” refers to both expression control sequences that arc contiguous with the gene of interest and expression control sequences that act in trans or at a distance to control the gene of interest.
The term “heterologous” when used with reference to a protein or a nucleic acid indicates that the protein or the nucleic acid comprises two or more sequences or subsequences which are not found in the same relationship to each other in nature. For instance, the nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged to make a new functional nucleic acid. For example, in one embodiment, the nucleic acid has a promoter from one gene arranged to direct the expression of a coding sequence from a different gene. Thus, with reference to the coding sequence, the promoter is heterologous.
Identity or similarity with respect to a sequence is defined herein as the percentage of amino acid residues in the candidate sequence that are identical (i.e., same residue) or similar (i.e. , amino acid residue from the same group based on common side-chain properties, see below) with the peptide and polypeptide regions provided herein, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Percent (%) identity is a measure of the relationship between two polynucleotides or two polypeptides, as determined by comparing their nucleotide or amino acid sequences, respectively. In general, the two sequences to be compared are aligned to give a maximum correlation between the sequences. The alignment of the two sequences is examined and the number of positions giving an exact amino acid or nucleotide correspondence between the two sequences determined, divided by the total length of the alignment and multiplied by 100 to give a % identity figure. This % identity figure may be determined over the whole length of the sequences to be compared, which is particularly suitable for sequences of the same or very similar length and which are highly homologous, or over shorter defined lengths, which is more suitable for sequences of unequal length or which have a lower level of homology. There are a number of algorithms, and computer programs based thereon, which are available to be used the literature and/or publicly or commercially available for
performing alignments and percent identity. The selection of the algorithm or program is not a limitation.
Examples of suitable alignment programs including, e.g., the software CLUSTALW under Unix and then be imported into the Bioedit program (Hall, T. A. 1999, BioEdit: a user- friendly biological sequence alignment editor and analysis program for Windows 95/98/NT. Nucl. Acids. Symp. Scr. 41:95-98); the Clustal Omega available from EMBL-EBI (Sievers, Fabian, et al. “Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega.” Molecular systems biology 7.1 (2011): 539 and Goujon, Mickael, et al. “A new bioinformatics analysis tools framework at EMBL-EBI.” Nucleic acids research 38. suppl 2 (2010): W695-W699); the Wisconsin Sequence Analysis Package, version 9.1 (Devereux J. et al., Nucleic Acids Res., 12:387-395, 1984, available from Genetics Computer Group, Madison, Wis., USA). The programs BESTFIT and GAP, may be used to determine the % identity between two polynucleotides and the % identity between two polypeptide sequences.
Other programs for determining identity and/or similarity between sequences include, e.g, the BLAST family of programs available from the National Center for Biotechnology Information (NCB), Bethesda, Md., USA and accessible through the home page of the NCBI at ww w. jrebl..:?ln’.... ?ih...g A), the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used; and FASTA (Pearson W. R. and Lipman D. J., Proc. Natl. Acad. Sci. USA, 85:2444-8, 1988, available as part of the Wisconsin Sequence Analysis Package). SeqWeb Software (a web-based interface to the GCG Wisconsin Package
The term “a” or “an” refers to one or more. As such, the terms “a” (or “an”), “one or more,” and “at least one” are used interchangeably herein.
The words “comprise”, “comprises”, and “comprising” are to be interpreted inclusively rather than exclusively. The words “consist”, “consisting”, and its variants, are to be interpreted exclusively, rather than inclusively. While various embodiments in the specification are presented using “comprising” language, under other circumstances, a related embodiment is also intended to be interpreted and described using “consisting of’ or “consisting essentially of’ language.
The term “about” encompasses a variation within and including ± 10%, unless otherwise specified.
Unless defined otherwise in this specification, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art and by reference to published texts, which provide one skilled in the art with a general guide to many of the terms used in the present application.
Specific embodiments
1. A composition comprising: a pharmaceutically acceptable delivery vehicle, carrier, and/or diluent, and at least one nucleic acid of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or
(d) combinations of (a), (b), or (c).
2. The composition according to embodiment 1, wherein at least one nucleic acid comprises 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO:
1.
3. The composition according to embodiment 1, wherein at least one nucleic acid comprises 18 to 30 nucleotides in length which comprise at least 15 consecutive nucleotides of SEQ ID NO: 2.
4. The composition of embodiment 1 or 2, wherein said composition comprises an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2.
5. The composition of embodiment 1 or 2, wherein said composition comprises an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2.
6. The composition of any one of embodiments 1 to 5 wherein the nucleic acid is an antisense oligonucleotide having at least one modified internucleoside linkage, sugar moiety, or nucleobase.
7. The composition of any one of embodiments 1 to 5, wherein the nucleic acid is a chimeric oligonucleotide having a gap segment positioned between 5' and 3' wing segments.
8. The composition of embodiment 6, wherein the gap segment of the chimeric oligonucleotide is comprised of 2'-deoxynucleotides and the wing segments are comprised of nucleotides having modified sugar moieties.
9. The composition according to any one of embodiments 1 to 8, wherein the composition comprises a non-viral vector comprising the nucleic acid which is a lipid nanoparticle, lipidoid, or liposome.
10. A method for treating an osteoclastogenesis-related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis, said method comprising delivering a nucleic acid that specifically binds to a human osteoclastspecific NFATcl regulatory region in chromosome 18.
11. The method according to embodiment 10, wherein the nucleic acid is an anti-sense oligonucleotide, an RNAi, siRNA, or combinations thereof.
12. The method according to embodiment 10 or 11, wherein the nucleic acid is delivered via a lipid nanoparticle, lipidoid, or liposome.
13. The method according to any one of embodiments 10 to 12, wherein the nucleic acid comprises at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or
(d) combinations of (a), (b), or (c).
14. The method of embodiment 13, wherein at least one antisense oligonucleotide comprises 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO:
1.
15. The method of embodiment 13, wherein at least one antisense oligonucleotide comprises 18 to 30 nucleotides in length which comprises SEQ ID NO: 2.
16. The method of embodiment 13, wherein said composition comprises an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2.
17. The method of embodiment 13, wherein said composition comprises an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2.
18. The method of embodiment 13, wherein the composition is delivered intravenously, via site-specific administration to a joint, via targeted delivery to the bone, or intrathecally.
19. Use of at least one antisense oligonucleotide (ASO) of 15 to 50 nucleotides in length in preparing a medicament, wherein the ASO comprises at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or
(d) combinations of (a), (b), or (c).
20. A composition suitable for treatment of an osteoclastogenesis-related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis, said composition comprising at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or
(d) combinations of (a), (b), or (c).
EXAMPLES:
The following examples investigated the super enhancer (SE) landscape during human osteoclastogenesis and defined RANKL-responsive SEs and SE-associated eRNAs in human osteoclasts. Along with key osteoclastogenic transcription factors enriched in RANKL-responsive SEs, we found Basic Leucine Zipper ATF-Like Transcription Factor (BATE) binding motifs in RANKL-responsive SEs. Moreover, wc discovered BATF1/3 as new positive regulators of osteoclasts. We also identified eRNAs transcribed from superenhancers (named SE-eRNAs) of human osteoclasts. Furthermore, RANKL-responsive SE- eRNAs were found in RA synovial macrophages that are prone to form osteoclasts. Accordingly, knock-down of SE-eRNA in the vicinity NFATcl gene suppressed NFATcl expression and RANKL-induced osteoclastogenesis. This study offers the first evidence of the potential role of RANKL-responsive SE-eRNAs in reprograming the differentiation of myeloid cells toward human osteoclasts and provides insights into the development of hyperactive osteoclasts in the pathological setting.
AP-1 Activator protein- 1
ASO-LNA Antisense oligonucleotides
ATAC-seq Transposase- Accessible Chromatin with high-throughput sequencing ATF3 Activating Transcription Factor 3 BATE Basic Leucine Zipper ATF-Like Transcription Factor BET Bromodomain and extraterminal BRD4 Bromodomain-containing protein 4 CAGE Cap-analysis of gene expression
ChlP-seq Chromatin immunoprecipitation (ChlP)-sequencing Dnmt3a DNA methyltransferase 3a dREG Regulatory-element detection from gro-seq Elf4 Enhancer RNA E74 Like ETS Transcription Factor 4 FBS Fetal Fetal bovine serum Fral Fos-related antigen 1
H3K27ac Histone 3 acetylation at lysine 27
H3K4mel Histone 3 mono-methylation at lysine 4
IRF8 Interferon Regulatory Factor 8
ITGAV Integrin Subunit Alpha V KLF2 Kruppel Like Factor 2 MAFA MAF BZIP Transcription Factor A
MCSF Macrophage colony stimulating factor
MMP9 Matrix metalloproteinase-9
NFATcl Nuclear factor of activated T cells, cl
Nuc-seq Nuclear RNA sequencing
OVX Ovariectomy
PBMC Peripheral blood mononuclear cells
Pol II RNA polymerase II
POSTN Periostin
PPARGC1B Peroxisome proliferator-activated receptor gamma coactivator 1-beta PRDM1 PR/SET Domain 1 Pro-seq Precision nuclear run-on and sequencing RA Rheumatoid arthritis
RANKL Receptor activator of NF-KB ligand
ROSE algorithm The rank ordering of super-enhancer algorithm
RUNX2 Runt-related transcription factor 2
SAM S-adenosylmethionine
SE Super-enhancer
SE-eRNA Super-enhancer-associated enhancer RNA
SF Human synovial fluid siRNAs Small interfering RNAs
TE Typical enhancers
TF Transcription factors
TNFRSF1B TNF receptor superfamily member IB TRAP Tartrate -resistant acid phosphatase
EXAMPLE 1 : Materials and methods
Human studies Human synovial fluid (SF) samples were collected from RA patients as previously described52. The protocol was approved by the Ethics Committee of Seoul National University Hospital Institutional Review Board (1511-094-721) and by the Hospital for Special Surgery Institutional Review Board (2016-957, 2016-958, and 2016-139). The diagnosis of RA was based on the 1987 revised criteria of the American College of Rheumatology53. There was limited information about patients’ medications, and correlation of the following findings with therapy was not possible.
