WO2024105633A1 - Compositions for mitophagy induction and uses thereof - Google Patents
Compositions for mitophagy induction and uses thereof Download PDFInfo
- Publication number
- WO2024105633A1 WO2024105633A1 PCT/IB2023/061649 IB2023061649W WO2024105633A1 WO 2024105633 A1 WO2024105633 A1 WO 2024105633A1 IB 2023061649 W IB2023061649 W IB 2023061649W WO 2024105633 A1 WO2024105633 A1 WO 2024105633A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cell
- mts
- polypeptide
- mitochondrial
- disease
- Prior art date
Links
- 230000021125 mitochondrion degradation Effects 0.000 title claims abstract description 43
- 239000000203 mixture Substances 0.000 title description 7
- 230000006698 induction Effects 0.000 title description 6
- 108090000765 processed proteins & peptides Proteins 0.000 claims abstract description 186
- 229920001184 polypeptide Polymers 0.000 claims abstract description 184
- 102000004196 processed proteins & peptides Human genes 0.000 claims abstract description 184
- 230000002438 mitochondrial effect Effects 0.000 claims abstract description 107
- 108010042407 Endonucleases Proteins 0.000 claims abstract description 90
- 102000039446 nucleic acids Human genes 0.000 claims abstract description 52
- 108020004707 nucleic acids Proteins 0.000 claims abstract description 52
- 150000007523 nucleic acids Chemical class 0.000 claims abstract description 52
- 208000012268 mitochondrial disease Diseases 0.000 claims abstract description 37
- 108020005196 Mitochondrial DNA Proteins 0.000 claims abstract description 28
- 230000005782 double-strand break Effects 0.000 claims abstract description 26
- 230000000368 destabilizing effect Effects 0.000 claims abstract description 25
- 230000008685 targeting Effects 0.000 claims abstract description 18
- 230000007306 turnover Effects 0.000 claims abstract description 16
- 210000004027 cell Anatomy 0.000 claims description 264
- 238000000034 method Methods 0.000 claims description 146
- 108090000623 proteins and genes Proteins 0.000 claims description 77
- 239000003381 stabilizer Substances 0.000 claims description 72
- 102000004169 proteins and genes Human genes 0.000 claims description 66
- IEDVJHCEMCRBQM-UHFFFAOYSA-N trimethoprim Chemical group COC1=C(OC)C(OC)=CC(CC=2C(=NC(N)=NC=2)N)=C1 IEDVJHCEMCRBQM-UHFFFAOYSA-N 0.000 claims description 58
- 230000001687 destabilization Effects 0.000 claims description 52
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 42
- 239000002245 particle Substances 0.000 claims description 37
- 230000001965 increasing effect Effects 0.000 claims description 27
- 201000010099 disease Diseases 0.000 claims description 26
- 230000001177 retroviral effect Effects 0.000 claims description 25
- 230000007812 deficiency Effects 0.000 claims description 24
- 102100029175 cGMP-specific 3',5'-cyclic phosphodiesterase Human genes 0.000 claims description 23
- 230000004770 neurodegeneration Effects 0.000 claims description 23
- 208000015122 neurodegenerative disease Diseases 0.000 claims description 23
- 208000024827 Alzheimer disease Diseases 0.000 claims description 22
- 230000001939 inductive effect Effects 0.000 claims description 20
- -1 GTP3BP Proteins 0.000 claims description 19
- 102000018679 Tacrolimus Binding Proteins Human genes 0.000 claims description 18
- 108010027179 Tacrolimus Binding Proteins Proteins 0.000 claims description 18
- 101100189582 Dictyostelium discoideum pdeD gene Proteins 0.000 claims description 15
- 101150098694 PDE5A gene Proteins 0.000 claims description 15
- 201000002169 Mitochondrial myopathy Diseases 0.000 claims description 14
- 208000026350 Inborn Genetic disease Diseases 0.000 claims description 13
- 206010028980 Neoplasm Diseases 0.000 claims description 13
- 201000011510 cancer Diseases 0.000 claims description 13
- 206010012601 diabetes mellitus Diseases 0.000 claims description 13
- 208000016361 genetic disease Diseases 0.000 claims description 13
- 230000004048 modification Effects 0.000 claims description 13
- 238000012986 modification Methods 0.000 claims description 13
- 201000000639 Leber hereditary optic neuropathy Diseases 0.000 claims description 12
- 230000015556 catabolic process Effects 0.000 claims description 12
- 238000006731 degradation reaction Methods 0.000 claims description 12
- 208000035475 disorder Diseases 0.000 claims description 12
- 108091008146 restriction endonucleases Proteins 0.000 claims description 12
- 206010002026 amyotrophic lateral sclerosis Diseases 0.000 claims description 11
- 108020004999 messenger RNA Proteins 0.000 claims description 11
- 230000004060 metabolic process Effects 0.000 claims description 11
- 108010058682 Mitochondrial Proteins Proteins 0.000 claims description 10
- 102000006404 Mitochondrial Proteins Human genes 0.000 claims description 10
- 208000017442 Retinal disease Diseases 0.000 claims description 10
- 210000004899 c-terminal region Anatomy 0.000 claims description 10
- 208000023692 inborn mitochondrial myopathy Diseases 0.000 claims description 10
- 208000035473 Communicable disease Diseases 0.000 claims description 9
- 102000000634 Cytochrome c oxidase subunit IV Human genes 0.000 claims description 9
- 206010019280 Heart failures Diseases 0.000 claims description 9
- 208000023105 Huntington disease Diseases 0.000 claims description 9
- 206010061598 Immunodeficiency Diseases 0.000 claims description 9
- 208000029462 Immunodeficiency disease Diseases 0.000 claims description 9
- 230000015572 biosynthetic process Effects 0.000 claims description 9
- 230000007813 immunodeficiency Effects 0.000 claims description 9
- 208000015181 infectious disease Diseases 0.000 claims description 9
- 239000002773 nucleotide Substances 0.000 claims description 9
- 125000003729 nucleotide group Chemical group 0.000 claims description 9
- 102000002666 Carnitine O-palmitoyltransferase Human genes 0.000 claims description 8
- 108010018424 Carnitine O-palmitoyltransferase Proteins 0.000 claims description 8
- 201000008990 Charcot-Marie-Tooth disease type 2A1 Diseases 0.000 claims description 8
- 201000000915 Chronic Progressive External Ophthalmoplegia Diseases 0.000 claims description 8
- 208000016621 Hearing disease Diseases 0.000 claims description 8
- 208000032087 Hereditary Leber Optic Atrophy Diseases 0.000 claims description 8
- 206010048804 Kearns-Sayre syndrome Diseases 0.000 claims description 8
- 208000009564 MELAS Syndrome Diseases 0.000 claims description 8
- 208000014844 Mitochondrial neurogastrointestinal encephalomyopathy Diseases 0.000 claims description 8
- 208000002009 Pyruvate Dehydrogenase Complex Deficiency Disease Diseases 0.000 claims description 8
- 208000021886 Pyruvate carboxylase deficiency Diseases 0.000 claims description 8
- 230000005856 abnormality Effects 0.000 claims description 8
- 208000029829 aminoglycoside-induced deafness Diseases 0.000 claims description 8
- 208000003531 maternally-inherited Leigh syndrome Diseases 0.000 claims description 8
- 201000002697 mitochondrial DNA depletion syndrome Diseases 0.000 claims description 8
- 239000008194 pharmaceutical composition Substances 0.000 claims description 8
- 208000015445 pyruvate dehydrogenase deficiency Diseases 0.000 claims description 8
- 208000002320 spinal muscular atrophy Diseases 0.000 claims description 8