Cell culture. Peripheral blood mononuclear cells (PBMCs) from blood leukocyte preparations purchased from the New York Blood Center or mononuclear cells from SF of RA patients were isolated by density gradient centrifugation with Ficoll (Invitrogen, Carlsbad, CA). CD14+ cells were obtained by isolation using anti-CD14 magnetic beads, as recommended by the manufacturer (Miltenyi Biotec, CA). Human CD14+ cells were cultured in a-MEM medium (Invitrogen) supplemented with 10 % fetal bovine scrum (FBS, Hyclonc; SH30070.03) and 1% L-glutamin with 20 ng/ml of M-CSF for 24 hours to generate osteoclast precursor cells (OCPs) The purity of monocytes was >97%, as verified by flow cytometric analysis 22.
For human osteoclastogenesis assays, cells were added to 96 well plates in triplicate at a seeding density of 5xl04 cells per well. Osteoclast precursors were incubated with 20 ng/ml of M-CSF and 40 ng/ml of human soluble RANKE (Peprotech, Rocky Hill, NJ) up to 5 days in a-MEM supplemented with 10 % FBS and 1% E-glutamine. Cytokines were replenished every 3 days. On each day, cells were fixed and stained for tartrate-resistant acid phosphatase (TRAP) using the Acid Phosphatase Eeukocyte diagnostic kit (Sigma; 387A) as recommended by the manufacturer. Multinucleated (greater than 3 nuclei), TRAP-positive osteoclasts were counted in triplicate wells. All cell-cultures were performed by a modification of previously published method22.
Gene expression analysis. For real-time qPCR, DNA free RNA was obtained using the RNeasy Mini Kit from QIAGEN with DNase treatment, and 0.5 mg of total RNA was reverse transcribed using a First Strand cDNA Synthesis kit (Fermentas, Hanover, MD). Real-time PCR was performed in triplicate using the iCycler iQ thermal cycler and detection system (Applied Biosystems, Carlsbad, CA) following the manufacturer’s protocols. Expression of the tested gene was normalized relative to the levels of TBP. The primer sequences are listed in the following Table.
Immunoblot Whole cell extracts were prepared by lysis in buffer containing lx Lamin sample buffer (Bio-rad) and 2-Mercaptoethanol (Sigma). Proteins were separated on 7.5 % SDS-PAGE gels, transferred to polyvinylidene difluoride membranes (PVDF, Millipore; ISEQ00010), and detected by antibodies as listed in the figure legends. NF ATcl (556602, BD Pharmagen) and a-tubulin (T9026, Sigma Aldrich).
TRAP staining Cells were fixed and stained for TRAP using the Acid Phosphatase Leukocyte diagnostic kit (Sigma) as recommended by the manufacturer. Nuclei was stained with methyl green. Multinucleated (more than three nuclei), TRAP-positive osteoclasts were counted in triplicate wells.
RNA Interference 0.2 nmol of antisense LNAs (LNA™ GapmeR) or siRNAs (BATF1 and BATF3) were designed and synthesized by Qiagen (Exiqon) and Ambion, respectively. Knock down of human Se-RNA; NF ATcl or control siRNA were transfected into primary human CD14+ monocytes with the Amaxa Nucleofector device set to program Y-001 using the Human Monocyte Nucleofector kit (Amaxa), as previously described54. Custom antisense-LNA GapmeR for SE-RNA:NFATcl_l:
5 ’ TGTTT AC ACGCT A AC AGGATG AC AGC AG AC ACTGTGTG AA ATC AGTC AGT3 ’ (SEQ ID NO: 1; Figure 7); SE-RNA:NFATcl_2: 5’ATTCAGCTCCACTTA3‘ (SEQ ID NO: 2; FIG 8); Pre-designed siRNA for BATF1 (Ambion, siRNA ID# 3851): 5’GGGAACGGUUAUUUUUCUAtt3’ (SEQ ID NO: 13): Pre-designed siRNA BATF3 (Ambion, siRNA ID# 116229): 5’GCUAGUAGGUUCUGCUGUUtt3’ (SEQ ID NO: 14) (Figure 3).
Fluorescence in situ hybridization Fluorescence-conjugated Se- RNA:NFATcl probes for RNA-FISH were generated according to protocols of Biosearch Technologies. Hybridization was carried according to the protocol of Biosearch Technologies. The stained cells were imaged using a Zeiss Axioplan microscope (Zeiss) with an attached Leica DC 200 digital camera (Leica).
ChlP-sequencing Cells were crosslinked for 5 min at room temperature by the addition of one tenth of the volume of 11% formaldehyde solution (11% formaldehyde, 50 mM HEPES, pH 7.5, 100 mM NaCl, 1 mM EDTA, pH 8.0, and 0.5 mM EGTA, pH 8.0) to the growth medium, followed by 5 min of quenching with 100 mM glycine. Cells were pelleted at 4 °C and washed with ice-cold PBS. The crosslinked cells were lysed with lysis
buffer (50 mM HEPES-KOH, pH 7.5, 140 mM NaCl, 1 mM EDTA, 10% glycerol, 0.5% NP-40 and 0.25% Triton X-100) with protease inhibitors on ice for 10 min and were washed with washing buffer (10 mM Tris-HCl, pH 8.0, 200 mM NaCl, 1 mM EDTA and 0.5 mM EGTA) for 10 min. The lysis samples were resuspended and sonicated in sonication buffer (10 mM Tris-HCl, pH 8.0, 100 mM NaCl, 1 mM EDTA, 0.5 mM EGTA, 0.1% sodium deoxy cholate and 0.5% N-lauroylsarcosinc) using a Bioruptor (Diagcnodc) with 30 s on and 30 s off on a high-power output for 12 cycles. After sonication, samples were centrifuged at 12,000 r.p.m. for 10 min at 4 °C, and 1% of sonicated cell extracts was saved as input. Precleared extracts were then incubated with 5 mg of H3K27ac antibody (ab4729) and RNA polymerase II (pol II, MMS-126R) from abeam overnight at 4 °C. After overnight incubation, antibody-bound agarose beads were washed twice with sonication buffer, once with sonication buffer with 500 mM NaCl, once with LiCl wash buffer (10 mM Tris- HC1, pH 8.0, 1 mM EDTA, 250 mM LiCl and 1% NP-40) and once with TE with 50 mM NaCl. After washing, DNA was eluted in freshly prepared elution buffer (1% SDS and 0.1 M NaHCO3). Cross-links were reversed by overnight incubation at 65 °C. RNA and protein were digested using RNase A and proteinase K, respectively, and DNA was purified with ChIP DNA Clean & Concentrator (Zymo Research). 10 ng of purified immunoprecipitated DNA per sample was ligated with adaptors, and 100-300 bp DNA fragments were purified to prepare DNA libraries using an Illumina TruSeq ChIP Library Prep Kit following the manufacturer’s instructions. ChIP libraries were sequenced (50 bp single end reads) using an Illumina HiSeq 2500 Sequencer at the Weill Cornell Medicine Epigenomic Core Facility per the manufacturer’s recommended protocol.
ATAC-sequencing ATAC-seq was performed as previously described. To prepare nuclei, we spun 50,000 cells at 500g for 5 min, which was followed by a wash using 50 mL of cold lx PBS and centrifugation at 500g for 5 min. Cells were lysed using cold lysis buffer (10 mM Tris-HCl, pH 7.4, 10 mM NaCl, 3 mM MgC12 and 0.1% IGEPAL CA-630). Immediately after lysis, nuclei were spun at 500g for 10 min in a refrigerated centrifuge. Immediately following the nuclei prep, the pellet was resuspended in the transposase reaction mix (Illumina). The transposition reaction was carried out for 30 min at 37 °C. Directly following transposition, the sample was purified using a Qiagen MinElute kit. Then, we amplified library fragments using lx NEB next PCR master mix and custom Nextera PCR primers as previously described, using the following PCR conditions: 72 °C for 5 min; 98 °C for 30 s; and thermocycling at 98 °C for 10s, 63 °C for 30s and 72 °C for Imin.The
libraries were purified using a Qiagen PCR cleanup kit yielding a final library concentration of ~30 nM in 20 ml. Libraries were amplified for a total of 10-13 cycles and were subjected to high-throughput sequencing using the Illumina HiSeq 2500 Sequencer (single end).
ChlP-seq and ATAC-seq analysis. Quality of reads was assessed with FastQC (vO.11.9). Low-quality reads were filtered with skewer (v.0.2.2). Reads were then mapped to reference genome GRCh37 (hgl9) with bowtic2 (v2.3.5.1) with “—very-sensitive" parameters. Ambiguous alignments were filtered out with samtools (vl.10) “view -q 10", then the alignments were sorted according to the coordinates with samtools sort. Definition of super- or typical enhancers was performed with HOMER “findPeaks -style super - typical”. For visualization of average coverages of the peaks, HOMER annotatePeaks.pl was run with “-hist -size given -d” options. For building genomic coverage tracks, the alignment files were processed to bedGraph files with bedtools (v2.27.1) genomecov, then converted to bigWig format with bedSort and bedGraphToBigWig (UCSC tools), then uploaded to UCSC genome browser. HOMER annotatePeaks.pl was used to assign each interval to its nearest gene, whose list is then presented as a Chow-Ruskey plot with R “vennerable" package. ChlP-seq read count at enhancer regions is measured with bedtools multicov, then normalized to RPKM with sequencing depth and interval length. Enhancers' “sensitivity to IBET” was determined with HOMER getDifferentialPeaks with -F 1.5 option.
RNA-sequencing Nuclear RNA was first extracted using RNeasy mini kit (Qiagen). True-seq sample preparation kits (Illumina) were then used to generate libraries with multiplexed barcode adaptors. All samples passed quality control analysis on a Bioanalyzer 2100 (Agilent). Paired-end reads were obtained on an Illumina HiSeq 2500/1500 in the Weill Cornell Medical College Genomics Resources Core Facility. The reads were aligned to hgl9 using STAR aligner (v2.7.3a) with default parameters. We used bedtools multicov to count reads that are mapped within the intervals, then the counts were normalized to FPKM considering both sequencing depth and interval length. The read count matrix is further analyzed with DESeq2 to find differential transcription.
PRO-sequencing Cells were incubated in the nuclear’ run-on reaction condition (5 mM Tris-HCl pH 8.0, 2.5 mM MgCh, 0.5 mM DTT, 150 mM KC1, 0.5% Sarkosyl, 0.4 units / pl of RNase inhibitor) with biotin-NTPs and rNTPs supplied (18.75 pM rATP, 18.75 pM rGTP, 1.875 pM biotin- 11 -CTP, 1.875 pM biotin- 11 -UTP for uPRO; 18.75 pM rATP, 18.75 pM rGTP, 18.75 pM rUTP, 0.75 pM CTP, 7.5 pM biotin-11-CTP for pChRO) for 5 min at 37°C. Run-On RNA was extracted using TRIzol, and fragmented under 0.2 N NaOH for 15
min on ice. Fragmented RNA was neutralized, and buffer exchanged by passing through P- 30 columns (Biorad). 3' RNA adaptor is ligated at 5 pM concentration for 1 hours at room temperature using T4 RNA ligase (NEB), followed by 2 consecutive streptavidin bead bindings and extractions. Extracted RNA is converted to cDNA using template switch reverse transcription with 1 pM RPl-short RT primer, 3.75 pM RTP-Template Switch Oligo, lx Template Switch Enzyme and Buffer (NEB) at 42°C for 30 min. After a SPRI bead cleanup, the cDNA is PCR amplified up to 20 cycles using primers compatible with Illumina Small RNA sequencing. Low quality reads along with Illumina small RNA 3’ adapter were filtered. PRO-seq reads were aligned to hgl9 genome with STAR aligner with default options. 5 ’-end positions of the alignments upon each strand were summarized to bedGraph tracks with bedtools genomecov -5 -strand + (or -), then converted to bigWig formatted files. The bigWig files, along with pre-trained support vector matrix (SVM) that is provided with the program, were run as inputs to the dREG program. Among resulting dREG peaks, ones with peak score greater than 0.7 were selected for further analysis. The remaining dREG peaks were then run in dREG-HD. “Relaxed” dREG-HD result peaks from all samples were merged with cat command, bedSort and bedtools merge -d 100. Read counts of each merged dREG-HD site extended by 250 bases from the center were assessed with bedtools multicov. Differential expression analysis was done with DESeq2, and differentially expressed dREG peak was defined as ones with adjusted p-value smaller than 0.01.
Data Accessions H3K27ac ChlP-seq data for osteoblasts and CD4+ T cells arc available in ENCODE project (www_encodeproject_org), under entry codes ENCFF000CVS and ENCFF000CVU for osteoblasts and ENCFF017MGJ and ENCFF416ZFL for CD4+ T cells. Data for this project have been deposited at NCBI’s Gene Expression Omnibus (GEO) and GSE number is GSE203587. Next-generation sequencing experiments used in this study are listed in FIG. 8a. All data are from two (ChlP-seq and Nuc-seq) or three (ATAC-seq and Pro-seq) replicate experiments.
Statistical Analyses All statistical analyses were performed with GraphPad Prism 8, or R (v4.1.0) software. Detailed information about statistical analysis, including tests and values used is provided in the figure legends. Shapiro-Wilk normality tests were performed; for data that fell within a Gaussian distribution, we performed appropriate parametric statistical tests and for those that did not fall within an equal variance-Gaussian distribution, we performed non-parametric statistical tests. The non-parametric Kruskal-Wallis test was
performed with the Bonferroni correction for comparisons for read density in each region. A p value less than 0.05 is considered statistically significant.
EXAMPLE 2: RANKL regulates SEs in the vicinity of master osteoclast regulators in human osteoclast
RANKL is a key driver to induce human osteoclast differentiation22. Human CD14+ monocytes differentiated into tartrate -resistant acid phosphatase (TRAP)-positive multinuclear osteoclasts with characteristic actin ring formation under M-CSF and RANKL stimulation (FIG. la). Osteoclasts are defined as TRAP-positive multinuclear cells (> 3 nuclei). Phalloidin staining showed actin ring formation of osteoclasts. These studies were designed to identify cell-specific programs in human osteoclasts. Reprogramming of superenhancers (SEs) by micro-environmental stimuli contributes to a distinct cell-specific phenotype in health and disease26. The hypothesis that osteoclast-specific SEs are established early by RANKL stimulation and contribute to osteoclast-specific gene expression was tested. Super-enhancers exhibit higher enrichment of histone 3 lysine 27 acetylation (H3K27ac) and DNase I hypersensitive regions than typical enhancers25. ChlP-scq was performed against H3K27ac after differentiating CD14+ cells with or without RANKL stimulation (FIG. 8A) to catalog SEs. Bioinformatic analysis using the rank ordering of super-enhancer (ROSE) algorithm25 defined 808 SEs in CD 14+ monocytes (CTRL) and 962 SEs in RANKL-treated OCPs (RANKL) (FIG. 8B). Among SEs in osteoclasts, 200 Ses were induced by RANKL relative to control and were closely correlated with gene sets related to bone disease showing pathological bone resorption such as RA and bone inflammation diseases (FTG.1C). 148 Ses that were suppressed by RANKL are associated with disease by infectious agents and regulation of immune system processes (FIG. 1C, FIG. 8B). The 200 upregulated SEs exhibited higher enrichment of H3K27ac compared to typical enhancers (TEs) (FIG. Id). In total SEs and TEs, basal H3K27ac levels were higher in SEs compared to in TEs, and RANKL treatment increased or decreased H3K27ac enrichment in both SEs and TEs (FIG. IE). It has been shown that SEs are strongly associated with the expression of master transcription factors (TFs) that control cell identity and state25,27. Accordingly, RANKL- induced SEs were located in the vicinity of NFATC1, a master regulator of ostcoclastogcncsis16, and key regulators of osteoclast differentiation such as MYC55, PR/SET Domain 1 (PRDM1)56, and matrix metalloproteinase-9 (MMP9)57 (FIGs.lE-F ). In contrast, H3K27ac read density near negative regulators such as interferon regulatory factor 8
(IRF8)58, Kruppel Like factor 2 (KLF2)59, and TNF receptor superfamily member IB (TNFRSF1B)60 was decreased in response to RANKL (FIGs.le-f). These results suggest that RANKL-regulated SEs are strongly associated with both positive and negative TFs to promote osteoclast differentiation.
EXAMPLE 3: RANKL-induced SEs are osteoclast-specific and mark osteoclast identity genes
To further examine whether defined SEs in osteoclast are cell-type specific, the study of RANKL-regulated SEs was extended to other cell types which are related to bone inflammation and RA.H3K27ac ChlP-seq datasets of osteoblasts, CD34+ progenitors and CD4+ T cells were retrieved from the ENCODE project and identified SEs in each cell type. In the defined RANKL-induced 200 SEs, H3K27ac read density was exclusively enriched in human osteoclasts, but not in other cell types (FIG. 2a). A set of SEs from osteoblasts, CD34+ progenitors and CD4+ T cells revealed that H3K27ac was highly enriched in celltype specific SEs (FIG. 2b). Chow-Ruskey diagrams of TE-associated and SE-associated genes in osteoclasts, osteoblasts, CD34+ progenitor cells, and T cells revealed that TEs shared genes that are active in all four cell types (FIG. 2c, the circle in the middle), while SE elements represented the overlap of a few cell types and spanned domains that were almost cell-type specific (FIG. 2c). For example, osteoclast-specific SEs, but not SEs from other cell types, were associated with the gene encoding NFATC1 and TNFRSF11A, which are well- defined osteoclast genes (FIG. 2d). In contrast, osteoblast-specific SEs were associated with the genes encoding runt-related transcription factor 2 (RUNX2) and periostin (POSTN), which are required for osteoblast differentiation (FIG. 2d). Gene ontology analysis using the set of genes associated with SEs in osteoblasts and CD4+ T cells showed that significant biological process terms were strongly descriptive of cell-type specific function (FIG. 9). This result suggests that the genes associated with RANKL-induced SEs are highly cell-type specific relative to TE-associated genes.
EXAMPLE 4: Chromatin accessibility is dynamically regulated in osteoclast SEs
Active enhancers are cis-regulatory elements that modulate the quality and quantity of gene expression24. These regions are characterized primarily by specific combinations of posttranslational modifications of histone proteins (H3K27ac and H3K4mel) and “open” chromatins where major TFs can bind31. Assay for Transposase-Accessible Chromatin with
high-throughput sequencing (ATAC-seq) was performed to investigate chromatin changes during osteoclast differentiation. The ATAC-Seq peaks were binned according to their respective fold changes in response to RANKL (FIG. 3A). Increased chromatin accessibility showed both higher fold change (FIG. 3A, black box in left panel) and at least within twofold change in response to RANKL, but decreased ATAC-seq peaks were uniformly distributed within two-fold change (FIG. 3A, right panel). Highly opened chromatin regions (a black box from FIG. 3A, n=5,899) were correlated with increased read densities of H3K27ac, suggesting that the enhancer regulation during osteoclast differentiation is closely related to local chromatin accessibility. Consistently, the most regulated peaks were located in the potential enhancer regions such as the distal, intragenic, or intergenic regions rather than the promoter. Chromatin accessibility increased in both TE and SE in response to RANKL but was significantly increased in TE with a relatively narrow width (> 500 bp) (FIGs. 3B-C). These suggest that chromatin accessibility is regulated in several specific regions within SE (FIGs. 3B-C), which has a very wide width (> 12.5 kb) defined by H3K27ac (FIG. 1C). Accordingly, relatively narrow ATAC-seq peaks, in contrast to H3K27ac, was upregulated within RANKL-sensitive SEs near genes of NFATC1, PRDM1, and MYC (FIG. 3D). To gain insight into the mechanisms involved in SE regulation, TF binding motifs were investigated in open chromatin within RANKL-regulated SEs. Open chromatin within RANKL-induced SEs were significantly enriched for TF motifs bound by BATF, activator protein-1 (AP-1), NFAT:AP-1, NF AT, and NF-kB, which has shown to be an important TF for osteoclast differentiation2 (FIG. 3E). Sequence motifs bound by TFs regulating osteoclast differentiation, such as PU.1 :IRF8, MAF BZIP transcription factor A (MAFA), and IRF8, were enriched in down-regulated ATAC-seq peaks within SEs (FIG. 3F). These results suggest that RANKL-sensitive SEs may be formed as a consequence of the binding of RANKL-dependent signaling TFs to specific open chromatin regions.