- 101710088194 Dehydrogenase Proteins 0.000 claims description 7
- 102000004190 Enzymes Human genes 0.000 claims description 7
- 108090000790 Enzymes Proteins 0.000 claims description 7
- 101000835023 Homo sapiens Transcription factor A, mitochondrial Proteins 0.000 claims description 7
- 206010062940 Neuropathy, ataxia, retinitis pigmentosa syndrome Diseases 0.000 claims description 7
- 238000010459 TALEN Methods 0.000 claims description 7
- QJJXYPPXXYFBGM-LFZNUXCKSA-N Tacrolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1\C=C(/C)[C@@H]1[C@H](C)[C@@H](O)CC(=O)[C@H](CC=C)/C=C(C)/C[C@H](C)C[C@H](OC)[C@H]([C@H](C[C@H]2C)OC)O[C@@]2(O)C(=O)C(=O)N2CCCC[C@H]2C(=O)O1 QJJXYPPXXYFBGM-LFZNUXCKSA-N 0.000 claims description 7
- 102100026155 Transcription factor A, mitochondrial Human genes 0.000 claims description 7
- NMFHJNAPXOMSRX-PUPDPRJKSA-N [(1r)-3-(3,4-dimethoxyphenyl)-1-[3-(2-morpholin-4-ylethoxy)phenyl]propyl] (2s)-1-[(2s)-2-(3,4,5-trimethoxyphenyl)butanoyl]piperidine-2-carboxylate Chemical group C([C@@H](OC(=O)[C@@H]1CCCCN1C(=O)[C@@H](CC)C=1C=C(OC)C(OC)=C(OC)C=1)C=1C=C(OCCN2CCOCC2)C=CC=1)CC1=CC=C(OC)C(OC)=C1 NMFHJNAPXOMSRX-PUPDPRJKSA-N 0.000 claims description 7
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 claims description 7
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 claims description 7
- 229960002930 sirolimus Drugs 0.000 claims description 7
- 102000052866 Amino Acyl-tRNA Synthetases Human genes 0.000 claims description 6
- 108700028939 Amino Acyl-tRNA Synthetases Proteins 0.000 claims description 6
- 108090000365 Cytochrome-c oxidases Proteins 0.000 claims description 6
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 claims description 6
- 102000003869 Frataxin Human genes 0.000 claims description 6
- 108090000217 Frataxin Proteins 0.000 claims description 6
- 102000005298 Iron-Sulfur Proteins Human genes 0.000 claims description 6
- 108010081409 Iron-Sulfur Proteins Proteins 0.000 claims description 6
- 101710159527 Maturation protein A Proteins 0.000 claims description 6
- 101710091157 Maturation protein A2 Proteins 0.000 claims description 6
- 108010006519 Molecular Chaperones Proteins 0.000 claims description 6
- 102000005431 Molecular Chaperones Human genes 0.000 claims description 6
- 208000018737 Parkinson disease Diseases 0.000 claims description 6
- 108091005804 Peptidases Proteins 0.000 claims description 6
- 108010068204 Peptide Elongation Factors Proteins 0.000 claims description 6
- 102000002508 Peptide Elongation Factors Human genes 0.000 claims description 6
- 239000004365 Protease Substances 0.000 claims description 6
- 101710150114 Protein rep Proteins 0.000 claims description 6
- 101710152114 Replication protein Proteins 0.000 claims description 6
- 210000003958 hematopoietic stem cell Anatomy 0.000 claims description 6
- 210000002901 mesenchymal stem cell Anatomy 0.000 claims description 6
- 108091064355 mitochondrial RNA Proteins 0.000 claims description 6
- 150000003904 phospholipids Chemical class 0.000 claims description 6
- 238000012545 processing Methods 0.000 claims description 6
- 208000011580 syndromic disease Diseases 0.000 claims description 6
- 238000003786 synthesis reaction Methods 0.000 claims description 6
- 230000014626 tRNA modification Effects 0.000 claims description 6
- 231100000167 toxic agent Toxicity 0.000 claims description 6
- 230000004102 tricarboxylic acid cycle Effects 0.000 claims description 6
- 239000001226 triphosphate Substances 0.000 claims description 6
- 235000011178 triphosphate Nutrition 0.000 claims description 6
- UNXRWKVEANCORM-UHFFFAOYSA-N triphosphoric acid Chemical compound OP(O)(=O)OP(O)(=O)OP(O)(O)=O UNXRWKVEANCORM-UHFFFAOYSA-N 0.000 claims description 6
- 206010003591 Ataxia Diseases 0.000 claims description 5
- 208000023275 Autoimmune disease Diseases 0.000 claims description 5
- 208000010693 Charcot-Marie-Tooth Disease Diseases 0.000 claims description 5
- 208000024412 Friedreich ataxia Diseases 0.000 claims description 5
- 101000693985 Homo sapiens Twinkle mtDNA helicase Proteins 0.000 claims description 5
- 102100027193 Twinkle mtDNA helicase Human genes 0.000 claims description 5
- ACTIUHUUMQJHFO-UPTCCGCDSA-N coenzyme Q10 Chemical compound COC1=C(OC)C(=O)C(C\C=C(/C)CC\C=C(/C)CC\C=C(/C)CC\C=C(/C)CC\C=C(/C)CC\C=C(/C)CC\C=C(/C)CC\C=C(/C)CC\C=C(/C)CCC=C(C)C)=C(C)C1=O ACTIUHUUMQJHFO-UPTCCGCDSA-N 0.000 claims description 5
- 238000012217 deletion Methods 0.000 claims description 5
- 230000037430 deletion Effects 0.000 claims description 5
- 208000036546 leukodystrophy Diseases 0.000 claims description 5
- 201000001119 neuropathy Diseases 0.000 claims description 5
- 230000007823 neuropathy Effects 0.000 claims description 5
- 208000033808 peripheral neuropathy Diseases 0.000 claims description 5
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 5
- 208000023434 Alpers-Huttenlocher syndrome Diseases 0.000 claims description 4
- 102000006589 Alpha-ketoglutarate dehydrogenase Human genes 0.000 claims description 4
- 108020004306 Alpha-ketoglutarate dehydrogenase Proteins 0.000 claims description 4
- 206010001939 Aminoaciduria Diseases 0.000 claims description 4
- 208000015012 Autosomal recessive progressive external ophthalmoplegia Diseases 0.000 claims description 4
- 108091033409 CRISPR Proteins 0.000 claims description 4
- 102100027943 Carnitine O-palmitoyltransferase 1, liver isoform Human genes 0.000 claims description 4
- 101710120614 Carnitine O-palmitoyltransferase 1, liver isoform Proteins 0.000 claims description 4
- 101710108984 Carnitine O-palmitoyltransferase 1, muscle isoform Proteins 0.000 claims description 4
- 206010008025 Cerebellar ataxia Diseases 0.000 claims description 4
- 206010008027 Cerebellar atrophy Diseases 0.000 claims description 4
- 206010008635 Cholestasis Diseases 0.000 claims description 4
- 108010014080 DNA Polymerase gamma Proteins 0.000 claims description 4
- 102000016903 DNA Polymerase gamma Human genes 0.000 claims description 4
- 206010011878 Deafness Diseases 0.000 claims description 4
- 206010013887 Dysarthria Diseases 0.000 claims description 4
- 208000006517 Fumaric aciduria Diseases 0.000 claims description 4
- 108700036912 Fumaric aciduria Proteins 0.000 claims description 4
- 206010053759 Growth retardation Diseases 0.000 claims description 4
- 206010065973 Iron Overload Diseases 0.000 claims description 4
- 208000006136 Leigh Disease Diseases 0.000 claims description 4
- 208000017507 Leigh syndrome Diseases 0.000 claims description 4
- 108700010229 Leukoencephalopathy with Brainstem and Spinal Cord Involvement and Lactate Elevation Proteins 0.000 claims description 4
- 208000025121 Leukoencephalopathy with brain stem and spinal cord involvement-high lactate syndrome Diseases 0.000 claims description 4
- 102000003960 Ligases Human genes 0.000 claims description 4
- 108090000364 Ligases Proteins 0.000 claims description 4
- 201000009035 MERRF syndrome Diseases 0.000 claims description 4
- 208000014413 Maternally-inherited diabetes and deafness Diseases 0.000 claims description 4
- 206010058799 Mitochondrial encephalomyopathy Diseases 0.000 claims description 4
- 206010069825 Myoclonic epilepsy and ragged-red fibres Diseases 0.000 claims description 4
- 208000013234 Pearson syndrome Diseases 0.000 claims description 4
- 108010017070 Zinc Finger Nucleases Proteins 0.000 claims description 4
- 208000012892 autosomal dominant progressive external ophthalmoplegia Diseases 0.000 claims description 4
- 230000002146 bilateral effect Effects 0.000 claims description 4
- 230000007870 cholestasis Effects 0.000 claims description 4
- 231100000359 cholestasis Toxicity 0.000 claims description 4
- 231100000895 deafness Toxicity 0.000 claims description 4
- 208000016097 disease of metabolism Diseases 0.000 claims description 4
- 230000005584 early death Effects 0.000 claims description 4
- 208000014346 fumarase deficiency Diseases 0.000 claims description 4
- 231100000001 growth retardation Toxicity 0.000 claims description 4
- 208000016354 hearing loss disease Diseases 0.000 claims description 4
- 210000005260 human cell Anatomy 0.000 claims description 4
- 208000030159 metabolic disease Diseases 0.000 claims description 4
- 201000011540 mitochondrial DNA depletion syndrome 4a Diseases 0.000 claims description 4
- 230000017074 necrotic cell death Effects 0.000 claims description 4
- 206010030875 ophthalmoplegia Diseases 0.000 claims description 4
- 208000037118 sensory ataxia Diseases 0.000 claims description 4
- 208000003755 striatonigral degeneration Diseases 0.000 claims description 4
- 108010030844 2-methylcitrate synthase Proteins 0.000 claims description 3
- 102100030877 28S ribosomal protein S16, mitochondrial Human genes 0.000 claims description 3
- 102100029442 28S ribosomal protein S22, mitochondrial Human genes 0.000 claims description 3
- 102100039358 3-hydroxyacyl-CoA dehydrogenase type-2 Human genes 0.000 claims description 3
- 102100034767 3-hydroxyisobutyryl-CoA hydrolase, mitochondrial Human genes 0.000 claims description 3
- 102100039522 39S ribosomal protein L3, mitochondrial Human genes 0.000 claims description 3
- 102100034147 39S ribosomal protein L44, mitochondrial Human genes 0.000 claims description 3
- 102100029005 ATP-dependent RNA helicase SUPV3L1, mitochondrial Human genes 0.000 claims description 3
- 108091006112 ATPases Proteins 0.000 claims description 3
- 108010009924 Aconitate hydratase Proteins 0.000 claims description 3
- 102000057290 Adenosine Triphosphatases Human genes 0.000 claims description 3
- 102100037411 Alanine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 101100000338 Arabidopsis thaliana ABCC11 gene Proteins 0.000 claims description 3
- 101100000344 Arabidopsis thaliana ABCC12 gene Proteins 0.000 claims description 3
- 102100026198 Aspartate-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100028724 BolA-like protein 3 Human genes 0.000 claims description 3
- 102100034476 CCA tRNA nucleotidyltransferase 1, mitochondrial Human genes 0.000 claims description 3
- 108010071536 Citrate (Si)-synthase Proteins 0.000 claims description 3
- 102100037021 Citrate synthase, mitochondrial Human genes 0.000 claims description 3
- 102100039259 Cytochrome c oxidase subunit 8A, mitochondrial Human genes 0.000 claims description 3
- 108050008072 Cytochrome c oxidase subunit IV Proteins 0.000 claims description 3
- 102100039868 Cytoplasmic aconitate hydratase Human genes 0.000 claims description 3
- 102100036853 Deoxyguanosine kinase, mitochondrial Human genes 0.000 claims description 3
- 102100039147 Dimethyladenosine transferase 2, mitochondrial Human genes 0.000 claims description 3
- 101100010303 Drosophila melanogaster PolG1 gene Proteins 0.000 claims description 3
- 101100519466 Drosophila melanogaster mtDNA-helicase gene Proteins 0.000 claims description 3
- 102100021309 Elongation factor Ts, mitochondrial Human genes 0.000 claims description 3
- 102100033238 Elongation factor Tu, mitochondrial Human genes 0.000 claims description 3
- 102100021822 Enoyl-CoA hydratase, mitochondrial Human genes 0.000 claims description 3
- 101710180035 Enoyl-CoA hydratase, mitochondrial Proteins 0.000 claims description 3
- 241000588724 Escherichia coli Species 0.000 claims description 3
- 102100039111 FAD-linked sulfhydryl oxidase ALR Human genes 0.000 claims description 3
- 108010036781 Fumarate Hydratase Proteins 0.000 claims description 3
- 102100036160 Fumarate hydratase, mitochondrial Human genes 0.000 claims description 3
- 102100036589 Glycine-tRNA ligase Human genes 0.000 claims description 3
- 102100029015 Histidine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 101000635698 Homo sapiens 28S ribosomal protein S16, mitochondrial Proteins 0.000 claims description 3
- 101000699890 Homo sapiens 28S ribosomal protein S22, mitochondrial Proteins 0.000 claims description 3
- 101001035740 Homo sapiens 3-hydroxyacyl-CoA dehydrogenase type-2 Proteins 0.000 claims description 3
- 101000872461 Homo sapiens 3-hydroxyisobutyryl-CoA hydrolase, mitochondrial Proteins 0.000 claims description 3
- 101000670350 Homo sapiens 39S ribosomal protein L3, mitochondrial Proteins 0.000 claims description 3
- 101000711597 Homo sapiens 39S ribosomal protein L44, mitochondrial Proteins 0.000 claims description 3
- 101000696694 Homo sapiens ATP-dependent RNA helicase SUPV3L1, mitochondrial Proteins 0.000 claims description 3
- 101000662481 Homo sapiens Alanine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000630206 Homo sapiens Aspartate-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000695321 Homo sapiens BolA-like protein 3 Proteins 0.000 claims description 3
- 101000849001 Homo sapiens CCA tRNA nucleotidyltransferase 1, mitochondrial Proteins 0.000 claims description 3
- 101000745956 Homo sapiens Cytochrome c oxidase subunit 8A, mitochondrial Proteins 0.000 claims description 3
- 101000838688 Homo sapiens D-aminoacyl-tRNA deacylase 1 Proteins 0.000 claims description 3
- 101000928003 Homo sapiens Deoxyguanosine kinase, mitochondrial Proteins 0.000 claims description 3
- 101000889470 Homo sapiens Dimethyladenosine transferase 2, mitochondrial Proteins 0.000 claims description 3
- 101000895350 Homo sapiens Elongation factor Ts, mitochondrial Proteins 0.000 claims description 3
- 101000959079 Homo sapiens FAD-linked sulfhydryl oxidase ALR Proteins 0.000 claims description 3
- 101000696493 Homo sapiens Histidine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101001032502 Homo sapiens Iron-sulfur cluster assembly enzyme ISCU, mitochondrial Proteins 0.000 claims description 3
- 101000875643 Homo sapiens Isoleucine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000966742 Homo sapiens Leucine-rich PPR motif-containing protein, mitochondrial Proteins 0.000 claims description 3
- 101000624540 Homo sapiens Leucine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000787809 Homo sapiens Methionine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000574302 Homo sapiens Mitochondrial genome maintenance exonuclease 1 Proteins 0.000 claims description 3
- 101000972158 Homo sapiens Mitochondrial tRNA-specific 2-thiouridylase 1 Proteins 0.000 claims description 3
- 101001111288 Homo sapiens NFU1 iron-sulfur cluster scaffold homolog, mitochondrial Proteins 0.000 claims description 3
- 101000938567 Homo sapiens Persulfide dioxygenase ETHE1, mitochondrial Proteins 0.000 claims description 3
- 101000842043 Homo sapiens Phenylalanine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000735427 Homo sapiens Poly(A) RNA polymerase, mitochondrial Proteins 0.000 claims description 3