EXAMPLE 5: BET protein inhibition differentially regulates SE-eRNA expression BET protein inhibition effectively suppresses osteoclastogenesis and arthritic bone erosion22. Bromodomain-containing protein 4 (BRD4) occupancy at SEs in other cell types is highly sensitive to BET inhibition26, 1-BET151 -mediated suppression of osteoclastogenesis was assessed to determine if it results from disturbing SE formation which drives target gene expression in human osteoclasts. H3K27ac ChlP-seq was performed in the presence of I- BET151 and examined intensity of H3K27ac signals in pre-defined osteoclast-specific SEs.
I-BET treatment significantly decreased H3K27ac signals in RANKL-induced SEs (FIG.4a). Of note, I-BET 151 treatment was found to not cause a decrease of H3K27ac in all RANKL- induced SEs, whereas significant changes in 49 out of 200 SEs contributed to an overall decrease of H3K27ac read densities in RANKL-induced SEs (FIGs.4b-c). For instance, SEs in the vicinity of NFATC1 and peroxisome proliferator-activated receptor gamma coactivator 1-bcta (PPARGC1B) genes were highly sensitive to I-BET151, while SEs near integrin subunit alpha V (ITGAV) were relatively resistant to inhibitors (FIG.4d). Concomitantly, the expression of NFATcl and PPARGC1B were suppressed by I-BET151 treatment while I-BET151 had a minimal effect on ITGAV (FIG.10a). Motif analysis further showed that IBET-sensitive H3K27ac peaks within RANKL-induced SEs were enriched for TF motifs bound by AP-1, activating transcription factor 3 (ATF3), and BATF (FIG.4e), which are similar to the results of motif enrichment derived from chromatin accessibility (FIG.3g). IBET-insensitive H3K27ac peaks were also enriched for PU.l family motifs (FIG.4e). Identified I-BET sensitive SEs were associated with AP-1 family transcription factors that play a key role in osteoclastogenesis61. BATF was consistently found in SE regions (FIGs. 3g and 4e). However, the role of BATF in osteoclasts remains unclear. The chromatin landscape was examined near BATF, Fos and NF-kB motif sites. As with key osteoclastogenic transcription factors, Fos and NF-kB motif sites (FIG. 10g and FIG.10b), BATF motif sites within RANKL-induced SEs are surrounded by robust signals of H3K27ac and ATAC-seq relative to TEs, indicating that the BATF binding site is in active SEs (FIG.4g). BATF belongs to the AP-1 family and consists of three members: BATF, BATF2, and BATF362 M. BATF1 was diminished initially and increased later, while BATF3 was induced by RANKL during osteoclastogenesis (FIG.10c). However, BATF2 was not detected (data not shown). It has been shown that BATF1 and BATF3 compensate each other64. To test the role of BATF in osteoclastogenesis, both BATF1 and BATF3 were knocked-down using small interfering RNAs (siRNAs) in CD14+ cells. KD cells were cultured with M-CSF and RANKL to differentiate into osteoclasts. Intriguingly, decreased BATF1/3 expression suppressed osteoclastogenesis (FIGs.4h-i), suggesting that BATF1/3 is a positive regulator of osteoclast differentiation. Taken together, these results suggest that BET proteins modulate osteoclast differentiation by selectively regulating RANKL-induced SE formation.
EXAMPLE 6: RNA Polymerase II is recruited to osteoclast SE regions to promote enhancer RNA transcription
ChlP-seq and RNA-seq were performed to investigate how Pol II recruitment and eRNA expression are associated with SEs and TEs across the human osteoclast genome. As measured by ChlP-seq reads, Pol II was found to be enriched in overall enhancers in human osteoclasts (FIG.5a). However, it is notable that RANKL-induccd SEs, relative to TEs, were highly occupied by a large portion of Pol II (FIGs.5b-c). The evidence that the high level of Pol II is occupied within RANKL-induced SEs suggests that these large m-clcmcnt domains may promote transcription of eRNA which drive high-level expression of SE-associated genes. Thus, to measure eRNA expression in osteoclasts, nuclear RNA and examined nascent transcripts were isolated by Nuclear RNA (Nuc)-seq, as previously described67. The presence of higher levels of eRNAs transcribed from SEs than from TEs and RANKL treatment further increased expression of eRNA from SEs (FIG.5d) were observed. Expression of eRNA was increased in 83 - 84 % RANKL-induced SEs, while 85 - 88 % down-regulated SEs showed decreased eRNA expression (FIG.5e). Correlated expression of SE and eRNAs included a set of gene encoding major TFs important for osteoclasts such as NFATC1 and MYC (FIG.5f). These studies also found that both recruitment of Pol II and eRNA expression were enriched in SEs regions in proximity to key osteoclast regulators such as NFATC1, PRDM1, and MYC (FIGs.5g-h), suggesting that osteoclast-specific SE activities may be inferred from eRNA expression and signals.
EXAMPLE 7: Global identification and characteristics of SE-eRNA in human osteoclast
To obtain a better resolution map of eRNA at RANKL-sensitive SEs in osteoclasts, a precision nuclear run-on and sequencing (PRO-seq) assay68 were performed which offers single nucleotide resolution of nascent RNA 3’ ends. Similar to Nuc-seq (FIG.5h), nascent transcripts were found to be highly enriched at NFATC1 -associated SE as well as PRDM1- and MYC-associated SE (FIG.6a, FIG. 11). Using discriminative regulatory-element detection from gro-seq (dREG) analysis 69 , a sensitive machine learning method that predicts active transcriptional regulatory elements and eRNA loci, most dREG peaks were found to be distributed in potential enhancer regions (intergenic: 26 %, intragenic 48.5%), while promoter regions included 19.1% dREG peaks (FIGs.6a-b). The 1,308 dREG peaks were found in the SE region and the 1,569 dREG peaks were located in the TE, suggesting that a
significant amount of eRNAs is transcribed in the SE region (FIG.6b). Predicted eRNA loci derived from dREG analysis were highly occupied by several marks of active enhancer including H3K27ac, open chromatin (ATAC-seq), and Pol II (FIGs.6c-d). RANKL stimulation dramatically increased signaling intensities of H3K27ac, Pol II ChlP-seq, and ATAC-seq, indicating that dREG analysis successfully predicted eRNA loci in both SE and TE (FIGs.6c-d). dREG was also found within RANKL-induccd enhancers were significantly enriched for TF motifs bound by AP-1 family such as ATF3, Fos, Fral, and BATF (FIG.6e). In contrast, down-regulated dREG regions were enriched for TF motifs bound by PU.1 family TFs such as ETS1, Elf4, and PU.l (FIG.6f). RANKL significantly increased the expression of eRNAs in both I-BET-sensitive and -insensitive SEs. However, the abundance of RANKL-induced eRNAs in I-BET-sensitive SEs was comparatively higher than in LBET insensitive SEs (FIG.6g). This result suggests that BET proteins may associate with SE- eRNA transcription by interacting with SEs during osteoclastogenesis. Taken together, these data suggest that RANKL-regulated signaling input is associated with SE-eRNA expression.
EXAMPLE 8: SE-eRNA regulates expression of SE-associated TF in human osteoclast
Consistent with H3K27ac ChlP-seq data (FIG. 1c), gene ontology analysis revealed that dREG peaks were strongly associated with osteoclast and bone-related diseases, including rheumatoid arthritis (RA) (FIG. 7a). NFATcl is a master regulator of osteoclasts15. During osteoclastogenesis, NFATcl mRNA expression was induced by RANKL in a timedependent manner (FIG. 7b). Freshly isolated synovial CD14+ cells from RA patients have a higher osteoclastogenic potential and express NFATcl protein without RANKL stimulation70. Consistently, quantitative RT-PCR analysis showed increased expression of NFATcl in RA synovial CD14+ cells relative to disease control CD14+ cells from healthy donors (FIG. 7c). To evaluate the functional role of SE-eRNA in human osteoclasts, NFATC1 -associated SE-eRNA (SE-eRNA:NFATCl) were examined using fluorescent in situ hybridization (FISH) with antisense probes. SE-eRNA:NFATCl was presented only in RANKL-treated cells (FIG. 7d). Subsequently, knockdown of SE-eRNA:NFATCl was observed to be mediated by antisense oligonucleotides (ASO-LNA) suppressed the differentiation of human OCPs into multi-nucleated TRAP-positive cells (FIGs. 7e-f, FIGs. 12a-b, see Example 1 for details of experiment). Moreover, knockdown of SE- eRNA:NFATCl abrogated RANKL-induced expression of NFATcl mRNA (FIG. 7g, FIG.