- 101001066878 Homo sapiens Polyribonucleotide nucleotidyltransferase 1, mitochondrial Proteins 0.000 claims description 3
- 101000874919 Homo sapiens Probable arginine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000640325 Homo sapiens Probable asparagine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000630121 Homo sapiens Probable cysteine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000630267 Homo sapiens Probable glutamate-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000695548 Homo sapiens Probable proline-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000623849 Homo sapiens Protein MTO1 homolog, mitochondrial Proteins 0.000 claims description 3
- 101000969776 Homo sapiens Protein Mpv17 Proteins 0.000 claims description 3
- 101000651437 Homo sapiens Protein SERAC1 Proteins 0.000 claims description 3
- 101001078484 Homo sapiens Ribonuclease H1 Proteins 0.000 claims description 3
- 101000674040 Homo sapiens Serine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000661446 Homo sapiens Succinate-CoA ligase [ADP-forming] subunit beta, mitochondrial Proteins 0.000 claims description 3
- 101000832009 Homo sapiens Succinate-CoA ligase [ADP/GDP-forming] subunit alpha, mitochondrial Proteins 0.000 claims description 3
- 101000658120 Homo sapiens Threonine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101001008959 Homo sapiens Thymidine kinase 2, mitochondrial Proteins 0.000 claims description 3
- 101000796134 Homo sapiens Thymidine phosphorylase Proteins 0.000 claims description 3
- 101000834946 Homo sapiens Transcription elongation factor, mitochondrial Proteins 0.000 claims description 3
- 101001116554 Homo sapiens Transcription termination factor 1, mitochondrial Proteins 0.000 claims description 3
- 101000836189 Homo sapiens Translational activator of cytochrome c oxidase 1 Proteins 0.000 claims description 3
- 101000787286 Homo sapiens Valine-tRNA ligase Proteins 0.000 claims description 3
- 101000787276 Homo sapiens Valine-tRNA ligase, mitochondrial Proteins 0.000 claims description 3
- 101000669028 Homo sapiens Zinc phosphodiesterase ELAC protein 2 Proteins 0.000 claims description 3
- 101000766035 Homo sapiens tRNA (guanine(37)-N1)-methyltransferase Proteins 0.000 claims description 3
- 101000624356 Homo sapiens tRNA dimethylallyltransferase Proteins 0.000 claims description 3
- 101710134930 Import motor subunit, mitochondrial Proteins 0.000 claims description 3
- 102100038096 Iron-sulfur cluster assembly enzyme ISCU, mitochondrial Human genes 0.000 claims description 3
- 102000012011 Isocitrate Dehydrogenase Human genes 0.000 claims description 3
- 108010075869 Isocitrate Dehydrogenase Proteins 0.000 claims description 3
- 102100035997 Isoleucine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 108091007984 KARS Proteins 0.000 claims description 3
- 102100040589 Leucine-rich PPR motif-containing protein, mitochondrial Human genes 0.000 claims description 3
- 102100023342 Leucine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100035529 Lysine-tRNA ligase Human genes 0.000 claims description 3
- 101150073395 MTFMT gene Proteins 0.000 claims description 3
- 102000013460 Malate Dehydrogenase Human genes 0.000 claims description 3
- 108010026217 Malate Dehydrogenase Proteins 0.000 claims description 3
- 102100025860 Methionine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100028928 Methionyl-tRNA formyltransferase, mitochondrial Human genes 0.000 claims description 3
- 102100025785 Mitochondrial genome maintenance exonuclease 1 Human genes 0.000 claims description 3
- 102100022450 Mitochondrial tRNA-specific 2-thiouridylase 1 Human genes 0.000 claims description 3
- 102100024011 NFU1 iron-sulfur cluster scaffold homolog, mitochondrial Human genes 0.000 claims description 3
- 101150078890 POLG gene Proteins 0.000 claims description 3
- 102100030940 Persulfide dioxygenase ETHE1, mitochondrial Human genes 0.000 claims description 3
- 102100029354 Phenylalanine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 108010064209 Phosphoribosylglycinamide formyltransferase Proteins 0.000 claims description 3
- 102100034937 Poly(A) RNA polymerase, mitochondrial Human genes 0.000 claims description 3
- 102100034410 Polyribonucleotide nucleotidyltransferase 1, mitochondrial Human genes 0.000 claims description 3
- 102100036134 Probable arginine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100033917 Probable asparagine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100026179 Probable cysteine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100026125 Probable glutamate-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100028531 Probable proline-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100023083 Protein MTO1 homolog, mitochondrial Human genes 0.000 claims description 3
- 102100021273 Protein Mpv17 Human genes 0.000 claims description 3
- 102100027374 Protein SERAC1 Human genes 0.000 claims description 3
- LCTONWCANYUPML-UHFFFAOYSA-M Pyruvate Chemical compound CC(=O)C([O-])=O LCTONWCANYUPML-UHFFFAOYSA-M 0.000 claims description 3
- 108010053763 Pyruvate Carboxylase Proteins 0.000 claims description 3
- 102100039895 Pyruvate carboxylase, mitochondrial Human genes 0.000 claims description 3
- 102100025290 Ribonuclease H1 Human genes 0.000 claims description 3
- 101100501116 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) TUF1 gene Proteins 0.000 claims description 3
- 241000252141 Semionotiformes Species 0.000 claims description 3
- 102100040597 Serine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102000019259 Succinate Dehydrogenase Human genes 0.000 claims description 3
- 108010012901 Succinate Dehydrogenase Proteins 0.000 claims description 3
- 108010075728 Succinate-CoA Ligases Proteins 0.000 claims description 3
- 102000011929 Succinate-CoA Ligases Human genes 0.000 claims description 3
- 102100037811 Succinate-CoA ligase [ADP-forming] subunit beta, mitochondrial Human genes 0.000 claims description 3
- 102100024241 Succinate-CoA ligase [ADP/GDP-forming] subunit alpha, mitochondrial Human genes 0.000 claims description 3
- 101150026786 TUFM gene Proteins 0.000 claims description 3
- 102100034997 Threonine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100027624 Thymidine kinase 2, mitochondrial Human genes 0.000 claims description 3
- 102100031372 Thymidine phosphorylase Human genes 0.000 claims description 3
- 102100026162 Transcription elongation factor, mitochondrial Human genes 0.000 claims description 3
- 102100024950 Transcription termination factor 1, mitochondrial Human genes 0.000 claims description 3
- 102100027210 Translational activator of cytochrome c oxidase 1 Human genes 0.000 claims description 3
- 102100025336 Tyrosine-tRNA ligase, mitochondrial Human genes 0.000 claims description 3
- 102100025607 Valine-tRNA ligase Human genes 0.000 claims description 3
- 108091008852 Waterwitch Proteins 0.000 claims description 3
- 102100039877 Zinc phosphodiesterase ELAC protein 2 Human genes 0.000 claims description 3
- 210000002798 bone marrow cell Anatomy 0.000 claims description 3
- 210000002865 immune cell Anatomy 0.000 claims description 3
- 210000004962 mammalian cell Anatomy 0.000 claims description 3
- 108010034343 phosphoribosylamine-glycine ligase Proteins 0.000 claims description 3