12c) and protein (FIG. 7h, FIG. 12d) in human OCPs. To investigate whether RANKL- responsive SE-eRNAs would be associated with a high osteoclastogenic potential of synovial CD14+ cells from RA patients, we assessed the expression of SE-eRNAs profile in RA synovial CD14+ cells. Nuc-seq data derived from synovial CD14+ cells from RA patients or disease control revealed that some of RANKL-sensitive SE-eRNAs were highly upregulated in synovial CD14+ cells of RA patients (FIG. 7i). Among them, nascent RNA expression within NFATC1 -associated SE (SE-eRNA:NFATCl) was increased in synovial CD 14+ cells from RA patients (FIG. 7j). Consistently, quantitative RT-PCR analysis showed increased expression of eRNAs in NFATC1 -associated SE in RA synovial CD 14+ cells relative to disease control CD14+ cells from healthy donors (FIG. 7k). The previous study showed that administration of I-BET151 suppressed arthritic bone erosion and NFATcl expression22. 1- BET151 treatment also suppressed the expression of SE-eRNA:NFATCl (FIG.12e), suggesting that I-BET-mediated inhibition of NFATcl expression might be mediated, in pail, by decreased expression of SE-eRNA:NFATCl. SE-eRNA:PRDMl and SE- eRNA:MYC were also found to be significantly increased in RA synovial OCPs (FIG. 12f). Taken together, these data suggest that eRNAs transcribed from NFATC1 -associated SE are indispensable for the transcriptional activation of NFATC1 and osteoclast differentiation, and the presence of RANKL-responsive SE-eRNAs in RA synovial CD 14+ cells may contribute to the high osteoclastogenic potential of RA synovial macrophages.
Discussion
Dysregulation of osteoclast differentiation and activity was linked to pathological bone diseases and arthritic bone destruction2. The major transcription factors and key molecules driving osteoclastogenesis have been extensively studied, and their global or conditional knock-out mice exhibited defects in bone homeostasis and pathological bone destruction2. However, globally targeting these key factors is often accompanied with nonspecific effects, and conditional deletion approaches may not be available for the treatment of human patients. Identifying cell-specific signatures in human osteoclasts is fundamental to developing osteoclast-specific therapeutic interventions for pathological bone resorption of humans. Here, this study defined 348 RANKL-responsive SEs and SE-associated eRNAs in human early osteoclasts as osteoclast-specific epigenetic programs. RANKL-responsive SEs were enriched by BATF-motifs and BET proteins, which are important regulators of osteoclasts. Targeting SE-eRNAs in the proximity of the NFATcl gene suppressed NFATcl
expression and osteoclastogenesis. Moreover, RANKL-responsive SE-eRNAs were found to be present in synovial macrophages from RA patients. This study provides better understanding of the cell-specific programs driving osteoclastogenesis and insights into the control of pathological bone resorption and suggests that manipulating SE-eRNAs offers a novel, osteoclast-specific therapeutic strategy for treating osteoclast-mediated bone diseases.
Epigenetic mechanisms arc increasingly appreciated for their critical roles in transcriptional control in human health and disease. Among them, super enhancers play a crucial role in regulating genes associated with cell-identity and cell-fate decision25 29. this study identified RANKL-responsive SEs in human osteoclasts. Interestingly, the majority of RANKL-responsive SEs were found to be osteoclast-specific, as they are not detected in osteoblasts, CD4+ T cells, or CD34+ progenitor cells. In this analysis, the specificity of SEs in CD34+ progenitors were found to be ambiguous. CD34+ progenitor cells can differentiate into different cell types including monocytes, osteoclasts, T cells, and even osteoprogenitor cells72'74. Thus, it is possible that CD34+ progenitor cells may share common features in the histone code with other cells, which can be retained in subsequently differentiated cells. However, the importance of shared features between progenitors and differentiated cells remains unknown. Human and mouse osteoclasts share key transcription factors that drive osteoclastogenesis including NFATcl2. RANKL transduces the same signaling pathways, including the NF-kB and MAPK pathways, in both human and mouse osteoclasts. Although genes proximal to SEs are important for tissue or cell identity, SEs among different species do not exhibit sequence conservation75, and the overall sequence similarity in non-coding areas between different species is widely variable76,77. This study suggests the possibility that SEs of osteoclast-identity genes may vary among different species. For example, while this study identified that super enhancers in the vicinity of the NFATcl gene were located at the intergenic region of C-terminal of the NFATcl gene in human osteoclasts, Carey et al. showed NFATcl super-enhancer loci in mouse osteoclasts by specifically focusing on two intronic enhancers of NFATcl33. However, it remains un resolved whether SE-associated genes are modulated by the same regulatory regions in human and mouse cells, which is an area for the future research.
SEs are occupied with large numbers of transcription factor binding sites, and master transcription factors and BET proteins contribute to SE formation26. In this analysis, NFATcl, AP-1, BATF and NF-kB binding motifs are top-ranked motifs in RANKL- responsive SEs. Among them, the crucial contribution of NFATcl, AP-1, and NF-kB to
osteoclasts is well known. Carey et al. showed that PU.1 and MITF coordinated the expression of essential transcription factors for mouse bone marrow derived osteoclasts and the PU.1-Eomes-MITF complex is enriched in two intronic enhancer of NFATcl in mouse osteoclasts10,33. In this study, the PU.l motif was not in the top-ranked TF motifs in RANKL- responsive SEs. However, PU.l -binding motifs were found to be pre-existing in both induced and repressed RANKL-rcsponsivc SEs in control conditions (data not shown), which is consistent with the previous report showing that PU.l can reshape the chromatin landscape by occupying all open regulatory regions in multipotent myeloid precursor cells, including closed sites78'80. However, PU.l is able to activate or repress transcription by cooperative action with other TFs. Izawa et al. showed that osteoclast-specific PU.l binding sites overlapped with NFATcl binding sites81. In addition to the existing regulators of osteoclastogenesis, the BATF motif was found to be highly enriched in RANKL-responsive SEs, suggesting that BATF may be required for SE formation in human osteoclasts. However, the role of BATF proteins in osteoclasts has not been characterized yet. BATF deficiency was found attenuated osteoclast differentiation in human cells and identified BATF as a new regulator of osteoclastogenesis. Defining the mechanistic link between BATF and SE formation requires more investigation, but consistent with the observations provided herein, the BATF3 module has been found in SEs of Anaplastic large cell lymphoma82. The role of key TFs in osteoclasts is extensively studied with a specific focus on their function for activating the promoter. These data suggest that TFs might regulate osteoclastogenesis by controlling SE formation in addition to their well-defined functions. It has been shown that BET proteins are also highly enriched in SEs25. Indeed, targeting BRD4, a BET protein, with small molecule inhibitors, including JQ1 and I-BET, suppressed osteoclast differentiation and joint inflammation and bone erosion in murine models of inflammatory arthritis22,83. This study also showed that RANKL-responsive SEs are preferentially lost in the presence of a BET inhibitor. Given previous findings on the positive role of BET proteins in NFATcl expression and osteoclasts, these data suggest that BET proteins might regulate gene expression and osteoclastogenesis, in part, by controlling SE formation.
Non-coding RNAs contribute significantly to cellular functions and differentiation84,85. The expression and role of non-coding RNAs such as micro-RNAs, long non-coding (line) RNAs, and intronic eRNAs in osteoclasts and in osteoporosis have been documented86'88. Although several non -coding RNA species have been identified in mouse
and human osteoclasts, SE-eRNAs and their function in osteoclastogenesis and bone resorption are largely unexplored in human cells. eRNAs were found to be transcribed from SEs in human osteoclasts. Sakaguchi et al. identified 87 putative eRNAs from mouse osteoclasts using cap-analysis of gene expression (CAGE) and deleting the region containing intronic eRNAs in Nfatcl genes suppressed Nfatcl gene expression88. Thus, this suggests that the location of eRNAs for the same gene differs between mouse osteoclasts and human osteoclasts, supporting the importance of studies with human cells. Advances in gene editing technology such as CRISPR-Cas9 have enabled direct manipulation of SEs in human cells90,91. However, directly targeting super-enhancer regions in humans may disrupt the chromatin landscape and thus remains questionable as an approach. eRNAs are nascent RNA transcripts which can be easily targetable. Knock-down of SE-eRNA have been shown in the vicinity of NFATcl gene suppressed NFATC1 mRNA expression and osteoclastogenesis. Thus, the identification of osteoclast-specific SE-eRNAs provides a target for safe and effective therapeutics for bone diseases. Further comprehensive functional analysis of other SE-eRNAs and characterization of the mechanism of action of RANKL-responsive SEs would be required. Taken together, this study provides the first evidence of the link between SE-eRNAs and human osteoclast differentiation.
Rheumatoid arthritis (RA) is a chronic autoimmune disease that primarily affects the joints, along with various body systems92. Bone erosion is one of the key clinical features of RA 6, leading to joint destruction and impaired mobility93'96. Osteoclasts are the major cell type responsible for bone erosion of RA and are found at sites of structural bone damage in inflammatory arthritis97. Increased number and activity of osteoclasts play crucial roles in rapid bone erosion in RA98,99. Recent advances in genome wide sequencing and highdimensional molecular profiling provide insights into RA pathogenesis and the contribution of cell types and pathways to its pathogenesis. In addition, tissues from RA patients have been thoroughly analyzed by various techniques including genome-wide sequencing, histological analysis, and flow cytometry, which provide patient-specific pathogenesis of RA. Notably, RA synovial CD 14+ cells also have high osteoclastogenic potential and high levels of NFATcl expression70. This shows that nascent eRNAs in RA synovial CD14+ cells were overlapped with RANKL-responsive SE-eRNAs, suggesting that high-levels of osteoclast SE-eRNAs may be associated with a higher osteoclastogenic potential of RA synovial macrophages. In summary, this study is the first study in human osteoclasts to
define SEs and SE-eRNAs and will provide a better understanding of human osteoclast biology and open new therapeutic avenues for human pathological bone destruction.
References
1 Tsukasaki, M. & Takayanagi, H. Osteoimmunology: evolving concepts in bone- immune interactions in health and disease. Nat Rev Immunol 19, 626-642, doi:10.1038/s41577-019-0178-8 (2019).
2 Park-Min, K. H. Mechanisms involved in normal and pathological osteoclastogenesis. Cellular and molecular life sciences : CMLS 75, 2519-2528, doi:10.1007/s00018-018-2817-9 (2018).