- 108010078587 pseudouridylate synthetase Proteins 0.000 claims description 3
- 210000001082 somatic cell Anatomy 0.000 claims description 3
- 102100026250 tRNA (guanine(37)-N1)-methyltransferase Human genes 0.000 claims description 3
- 102100023397 tRNA dimethylallyltransferase Human genes 0.000 claims description 3
- 102100029783 tRNA pseudouridine synthase A Human genes 0.000 claims description 3
- PONZBUKBFVIXOD-UHFFFAOYSA-N 9,10-dicarbamoylperylene-3,4-dicarboxylic acid Chemical compound C=12C3=CC=C(C(O)=O)C2=C(C(O)=O)C=CC=1C1=CC=C(C(O)=N)C2=C1C3=CC=C2C(=N)O PONZBUKBFVIXOD-UHFFFAOYSA-N 0.000 claims description 2
- 102100037642 Elongation factor G, mitochondrial Human genes 0.000 claims description 2
- 101000880344 Homo sapiens Elongation factor G, mitochondrial Proteins 0.000 claims description 2
- 101001133003 Homo sapiens Mitochondrial translation release factor in rescue Proteins 0.000 claims description 2
- 101000741407 Homo sapiens Putative transferase CAF17, mitochondrial Proteins 0.000 claims description 2
- 102100033858 Mitochondrial translation release factor in rescue Human genes 0.000 claims description 2
- 102100038549 Putative transferase CAF17, mitochondrial Human genes 0.000 claims description 2
- 102100031780 Endonuclease Human genes 0.000 claims 14
- 102100024746 Dihydrofolate reductase Human genes 0.000 claims 4
- 108020001096 dihydrofolate reductase Proteins 0.000 claims 4
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 claims 2
- 241000045500 Diseae Species 0.000 claims 1
- 230000002068 genetic effect Effects 0.000 claims 1
- 102000004533 Endonucleases Human genes 0.000 abstract description 76
- 230000001225 therapeutic effect Effects 0.000 abstract description 2
- 150000001413 amino acids Chemical group 0.000 description 54
- 229960001082 trimethoprim Drugs 0.000 description 51
- 235000018102 proteins Nutrition 0.000 description 49
- 235000001014 amino acid Nutrition 0.000 description 29
- 108010022394 Threonine synthase Proteins 0.000 description 28
- 102000004419 dihydrofolate reductase Human genes 0.000 description 28
- 210000003470 mitochondria Anatomy 0.000 description 28
- 239000013598 vector Substances 0.000 description 27
- 239000002609 medium Substances 0.000 description 26
- 238000012258 culturing Methods 0.000 description 25
- 230000009467 reduction Effects 0.000 description 22
- 238000006467 substitution reaction Methods 0.000 description 20
- 229940024606 amino acid Drugs 0.000 description 17
- 210000002950 fibroblast Anatomy 0.000 description 13
- 230000014509 gene expression Effects 0.000 description 13
- 230000001052 transient effect Effects 0.000 description 13
- 230000000694 effects Effects 0.000 description 11
- 238000010186 staining Methods 0.000 description 11
- BNRNXUUZRGQAQC-UHFFFAOYSA-N sildenafil Chemical compound CCCC1=NN(C)C(C(N2)=O)=C1N=C2C(C(=CC=1)OCC)=CC=1S(=O)(=O)N1CCN(C)CC1 BNRNXUUZRGQAQC-UHFFFAOYSA-N 0.000 description 10
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 9
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 9
- 101000988412 Homo sapiens cGMP-specific 3',5'-cyclic phosphodiesterase Proteins 0.000 description 8
- 230000003612 virological effect Effects 0.000 description 8
- XDHNQDDQEHDUTM-UHFFFAOYSA-N bafliomycin A1 Natural products COC1C=CC=C(C)CC(C)C(O)C(C)C=C(C)C=C(OC)C(=O)OC1C(C)C(O)C(C)C1(O)OC(C(C)C)C(C)C(O)C1 XDHNQDDQEHDUTM-UHFFFAOYSA-N 0.000 description 7
- 230000003247 decreasing effect Effects 0.000 description 7
- 230000008437 mitochondrial biogenesis Effects 0.000 description 7
- 230000004065 mitochondrial dysfunction Effects 0.000 description 7
- 210000001700 mitochondrial membrane Anatomy 0.000 description 7
- 210000001744 T-lymphocyte Anatomy 0.000 description 6
- 210000004961 autolysosome Anatomy 0.000 description 6
- 230000003197 catalytic effect Effects 0.000 description 6
- 238000004520 electroporation Methods 0.000 description 6
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 6
- 238000000799 fluorescence microscopy Methods 0.000 description 6
- 230000010627 oxidative phosphorylation Effects 0.000 description 6
- 239000013612 plasmid Substances 0.000 description 6
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 5
- 108010029485 Protein Isoforms Proteins 0.000 description 5
- 102000001708 Protein Isoforms Human genes 0.000 description 5
- 108700019146 Transgenes Proteins 0.000 description 5
- 230000008436 biogenesis Effects 0.000 description 5
- 230000007423 decrease Effects 0.000 description 5
- 230000034659 glycolysis Effects 0.000 description 5
- 210000003712 lysosome Anatomy 0.000 description 5
- 230000001868 lysosomic effect Effects 0.000 description 5
- 238000004519 manufacturing process Methods 0.000 description 5
- 239000003550 marker Substances 0.000 description 5
- 239000003642 reactive oxygen metabolite Substances 0.000 description 5
- 230000035806 respiratory chain Effects 0.000 description 5
- 229960003310 sildenafil Drugs 0.000 description 5
- ZOOGRGPOEVQQDX-UUOKFMHZSA-N 3',5'-cyclic GMP Chemical compound C([C@H]1O2)OP(O)(=O)O[C@H]1[C@@H](O)[C@@H]2N1C(N=C(NC2=O)N)=C2N=C1 ZOOGRGPOEVQQDX-UUOKFMHZSA-N 0.000 description 4
- RCJYGWGQCPDYSL-HZPDHXFCSA-N 7-[(3-bromo-4-methoxyphenyl)methyl]-1-ethyl-8-[[(1r,2r)-2-hydroxycyclopentyl]amino]-3-(2-hydroxyethyl)purine-2,6-dione Chemical compound C=1C=C(OC)C(Br)=CC=1CN1C=2C(=O)N(CC)C(=O)N(CCO)C=2N=C1N[C@@H]1CCC[C@H]1O RCJYGWGQCPDYSL-HZPDHXFCSA-N 0.000 description 4
- 102220541496 Geminin_N18T_mutation Human genes 0.000 description 4
- 102000035195 Peptidases Human genes 0.000 description 4
- 102220519997 Protein DEK_H12L_mutation Human genes 0.000 description 4
- 102220601576 Putative uncharacterized protein FLJ42102_M42T_mutation Human genes 0.000 description 4
- SECKRCOLJRRGGV-UHFFFAOYSA-N Vardenafil Chemical compound CCCC1=NC(C)=C(C(N=2)=O)N1NC=2C(C(=CC=1)OCC)=CC=1S(=O)(=O)N1CCN(CC)CC1 SECKRCOLJRRGGV-UHFFFAOYSA-N 0.000 description 4
- PFYWPQMAWCYNGW-UHFFFAOYSA-M [6-(dimethylamino)-9-(2-methoxycarbonylphenyl)xanthen-3-ylidene]-dimethylazanium;perchlorate Chemical compound [O-]Cl(=O)(=O)=O.COC(=O)C1=CC=CC=C1C1=C2C=CC(=[N+](C)C)C=C2OC2=CC(N(C)C)=CC=C21 PFYWPQMAWCYNGW-UHFFFAOYSA-M 0.000 description 4
- 230000032683 aging Effects 0.000 description 4
- 230000004900 autophagic degradation Effects 0.000 description 4
- 210000004957 autophagosome Anatomy 0.000 description 4
- WEAJZXNPAWBCOA-INIZCTEOSA-N avanafil Chemical compound C1=C(Cl)C(OC)=CC=C1CNC1=NC(N2[C@@H](CCC2)CO)=NC=C1C(=O)NCC1=NC=CC=N1 WEAJZXNPAWBCOA-INIZCTEOSA-N 0.000 description 4
- 229960000307 avanafil Drugs 0.000 description 4
- 229950004399 beminafil Drugs 0.000 description 4
- ZISFCTXLAXIEMV-UHFFFAOYSA-N benzamidenafil Chemical compound C1=C(OC)C(OC)=CC=C1CNC(=O)C1=CC([N+]([O-])=O)=CC=C1NC(C)CO ZISFCTXLAXIEMV-UHFFFAOYSA-N 0.000 description 4
- KJJPAVCZQWWWMM-WKILWMFISA-N chembl2107736 Chemical compound C1=C(Cl)C(OC)=CC=C1CNC1=NC([C@@H]2CC[C@H](CC2)C(O)=O)=NC2=C1C1=CC=CC=C1S2 KJJPAVCZQWWWMM-WKILWMFISA-N 0.000 description 4
- 229950003418 dasantafil Drugs 0.000 description 4
- 238000002073 fluorescence micrograph Methods 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 238000002347 injection Methods 0.000 description 4
- 239000007924 injection Substances 0.000 description 4
- MVYUCRDXZXLFSB-UHFFFAOYSA-N lodenafil Chemical compound CCCC1=NN(C)C(C(N=2)=O)=C1NC=2C(C(=CC=1)OCC)=CC=1S(=O)(=O)N(CC1)CCN1CCOC(=O)OCCN(CC1)CCN1S(=O)(=O)C(C=1)=CC=C(OCC)C=1C(N1)=NC(=O)C2=C1C(CCC)=NN2C MVYUCRDXZXLFSB-UHFFFAOYSA-N 0.000 description 4
- 239000012528 membrane Substances 0.000 description 4
- 229950002245 mirodenafil Drugs 0.000 description 4
- MIJFNYMSCFYZNY-UHFFFAOYSA-N mirodenafil Chemical compound C1=C(C=2NC=3C(CCC)=CN(CC)C=3C(=O)N=2)C(OCCC)=CC=C1S(=O)(=O)N1CCN(CCO)CC1 MIJFNYMSCFYZNY-UHFFFAOYSA-N 0.000 description 4
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 4
- 238000003908 quality control method Methods 0.000 description 4
- 102200070470 rs137852621 Human genes 0.000 description 4
- 102220054556 rs200529550 Human genes 0.000 description 4
- 102220044906 rs587781669 Human genes 0.000 description 4
- 102220078610 rs797045482 Human genes 0.000 description 4
- 102220105260 rs879254392 Human genes 0.000 description 4
- 229960000835 tadalafil Drugs 0.000 description 4
- IEHKWSGCTWLXFU-IIBYNOLFSA-N tadalafil Chemical compound C1=C2OCOC2=CC([C@@H]2C3=C([C]4C=CC=CC4=N3)C[C@H]3N2C(=O)CN(C3=O)C)=C1 IEHKWSGCTWLXFU-IIBYNOLFSA-N 0.000 description 4
- 238000012546 transfer Methods 0.000 description 4
- IYFNEFQTYQPVOC-UHFFFAOYSA-N udenafil Chemical compound C1=C(C=2NC=3C(CCC)=NN(C)C=3C(=O)N=2)C(OCCC)=CC=C1S(=O)(=O)NCCC1CCCN1C IYFNEFQTYQPVOC-UHFFFAOYSA-N 0.000 description 4
- 229960000438 udenafil Drugs 0.000 description 4
- 229960002381 vardenafil Drugs 0.000 description 4
- 238000001262 western blot Methods 0.000 description 4
- 102220470089 60S ribosomal protein L21_F15S_mutation Human genes 0.000 description 3
- 102000012074 GAF domains Human genes 0.000 description 3
- 108050002598 GAF domains Proteins 0.000 description 3
- 208000010412 Glaucoma Diseases 0.000 description 3
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 3
- 208000025500 Hutchinson-Gilford progeria syndrome Diseases 0.000 description 3
- 102220534940 Integrator complex subunit 13_M66T_mutation Human genes 0.000 description 3
- 208000001344 Macular Edema Diseases 0.000 description 3
- 206010025415 Macular oedema Diseases 0.000 description 3
- 102220486989 Olfactory receptor 2T1_H25R_mutation Human genes 0.000 description 3
- 208000007932 Progeria Diseases 0.000 description 3
- 201000011032 Werner Syndrome Diseases 0.000 description 3
- 206010064930 age-related macular degeneration Diseases 0.000 description 3
- 230000008901 benefit Effects 0.000 description 3
- 230000008859 change Effects 0.000 description 3
- 230000007547 defect Effects 0.000 description 3
- 239000013604 expression vector Substances 0.000 description 3
- 230000004927 fusion Effects 0.000 description 3
- 239000001963 growth medium Substances 0.000 description 3
- 208000002780 macular degeneration Diseases 0.000 description 3
- 201000010230 macular retinal edema Diseases 0.000 description 3
- 239000000463 material Substances 0.000 description 3
- 229930182817 methionine Natural products 0.000 description 3
- 230000036961 partial effect Effects 0.000 description 3
- 230000001575 pathological effect Effects 0.000 description 3
- 239000013641 positive control Substances 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 230000010076 replication Effects 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 102200001904 rs104894321 Human genes 0.000 description 3
- 102200000381 rs121917855 Human genes 0.000 description 3
- 102200118243 rs33937393 Human genes 0.000 description 3
- 102220005320 rs33945546 Human genes 0.000 description 3
- 102220005150 rs33971048 Human genes 0.000 description 3
- 102220049017 rs587784387 Human genes 0.000 description 3
- 102200067619 rs80357382 Human genes 0.000 description 3
- 230000035882 stress Effects 0.000 description 3
- 239000000758 substrate Substances 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 230000035899 viability Effects 0.000 description 3
- 230000002407 ATP formation Effects 0.000 description 2
- 102100021921 ATP synthase subunit a Human genes 0.000 description 2
- 102100027573 ATP synthase subunit alpha, mitochondrial Human genes 0.000 description 2
- 108700028369 Alleles Proteins 0.000 description 2
- UGTJLJZQQFGTJD-UHFFFAOYSA-N Carbonylcyanide-3-chlorophenylhydrazone Chemical compound ClC1=CC=CC(NN=C(C#N)C#N)=C1 UGTJLJZQQFGTJD-UHFFFAOYSA-N 0.000 description 2
- 101150076793 Cox8a gene Proteins 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 102100022207 E3 ubiquitin-protein ligase parkin Human genes 0.000 description 2
- WHUUTDBJXJRKMK-UHFFFAOYSA-N Glutamic acid Natural products OC(=O)C(N)CCC(O)=O WHUUTDBJXJRKMK-UHFFFAOYSA-N 0.000 description 2
- 101000753741 Homo sapiens ATP synthase subunit a Proteins 0.000 description 2
- 101000936262 Homo sapiens ATP synthase subunit alpha, mitochondrial Proteins 0.000 description 2
- 101000936965 Homo sapiens ATP synthase-coupling factor 6, mitochondrial Proteins 0.000 description 2
- 101000619542 Homo sapiens E3 ubiquitin-protein ligase parkin Proteins 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 102220476922 Interleukin-1 receptor-associated kinase 3_K105I_mutation Human genes 0.000 description 2
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical group [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 2
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 2
- 241000713666 Lentivirus Species 0.000 description 2
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 2
- 108091093105 Nuclear DNA Proteins 0.000 description 2
- 102000004245 Proteasome Endopeptidase Complex Human genes 0.000 description 2
- 108090000708 Proteasome Endopeptidase Complex Proteins 0.000 description 2
- 102100039641 Protein MFI Human genes 0.000 description 2
- 238000011529 RT qPCR Methods 0.000 description 2
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 230000021736 acetylation Effects 0.000 description 2
- 238000006640 acetylation reaction Methods 0.000 description 2
- 238000004458 analytical method Methods 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 230000004908 autophagic flux Effects 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- 230000030833 cell death Effects 0.000 description 2
- 230000003833 cell viability Effects 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 230000009920 chelation Effects 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 2
- 208000030533 eye disease Diseases 0.000 description 2
- 230000004992 fission Effects 0.000 description 2
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 2
- 210000005061 intracellular organelle Anatomy 0.000 description 2
- 230000002132 lysosomal effect Effects 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 230000004066 metabolic change Effects 0.000 description 2
- 230000002503 metabolic effect Effects 0.000 description 2
- 125000001360 methionine group Chemical group N[C@@H](CCSC)C(=O)* 0.000 description 2
- 230000002025 microglial effect Effects 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 210000001539 phagocyte Anatomy 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 102000040430 polynucleotide Human genes 0.000 description 2
- 108091033319 polynucleotide Proteins 0.000 description 2
- 239000002157 polynucleotide Substances 0.000 description 2
- 229950010131 puromycin Drugs 0.000 description 2