3 Novack, D. V. & Teitelbaum, S. L. The osteoclast: friend or foe? Annual review of pathology 3, 457-484, doi:10.1146/annurev.pathmechdis.3.121806.151431 (2008).
4 Goldring, S. R. Pathogenesis of bone and cartilage destruction in rheumatoid arthritis. Rheumatology (Oxford) 42 Suppl 2, iil l-16, doi:10.1093/rheumatology/keg327 (2003).
5 Sato, K. & Takayanagi, H. Osteoclasts, rheumatoid arthritis, and osteoimmunology. Curr Opin Rheumatol 18, 419-426, doi:10.1097/01.bor.0000231912.24740.a5 (2006).
6 Schett, G. & Gravallese, E. Bone erosion in rheumatoid arthritis: mechanisms, diagnosis and treatment. Nat Rev Rheumatol 8, 656-664, doi:10.1038/nrrheum.2012.153 (2012).
7 Yasui, T. et al. Epigenetic regulation of osteoclast differentiation: possible involvement of Jmjd3 in the histone demethylation of Nfatc 1. J Bone Miner Res 26, 2665- 2671 (2011).
8 Asagiri, M. et al. Autoamplification of NFATcl expression determines its essential role in bone homeostasis. J Exp Med 202, 1261-1269 (2005).
9 Yasui, T., Hirose, J., Aburatani, H. & Tanaka, S. Epigenetic regulation of osteoclast differentiation. Ann N Y Acad Sci 1240, 7-13 (2011).
10 Carey, H. A. et al. Eomes partners with PU.l and MITF to Regulate Transcription Factors Critical for osteoclast differentiation. iScience 11, 238-245, doi:10.1016/j.isci.2018.12.018 (2019).
11 Park-Min, K. H. Epigenetic regulation of bone cells. Connect Tissue Res 58, 76-89, doi:10.1080/03008207.2016.1177037 (2017).
12 Li, J. et al. RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proc Natl Acad Sci U SA 97, 1566-1571 (2000).
13 Kong, Y. Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397, 315-323, doi: 10.1038/16852 (1999).
14 Dougall, W. C. et al. RANK is essential for osteoclast and lymph node development. Genes Dev 13, 2412-2424, doi:10.1101/gad,13.18.2412 (1999).
15 Takayanagi, H., Kim S. Koga T, Nishina H, Isshiki M, Yoshida H, Saiura A, Isobe M, Yokochi T, Inoue J, Wagner EF, Mak TW, Kodama T, Tankguchi T Induction and activation of the transcription factor NFATcl (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Developmental cell 3, 889-901 (2002).
16 Takayanagi, H. et al. Induction and activation of the transcription factor NFATcl (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell 3, 889-901, doi:10.1016/sl534-5807(02)00369-6 (2002).
17 Winslow, M. M. et al. Calcineurin/NFAT signaling in osteoblasts regulates bone mass. Developmental cell 10, 771-782, doi:10.1016/j.devcel.2006.04.006 (2006).
18 Aliprantis, A. O. et al. NFATcl in mice represses osteoprotegerin during osteoclastogenesis and dissociates systemic osteopenia from inflammation in cherubism. J Clin Invest 118, 3775-3789, doi:10.1172/JCI35711 (2008).
19 Arrowsmith, C. H., Bountra, C., Fish, P. V., Lee, K. & Schapira, M. Epigenetic protein families: a new frontier for drug discovery. Nat Rev Drug Discov 11, 384-400, doi:10.1038/nrd3674 (2012).
20 Berdasco, M. & Esteller, M. Clinical epigenetics: seizing opportunities for translation. Nat Rev Genet 20, 109-127, doi:10.1038/s41576-018-0074-2 (2019).
21 Nishikawa, K. et al. DNA methyltransferase 3a regulates osteoclast differentiation by coupling to an S-adenosylmethionine-producing metabolic pathway. Nat Med 21, 281- 287, doi:10.1038/nm.3774 (2015).
22 Park-Min, K. H. et al. Inhibition of osteoclastogenesis and inflammatory bone resorption by targeting BET proteins and epigenetic regulation. Nature communications 5, 5418, doi:10.1038/ncomms6418 (2014).
23 Meng, S. et al. BET Inhibitor JQ1 Blocks Inflammation and Bone Destruction. J Dent Res 93, 657-662, doi: 10.1177/0022034514534261 (2014).
24 Williamson, I., Hill, R. E. & Bickmore, W. A. Enhancers: from developmental genetics to the genetics of common human disease. Dev Cell 21, 17-19, doi:10.1016/j.devcel.2011.06.008 (2011).
25 Whyte, W. A. et al. Master transcription factors and mediator establish superenhancers at key cell identity genes. Cell 153, 307-319, doi: 10.1016/j. cell.2013.03.035 (2013).
26 Loven, J. et al. Selective inhibition of tumor oncogenes by disruption of superenhancers. Cell 153, 320-334, doi:10.1016/j.cell.2013.03.036 (2013).
27 Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934-947, doi:10.1016/j.cell.2013.09.053 (2013).
28 Stower, H. Gene expression: Super enhancers. Nat Rev Genet 14, 367, doi:10.1038/nrg3496 (2013).
29 Shin, H. Y. Targeting Super-Enhancers for Disease Treatment and Diagnosis. Mol Cells 41, 506-514, doi: 10.14348/molcells.2018.2297 (2018).
30 Arnold, P. R., Wells, A. D. & Li, X. C. Diversity and Emerging Roles of Enhancer RNA in Regulation of Gene Expression and Cell Fate. Front Cell Dev Biol 7, 377, doi: 10.3389/fcell.2019.00377 (2019).
31 Zentner, G. E. & Henikoff, S. Regulation of nucleosome dynamics by histone modifications. Nat Struct Mol Biol 20, 259-266, doi:10.1038/nsmb.2470 (2013).
32 Adam, R. C. et al. Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice. Nature 521, 366-370, doi:10.1038/naturel4289 (2015).
33 Carey, H. A. et al. Enhancer variants reveal a conserved transcription factor network governed by PU.l during osteoclast differentiation. Bone Res 6, 8, doi:10.1038/s41413-018- 0011-1 (2018).
34 Ko, J. Y., Oh, S. & Yoo, K. H. Functional Enhancers As Master Regulators of Tissue-Specific Gene Regulation and Cancer Development. Mol Cells 40, 169-177, doi: 10.14348/molcells.2017.0033 (2017).
35 Pefanis, E. et al. RNA exosome -regulated long non-coding RNA transcription controls super-enhancer activity. Cell 161, 774-789, doi: 10.1016/j. cell.2015.04.034 (2015).
36 Chang, H. C., Huang, H. C., Juan, H. F. & Hsu, C. L. Investigating the role of superenhancer RNAs underlying embryonic stem cell differentiation. BMC Genomics 20, 896, doi:10.1186/sl2864-019-6293-x (2019).
37 Liang, J. et al. Epstein-Barr virus super-enhancer eRNAs are essential for MYC oncogene expression and lymphoblast proliferation. Proc Natl Acad Sci U SA 113, 14121- 14126, doi:10.1073/pnas.l616697113 (2016).
38 Alvarez-Dominguez, J. R., Knoll, M., Gromatzky, A. A. & Lodish, H. F. The Super- Enhancer-Derived alncRNA-EC7/Bloodlinc Potentiates Red Blood Cell Development in trans. Cell Rep 19, 2503-2514, doi:10.1016/j.cclrcp.2017.05.082 (2017).
39 Kim, T. K. el al. Widespread transcription at neuronal activity-regulated enhancers. Nature 465, 182-187, doi:10.1038/nature09033 (2010).
40 Kim, Y. W ., Lee, S., Yun, J. & Kim, A. Chromatin looping and eRNA transcription precede the transcriptional activation of gene in the beta-globin locus. Biosci Rep 35, doi:10.1042/BSR20140126 (2015).
41 Benner, C., Isoda, T. & Murre, C. New roles for DNA cytosine modification, eRNA, anchors, and superanchors in developing B cell progenitors. Proc Natl Acad Sci U SA 112, 12776-12781, doi:10.1073/pnas.l512995112 (2015).
42 Kaikkonen, M. U. et al. Remodeling of the enhancer landscape during macrophage activation is coupled to enhancer transcription. Mol Cell 51, 310-325, doi:10.1016/j.molcel.2013.07.010 (2013).
43 Meng, F. L. et al. Convergent transcription at intragenic super-enhancers targets AID-initiated genomic instability. Cell 159, 1538-1548, doi: 10.1016/j. cell.2014.11.014 (2014).
44 Lai, F. et al. Activating RNAs associate with Mediator to enhance chromatin architecture and transcription. Nature 494, 497-501, doi:10.1038/naturell884 (2013).
45 Arner, E. et al. Transcribed enhancers lead waves of coordinated transcription in transitioning mammalian cells. Science 347, 1010-1014, doi:10.1126/science.l259418 (2015).
46 Imamura, K. et al. Diminished nuclear RNA decay upon Salmonella infection upregulates antibacterial noncoding RNAs. EMBO J 37, doi: 10.15252/embj.201797723 (2018).
47 Dorighi, K. M. et al. M113 and M114 Facilitate Enhancer RNA Synthesis and Transcription from Promoters Independently of H3K4 Monomethylation. Mol Cell 66, 568- 576 e564, doi:10.1016/j.molcel.2017.04.018 (2017).
48 Cheng, J. H., Pan, D. Z., Tsai, Z. T. & Tsai, H. K. Genome-wide analysis of enhancer RNA in gene regulation across 12 mouse tissues. Sci Rep 5, 12648, doi:10.1038/srepl2648 (2015).
49 Ren, C. et al. Functional annotation of structural ncRNAs within enhancer RNAs in the human genome: implications for human disease. Sci Rep 7, 15518, doi:10.1038/s41598- 017-15822-7 (2017).
50 Li, W. et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498, 516-520, doi:10.1038/naturel2210 (2013).
51 Andersson, R. et al. An atlas of active enhancers across human cell types and tissues. Nature 507, 455-461, doi:10.1038/naturel2787 (2014).
52 Gordon, R. A., Grigoriev, G., Lee, A., Kalliolias, G. D. & Ivashkiv, L. B. The interferon signature and STAT1 expression in rheumatoid arthritis synovial fluid macrophages are induced by tumor necrosis factor alpha and counter-regulated by the synovial fluid microenvironment. Arthritis and rheumatism 64, 3119-3128, doi: 10.1002/art.34544 (2012).
53 Arnett, F. C. et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis and rheumatism 31, 315-324 (1988).
54 Park-Min, K. H. et al. FcgammaRIILdependent inhibition of interferon-gamma responses mediates suppressive effects of intravenous immune globulin. Immunity 26, 67-78, doi:10.1016/j.immuni.2006.11.010 (2007).
55 Bae, S. et al. MYC-dependent oxidative metabolism regulates osteoclastogenesis via nuclear receptor ERRalpha. J Clin Invest Ml, 2555-2568, doi: 10.1172/JCI89935 (2017).
56 Miyauchi, Y. et al. The Blimpl-Bcl6 axis is critical to regulate osteoclast differentiation and bone homeostasis. J Exp Med 207, 751-762, doi:10.1084/jem.20091957 (2010).
57 Zhu, L. et al. Osteoclast-mediated bone resorption is controlled by a compensatory network of secreted and membrane -tethered metalloproteinases. Sci Transl Med 12, doi: 10.1126/scitranslmed.aaw6143 (2020).
58 Zhao, B. et al. Interferon regulatory factor-8 regulates bone metabolism by suppressing osteoclastogenesis. Nat Med 15, 1066-1071, doi:10.1038/nm.2007 (2009).
59 Kim, L, Kim, J. H„ Kim, K„ Seong, S. & Kim, N. The IRF2BP2-KLF2 axis regulates osteoclast and osteoblast differentiation. BMB Rep 52, 469-474 (2019).
60 Sun, L., Lian, J. X. & Meng, S. MiR-125a-5p promotes osteoclastogenesis by targeting TNFRSF1B. Cell Mol Biol Let 24, 23, doi:10.1186/sll658-019-0146-0 (2019).
61 Wagner, E. F. & Eferl, R. Fos/AP-1 proteins in bone and the immune system. Immunol Rev 208, 126-140, doi: 10.1 ll l/j.0105-2896.2005 ,00332.x (2005).
62 Dorsey, M. J. et al. B-ATF: a novel human bZIP protein that associates with members of the AP-1 transcription factor family. Oncogene 11, 2255-2265 (1995).
63 Echlin, D. R., Tae, H. J., Mitin, N. & Taparowsky, E. J. B-ATF functions as a negative regulator of AP-1 mediated transcription and blocks cellular transformation by Ras and Fos. Oncogene 19, 1752-1763, doi:10.1038/sj.onc.l203491 (2000).
64 Tussiwand, R. et al. Compensatory dendritic cell development mediated by BATF- IRF interactions. Nature 490, 502-507, doi:10.1038/naturel l531 (2012).
65 Xiao, S., Huang, Q., Ren, H. & Yang, M. The mechanism and function of super enhancer RNA. Genesis 59, e23422, doi: 10.1002/dvg.23422 (2021).
66 De Santa, F. et al. A large fraction of extragenic RNA pol II transcription sites overlap enhancers. PLoSBiol 8, M000384, doi: 10.1371/journal.pbio.1000384 (2010).
67 Dhaliwal, N. K. & Mitchell, J. A. Nuclear RNA Isolation and Sequencing. Methods Mol Biol 1402, 63-71, doi:10.1007/978-l-4939-3378-5_7 (2016).
68 Kwak, H., Fuda, N. J., Core, L. J. & Lis, J. T. Precise maps of RNA polymerase reveal how promoters direct initiation and pausing. Science 339, 950-953, doi: 10.1126/science.1229386 (2013).
69 Wang, Z., Chu, T., Choate, L. A. & Danko, C. G. Identification of regulatory elements from nascent transcription using dREG. Genome Res 29, 293-303, doi: 10.1101/gr.238279.118 (2019).
70 Yarilina, A., Xu, K., Chen, J. & Ivashkiv, L. B. TNF activates calcium-nuclear factor of activated T cells (NFAT)cl signaling pathways in human macrophages. Proceedings of the National Academy of Sciences of the United States of America 108, 1573-1578, doi: 10.1073/pnas.1010030108 (2011).
71 Hermann-Kleiter, N. & Baier, G. NF AT pulls the strings during CD4+ T helper cell effector functions. Blood ll5, 2989-2997, doi: 10.1182/blood-2009-10-233585 (2010).
72 Abdallah, B. M. et al. CD34 defines an osteoprogenitor cell population in mouse bone marrow stromal cells. Stem Cell Res 15, 449-458, doi:10.1016/j.scr.2015.09.005 (2015).
73 Tjonnfjord, G. E., Veiby, O. P., Steen, R. & Egeland, T. T lymphocyte differentiation in vitro from adult human prethymic CD34+ bone marrow cells. The Journal of experimental medicine 177, 1531-1539, doi:10.1084/jem.l77.6.1531 (1993).
74 Matayoshi, A. et al. Human blood-mobilized hematopoietic precursors differentiate into osteoclasts in the absence of stromal cells. Proceedings of the National Academy of Sciences of the United States of America 93, 10785-10790, doi:10.1073/pnas.93.20.10785 (1996).
75 Peng, Y., Kang, H., Luo, J. & Zhang, Y. A Comparative Analysis of SuperEnhancers and Broad H3K4me3 Domains in Pig, Human, and Mouse Tissues. Front Genet 12, 701049, doi:10.3389/fgene.2021.701049 (2021).
76 Ansari-Lari, M. A. et al. Comparative sequence analysis of a gene-rich cluster at human chromosome 12pl3 and its syntenic region in mouse chromosome 6. Genome Res 8, 29-40 (1998).
77 Hardison, R. C., Oeltjen, J. & Miller, W. Long human-mouse sequence alignments reveal novel regulatory elements: a reason to sequence the mouse genome. Genome Res 7, 959-966, doi: 10.1101/gr.7.10.959 (1997).
78 Ungerback, J. et al. Pioneering, chromatin remodeling, and epigenetic constraint in early T-cell gene regulation by SPI1 (PU.l). Genome Res 28, 1508-1519, doi: 10.1101/gr.231423.117 (2018).
79 Natoli, G., Ghisletti, S. & Barozzi, I. The genomic landscapes of inflammation. Genes Dev 25, 101-106, doi:10.1101/gad.2018811 (2011).
80 Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38, 576-589, doi:10.1016/j.molcel.2010.05.004 (2010).
81 Izawa, N. et al. Cooperation of PU.l With IRF8 and NFATcl Defines Chromatin Landscapes During RANKL-Induced Osteoclastogenesis. J Bone Miner Res 34, 1143-1154, doi:10.1002/jbmr.3689 (2019).
82 Liang, H. C. et al. Super-enhancer-based identification of a BATF3/IL-2R-module reveals vulnerabilities in anaplastic large cell lymphoma. Nature communications 12, 5577, doi:10.1038/s41467-021-25379-9 (2021).
83 Zhang, Q. G., Qian, J. & Zhu, Y. C. Targeting bromodomain-containing protein 4 (BRD4) benefits rheumatoid arthritis. Immunol Lett 166, 103-108, doi:10.1016/j.imlet.2015.05.016 (2015).
84 Cabili, M. N. et al. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes & development 25, 1915- 1927, doi:10.1101/gad.l7446611 (2011).
85 Derrien, T. et al. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome research 22, 1775-1789, doi: 10.1101/gr.132159.111 (2012).
86 Liu, W. et al. LncRNA-mRNA expression profiles and functional networks in osteoclast differentiation. J Cell Mol Med, doi: 10. I ll 1/jcmm.15560 (2020).
87 Silva, A. M. et al. Long noncoding RNAs: a missing link in osteoporosis. Bone Res 7, 10, doi:10.1038/s41413-019-0048-9 (2019).
88 Sakaguchi, Y. et al. Roles of Enhancer RNAs in RANKL-induced Osteoclast Differentiation Identified by Genome-wide Cap-analysis of Gene Expression using CRISPR/Cas9. Sci Rep 8, 7504, doi:10.1038/s41598-018-25748-3 (2018).
89 Kim, J. H. & Kim, N. Regulation of NFATcl in Osteoclast Differentiation. J Bone Metab 21, 233-241, doi:10.11005/jbm.2014.21.4.233 (2014).
90 Wu, Y. et al. Highly efficient therapeutic gene editing of human hematopoietic stem cells. Nat Med 25. 776-783, doi:10.1038/s41591-019-0401-y (2019).
91 Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442-451, doi:10.1016/j.cell.2013.06.044 (2013).
92 Smolen, J. S. et al. Rheumatoid arthritis. Nat Rev Dis Primers 4, 18001, doi:10.1038/nrdp.2018.1 (2018).
93 van der Heijde, D. M. Joint erosions and patients with early rheumatoid arthritis. British journal of rheumatology 34 Suppl 2, 74-78 (1995).
94 Machold, K. P. et al. Very recent onset rheumatoid arthritis: clinical and serological patient characteristics associated with radiographic progression over the first years of disease. Rheumatology 46, 342-349, doi:10.1093/rheumatology/kel237 (2007).
95 Ejbjerg, B. et al. Magnetic resonance imaging of wrist and finger joints in healthy subjects occasionally shows changes resembling erosions and synovitis as seen in rheumatoid arthritis. Arthritis and rheumatism 50, 1097-1106, doi:10.1002/art.20135 (2004).
96 Alippe, Y. & Mbalaviele, G. Omnipresence of inflammasome activities in inflammatory bone diseases. Semin Immunopathol 41, 607-618, doi:10.1007/s00281-019- 00753-4 (2019).