- 238000003259 recombinant expression Methods 0.000 description 2
- 230000029058 respiratory gaseous exchange Effects 0.000 description 2
- 230000004043 responsiveness Effects 0.000 description 2
- 230000006641 stabilisation Effects 0.000 description 2
- 238000011105 stabilization Methods 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 241001430294 unidentified retrovirus Species 0.000 description 2
- RHQDDDXTJJEYKY-UHFFFAOYSA-N (3-chlorophenyl)azaniumylideneazanide Chemical compound [N]Nc1cccc(Cl)c1 RHQDDDXTJJEYKY-UHFFFAOYSA-N 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- RUVJFMSQTCEAAB-UHFFFAOYSA-M 2-[3-[5,6-dichloro-1,3-bis[[4-(chloromethyl)phenyl]methyl]benzimidazol-2-ylidene]prop-1-enyl]-3-methyl-1,3-benzoxazol-3-ium;chloride Chemical compound [Cl-].O1C2=CC=CC=C2[N+](C)=C1C=CC=C(N(C1=CC(Cl)=C(Cl)C=C11)CC=2C=CC(CCl)=CC=2)N1CC1=CC=C(CCl)C=C1 RUVJFMSQTCEAAB-UHFFFAOYSA-M 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
- 102100036009 5'-AMP-activated protein kinase catalytic subunit alpha-2 Human genes 0.000 description 1
- CLGFIVUFZRGQRP-UHFFFAOYSA-N 7,8-dihydro-8-oxoguanine Chemical compound O=C1NC(N)=NC2=C1NC(=O)N2 CLGFIVUFZRGQRP-UHFFFAOYSA-N 0.000 description 1
- 101150037123 APOE gene Proteins 0.000 description 1
- 229940121819 ATPase inhibitor Drugs 0.000 description 1
- 206010067484 Adverse reaction Diseases 0.000 description 1
- 102100029470 Apolipoprotein E Human genes 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000713826 Avian leukosis virus Species 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- 101710172824 CRISPR-associated endonuclease Cas9 Proteins 0.000 description 1
- 208000024172 Cardiovascular disease Diseases 0.000 description 1
- ACTIUHUUMQJHFO-UHFFFAOYSA-N Coenzym Q10 Natural products COC1=C(OC)C(=O)C(CC=C(C)CCC=C(C)CCC=C(C)CCC=C(C)CCC=C(C)CCC=C(C)CCC=C(C)CCC=C(C)CCC=C(C)CCC=C(C)C)=C(C)C1=O ACTIUHUUMQJHFO-UHFFFAOYSA-N 0.000 description 1
- 108091035707 Consensus sequence Proteins 0.000 description 1
- 102000004654 Cyclic GMP-Dependent Protein Kinases Human genes 0.000 description 1
- 108010003591 Cyclic GMP-Dependent Protein Kinases Proteins 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 241000991587 Enterovirus C Species 0.000 description 1
- 208000010228 Erectile Dysfunction Diseases 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 108060002716 Exonuclease Proteins 0.000 description 1
- 101150066002 GFP gene Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 108020005004 Guide RNA Proteins 0.000 description 1
- 241001559542 Hippocampus hippocampus Species 0.000 description 1
- 108010033040 Histones Proteins 0.000 description 1
- 101000783681 Homo sapiens 5'-AMP-activated protein kinase catalytic subunit alpha-2 Proteins 0.000 description 1
- 101000878605 Homo sapiens Low affinity immunoglobulin epsilon Fc receptor Proteins 0.000 description 1
- 101000969594 Homo sapiens Modulator of apoptosis 1 Proteins 0.000 description 1
- 101000589784 Homo sapiens Pentatricopeptide repeat-containing protein 1, mitochondrial Proteins 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 1
- ROHFNLRQFUQHCH-UHFFFAOYSA-N Leucine Natural products CC(C)CC(N)C(O)=O ROHFNLRQFUQHCH-UHFFFAOYSA-N 0.000 description 1
- 102100038007 Low affinity immunoglobulin epsilon Fc receptor Human genes 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- 241000282567 Macaca fascicularis Species 0.000 description 1
- 102000009664 Microtubule-Associated Proteins Human genes 0.000 description 1
- 108010020004 Microtubule-Associated Proteins Proteins 0.000 description 1
- 102100021440 Modulator of apoptosis 1 Human genes 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000714177 Murine leukemia virus Species 0.000 description 1
- 241000713883 Myeloproliferative sarcoma virus Species 0.000 description 1
- 125000001429 N-terminal alpha-amino-acid group Chemical group 0.000 description 1
- 101710163270 Nuclease Proteins 0.000 description 1
- 108010016731 PPAR gamma Proteins 0.000 description 1
- 102100032227 Pentatricopeptide repeat-containing protein 1, mitochondrial Human genes 0.000 description 1
- 102100038825 Peroxisome proliferator-activated receptor gamma Human genes 0.000 description 1
- 229940123333 Phosphodiesterase 5 inhibitor Drugs 0.000 description 1
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 1
- 208000007014 Retinitis pigmentosa Diseases 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 206010039491 Sarcoma Diseases 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 241000713896 Spleen necrosis virus Species 0.000 description 1
- 101000910035 Streptococcus pyogenes serotype M1 CRISPR-associated endonuclease Cas9/Csn1 Proteins 0.000 description 1
- AYFVYJQAPQTCCC-UHFFFAOYSA-N Threonine Natural products CC(O)C(N)C(O)=O AYFVYJQAPQTCCC-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- KZSNJWFQEVHDMF-UHFFFAOYSA-N Valine Natural products CC(C)C(N)C(O)=O KZSNJWFQEVHDMF-UHFFFAOYSA-N 0.000 description 1
- 230000001133 acceleration Effects 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 239000000362 adenosine triphosphatase inhibitor Substances 0.000 description 1
- 230000006838 adverse reaction Effects 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 230000009435 amidation Effects 0.000 description 1
- 238000007112 amidation reaction Methods 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 239000007864 aqueous solution Substances 0.000 description 1
- 239000007900 aqueous suspension Substances 0.000 description 1
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 230000005033 autophagosome formation Effects 0.000 description 1
- OQFSQFPPLPISGP-UHFFFAOYSA-N beta-carboxyaspartic acid Natural products OC(=O)C(N)C(C(O)=O)C(O)=O OQFSQFPPLPISGP-UHFFFAOYSA-N 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 230000008512 biological response Effects 0.000 description 1
- 239000000090 biomarker Substances 0.000 description 1
- 238000007413 biotinylation Methods 0.000 description 1
- 230000006287 biotinylation Effects 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 230000008045 co-localization Effects 0.000 description 1
- 230000003081 coactivator Effects 0.000 description 1
- 235000017471 coenzyme Q10 Nutrition 0.000 description 1
- 229940110767 coenzyme Q10 Drugs 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 229910052802 copper Chemical group 0.000 description 1
- 239000010949 copper Chemical group 0.000 description 1
- 230000001186 cumulative effect Effects 0.000 description 1
- 235000018417 cysteine Nutrition 0.000 description 1
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 1
- 210000000172 cytosol Anatomy 0.000 description 1
- 238000013461 design Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 230000002500 effect on skin Effects 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 230000007613 environmental effect Effects 0.000 description 1
- 102000013165 exonuclease Human genes 0.000 description 1
- 230000002349 favourable effect Effects 0.000 description 1
- 230000004907 flux Effects 0.000 description 1
- 210000000245 forearm Anatomy 0.000 description 1
- 230000022244 formylation Effects 0.000 description 1
- 238000006170 formylation reaction Methods 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 101150118163 h gene Proteins 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 229910052739 hydrogen Inorganic materials 0.000 description 1