97 Gravallese, E. M. et al. Identification of cell types responsible for bone resorption in rheumatoid arthritis and juvenile rheumatoid arthritis. The American journal of pathology 152, 943-951 (1998).
98 Pettit, A. R. et al. TRANCE/RANKL knockout mice are protected from bone erosion in a serum transfer model of arthritis. The American journal of pathology 159, 1689-1699, doi:10.1016/S0002-9440(10)63016-7 (2001).
99 Redlich, K. et al. Osteoclasts are essential for TNF-alpha-mediated joint destruction. The Journal of clinical investigation 110, 1419-1427, doi:10.1172/JCI15582 (2002). All patents, patent applications, and publications, and references to GenBank or another publicly available sequences database cited throughout the disclosure, are expressly incorporated herein by reference in its entirety. In addition, US Provisional Patent Application No. 63/382,669 is also incorporated in its entirety. While this invention has been disclosed with reference to specific embodiments, it is apparent that other embodiments and variations of this invention are devised by others skilled in the art without departing from the true spirit and scope of the invention. The appended claims include such embodiments and equivalents.
Claims
1. A composition comprising: a pharmaceutically acceptable delivery vehicle, carrier, and/or diluent, and at least one nucleic acid of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof:
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or
(d) combinations of (a), (b), or (c).
2. The composition according to claim 1, wherein at least one nucleic acid comprises 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO: 1.
3. The composition according to claim 1, wherein at least one nucleic acid comprises 18 to 30 nucleotides in length which comprise at least 15 consecutive nucleotides of SEQ ID NO: 2.
4. The composition of claim 1 or 2, wherein said composition comprises an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2.
5. The composition of claim 1 or 2, wherein said composition comprises an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2.
6. The composition of any one of claims 1 to 5 wherein the nucleic acid is an antisense oligonucleotide having at least one modified intemucleoside linkage, sugar moiety, or nucleobase.
7. The composition of any one of claims 1 to 5, wherein the nucleic acid is a chimeric oligonucleotide having a gap segment positioned between 5' and 3' wing segments.
8. The composition of claim 6, wherein the gap segment of the chimeric oligonucleotide is comprised of 2'-deoxynucleotides and the wing segments are comprised of nucleotides having modified sugar moieties.
9. The composition according to any one of claims 1 to 8, wherein the composition comprises a non-viral vector comprising the nucleic acid which is a lipid nanoparticle, lipidoid, or liposome.
10. A method for treating an osteoclastogenesis-related disease, bone or joint destruction, bone-related impaired mobility, osteoporosis, and/or rheumatoid arthritis, said method comprising delivering a nucleic acid that specifically binds to a human osteoclastspecific NFATcl regulatory region in chromosome 18.
11. The method according to claim 10, wherein the nucleic acid is an anti-sense oligonucleotide, an RNAi, siRNA, or combinations thereof.
12. The method according to claim 10 or 11, wherein the nucleic acid is delivered via a lipid nanoparticle, lipidoid, or liposome.
13. The method according to any one of claims 10 to 12, wherein the nucleic acid comprises at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or
(d) combinations of (a), (b), or (c).
14. The method of claim 13, wherein at least one antisense oligonucleotide comprises 18 to 45, 20 to 40, 25 to 35, or about 30 consecutive nucleotides of SEQ ID NO: 1.
15. The method of claim 13, wherein at least one antisense oligonucleotide comprises 18 to 30 nucleotides in length which comprises SEQ ID NO: 2.
16. The method of claim 13, wherein said composition comprises an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 1 or an antisense oligonucleotide having 100% complementarity to one of SEQ ID NO: 2.
17. The method of claim 13, wherein said composition comprises an antisense oligonucleotide of SEQ ID NO: 1 and/or SEQ ID NO: 2.
18. The method of claim 13, wherein the composition is delivered intravenously, via site-specific administration to a joint, via targeted delivery to the bone, or intrathecally.
19. Use of at least one antisense oligonucleotide (ASO) of 15 to 50 nucleotides in length in preparing a medicament, wherein the ASO comprises at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or
(d) combinations of (a), (b), or (c).
20. A composition suitable for treatment of an osteoclastogenesis-related disease, bone or joint destruction, bonc-rclatcd impaired mobility, osteoporosis, and/or rheumatoid arthritis, said composition comprising at least one antisense oligonucleotide of 15 to 50 nucleotides in length comprising at least 15 consecutive nucleotides of a sequence comprising:
(a) TGTTTACACGCTAACAGGATGACAGCAGACACTGTGTGAAATCAGTCAGT (SEQ ID NO: 1), a sequence at least 99% identical to SEQ ID NO: 1, a sequence having at least 95% complementarity to SEQ ID NO: 1, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(b) ATTCAGCTCCACTTA (SEQ ID NO: 2), a sequence at least 99% identical to SEQ ID NO: 2, a sequence having at least 95% complementarity to SEQ ID NO: 2, or a pharmaceutically acceptable salt thereof, or combinations thereof;
(c) a sequence comprising at least 15 consecutive nucleotides of SEQ ID NO: 1 or 2, or a pharmaceutically acceptable salt thereof, or combinations thereof, or
(d) combinations of (a), (b), or (c).
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202263382669P | 2022-11-07 | 2022-11-07 | |
US63/382,669 | 2022-11-07 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024102688A1 true WO2024102688A1 (en) | 2024-05-16 |
Family
ID=91033400
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2023/078886 WO2024102688A1 (en) | 2022-11-07 | 2023-11-07 | Compositions for treating osteoclastogenesis disorders and/or rheumatoid arthritis |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024102688A1 (en) |
Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2006074061A2 (en) * | 2004-12-31 | 2006-07-13 | Sequenom, Inc. | Methods for identifying risk of low bmd and treatments thereof |
US20080125583A1 (en) * | 2005-02-11 | 2008-05-29 | International Business Machines Corporation | Ribonucleic acid interference molecules |
US20080242599A1 (en) * | 2005-05-03 | 2008-10-02 | Majd Zayzafoon | Compositions and Methods for Increasing Osteoblast Cell Differentiation and Bone Generation |
US20190032136A1 (en) * | 2010-04-19 | 2019-01-31 | Celera Corporation | Genetic polymorphisms associated with statin response and cardiovascular diseases, methods of detection and uses thereof |
-
2023
- 2023-11-07 WO PCT/US2023/078886 patent/WO2024102688A1/en unknown
Patent Citations (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2006074061A2 (en) * | 2004-12-31 | 2006-07-13 | Sequenom, Inc. | Methods for identifying risk of low bmd and treatments thereof |
US20080125583A1 (en) * | 2005-02-11 | 2008-05-29 | International Business Machines Corporation | Ribonucleic acid interference molecules |
US20080242599A1 (en) * | 2005-05-03 | 2008-10-02 | Majd Zayzafoon | Compositions and Methods for Increasing Osteoblast Cell Differentiation and Bone Generation |
US20190032136A1 (en) * | 2010-04-19 | 2019-01-31 | Celera Corporation | Genetic polymorphisms associated with statin response and cardiovascular diseases, methods of detection and uses thereof |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Garikipati et al. | Circular RNA CircFndc3b modulates cardiac repair after myocardial infarction via FUS/VEGF-A axis | |
Lim et al. | Targeting the highly abundant circular RNA circSlc8a1 in cardiomyocytes attenuates pressure overload induced hypertrophy | |
Xie et al. | Exosomes derived from microRNA-101-3p-overexpressing human bone marrow mesenchymal stem cells suppress oral cancer cell proliferation, invasion, and migration | |
Chen et al. | Malat1 regulates myogenic differentiation and muscle regeneration through modulating MyoD transcriptional activity | |
Rajman et al. | A microRNA‐129‐5p/Rbfox crosstalk coordinates homeostatic downscaling of excitatory synapses | |
US10253319B2 (en) | miRNA modulators of thermogenesis | |
Zhuang et al. | miR-195 competes with HuR to modulate stim1 mRNA stability and regulate cell migration | |
Boon et al. | MicroRNA-34a regulates cardiac ageing and function | |
Håkelien et al. | The regulatory landscape of osteogenic differentiation | |
Frey et al. | Association of snRNA genes with coiled bodies is mediated by nascent snRNA transcripts | |
Chen et al. | Long noncoding RNA BCAR4 promotes osteosarcoma progression through activating GLI2-dependent gene transcription | |
JP6414886B2 (en) | Long non-coding RNA for anti-cancer therapy | |
US20200158716A1 (en) | Cell atlas of healthy and diseased barrier tissues | |
US20180187190A1 (en) | New crispr assays | |
Czimmerer et al. | The IL-4/STAT6 signaling axis establishes a conserved microRNA signature in human and mouse macrophages regulating cell survival via miR-342-3p | |
Zhang et al. | MiR‐27 alleviates myocardial cell damage induced by hypoxia/reoxygenation via targeting TGFBR1 and inhibiting NF‐κB pathway | |
Xu et al. | hnRNPL-activated circANKRD42 back-splicing and circANKRD42-mediated crosstalk of mechanical stiffness and biochemical signal in lung fibrosis | |
Li et al. | Systematic transcriptome-wide analysis of mRNA–miRNA interactions reveals the involvement of miR-142-5p and its target (FOXO3) in skeletal muscle growth in chickens | |
Panagopoulos et al. | The involvement of microRNAs in osteoarthritis and recent developments: a narrative review | |
Wang et al. | Negative feedback of SNRK to circ-SNRK regulates cardiac function post-myocardial infarction | |
Li et al. | The functional mechanism of MicroRNA in oral lichen planus | |
Fleming et al. | Differential expression of miR-1, a putative tumor suppressing microRNA, in cancer resistant and cancer susceptible mice | |
Liu et al. | miR-221 modulates skeletal muscle satellite cells proliferation and differentiation | |
Yu et al. | XIST/let-7i/HMGA1 axis maintains myofibroblasts activities in oral submucous fibrosis | |
CN113234815B (en) | Application of lncRNA molecule in GBM |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23889570 Country of ref document: EP Kind code of ref document: A1 |