- 239000001257 hydrogen Substances 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- JGPMMRGNQUBGND-UHFFFAOYSA-N idebenone Chemical compound COC1=C(OC)C(=O)C(CCCCCCCCCCO)=C(C)C1=O JGPMMRGNQUBGND-UHFFFAOYSA-N 0.000 description 1
- 229960004135 idebenone Drugs 0.000 description 1
- 201000001881 impotence Diseases 0.000 description 1
- 239000000411 inducer Substances 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 239000000543 intermediate Substances 0.000 description 1
- 229910052742 iron Inorganic materials 0.000 description 1
- AGPKZVBTJJNPAG-UHFFFAOYSA-N isoleucine Natural products CCC(C)C(N)C(O)=O AGPKZVBTJJNPAG-UHFFFAOYSA-N 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 230000028161 membrane depolarization Effects 0.000 description 1
- 230000006680 metabolic alteration Effects 0.000 description 1
- XZWYZXLIPXDOLR-UHFFFAOYSA-N metformin Chemical compound CN(C)C(=N)NC(N)=N XZWYZXLIPXDOLR-UHFFFAOYSA-N 0.000 description 1
- 229960003105 metformin Drugs 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 230000004898 mitochondrial function Effects 0.000 description 1
- 108010045576 mitochondrial transcription factor A Proteins 0.000 description 1
- 238000010852 mitochondrial transfer Methods 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 238000012900 molecular simulation Methods 0.000 description 1
- 210000004897 n-terminal region Anatomy 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 108010007425 oligomycin sensitivity conferring protein Proteins 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 230000036284 oxygen consumption Effects 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 238000002135 phase contrast microscopy Methods 0.000 description 1
- COLNVLDHVKWLRT-UHFFFAOYSA-N phenylalanine Natural products OC(=O)C(N)CC1=CC=CC=C1 COLNVLDHVKWLRT-UHFFFAOYSA-N 0.000 description 1
- 239000002590 phosphodiesterase V inhibitor Substances 0.000 description 1
- 238000005086 pumping Methods 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 230000008263 repair mechanism Effects 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 102200015453 rs121912293 Human genes 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 125000006850 spacer group Chemical group 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 238000002560 therapeutic procedure Methods 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 238000010361 transduction Methods 0.000 description 1
- 230000026683 transduction Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 230000001960 triggered effect Effects 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 230000004906 unfolded protein response Effects 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 239000013603 viral vector Substances 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/16—Hydrolases (3) acting on ester bonds (3.1)
- C12N9/22—Ribonucleases RNAses, DNAses
-
- 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
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/0004—Oxidoreductases (1.)
- C12N9/0012—Oxidoreductases (1.) acting on nitrogen containing compounds as donors (1.4, 1.5, 1.6, 1.7)
- C12N9/0026—Oxidoreductases (1.) acting on nitrogen containing compounds as donors (1.4, 1.5, 1.6, 1.7) acting on CH-NH groups of donors (1.5)
- C12N9/0028—Oxidoreductases (1.) acting on nitrogen containing compounds as donors (1.4, 1.5, 1.6, 1.7) acting on CH-NH groups of donors (1.5) with NAD or NADP as acceptor (1.5.1)
- C12N9/003—Dihydrofolate reductase [DHFR] (1.5.1.3)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y105/00—Oxidoreductases acting on the CH-NH group of donors (1.5)
- C12Y105/01—Oxidoreductases acting on the CH-NH group of donors (1.5) with NAD+ or NADP+ as acceptor (1.5.1)
- C12Y105/01003—Dihydrofolate reductase (1.5.1.3)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/07—Fusion polypeptide containing a localisation/targetting motif containing a mitochondrial localisation signal
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/95—Fusion polypeptide containing a motif/fusion for degradation (ubiquitin fusions, PEST sequence)
Definitions
- Mitochondrial dysfunction is caused by a variety of factors, including mutations in genes encoding mitochondrial proteins, tRNA, and rRNA, accumulation of mutations in the mitochondrial genome, mismanagement of mitochondrial proteins, mismanagement of intracellular organelles such as the endoplasmic reticulum and lysosome, and mitochondrial protein quality control defects.
- Mitochondrial protein quality control defects can be caused by a variety of factors, such as mismanagement of intracellular organelles such as the endoplasmic reticulum and the lysosome.
- quality control by mitochondrial biogenesis and autophagy (mitophagy) and dynamics by fusion/fission is where various mitochondrial dysfunctions converge.
- Mitochondrial quality control defects associated with various diseases continue to be intensively studied as targets for disease therapy.
- Target diseases include not only mitochondrial diseases, but also neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and ALS, heart failure, diabetes, and immunodeficiency for cancer, infectious diseases, and autoimmune diseases.
- Small molecule compounds have been developed to enhance biogenesis, to induce mitophagy, and to suppress excessive fission, etc.
- coenzyme Q10, idebenone, and metformin have been shown to induce mitophagy.
- the disclosure provides polypeptides capable of inducing double strand breaks (DSBs) in mitochondria to enable a temporary and partial reduction in the number of mitochondria in a cell.
- DSBs double strand breaks
- mitochondria Compared to the nucleus, mitochondria havp noor gene repair mechanisms.
- mitochondria strongly transmit signals to the nucleus to promote replication of the mitochondrial genome and increase production of mitochondrial component proteins.
- DSBs can be introduced into mitochondrial DNA through use of a polypeptide comprising a mitochondrial targeting sequence (MTS) fused to an endonuclease.
- MTS mitochondrial targeting sequence
- the present disclosure provides polypeptides having a destabilization domain in addition to a mitochondrial targeting sequence and endonuclease sequence.
- the destabilization domain permits the polypeptide to retain structure and endonuclease activity; in absence of the stabilizing agent, the destabilization domain loses stability leading to degradation of the polypeptide by the proteasome.
- the disclosure provides polypeptides comprising a mitochondrial targeting sequence (MTS), an endonuclease sequence, for example XbaIR, and a destabilizing domain sequence.
- MTS mitochondrial targeting sequence
- an endonuclease sequence for example XbaIR
- a destabilizing domain sequence is useful for directing the polypeptide to the desired site of endonuclease activity, namely the mitochondrial genome.
- a destabilizing domain sequence which is stabilized by a stabilizing agent, enables sensitive ON/OFF control of the endonuclease.
- a cell can be contacted with the polypeptide in the presence of the stabilizing agent for a period of time (during which the polypeptide is active) and, subsequently, the stabilizing agent can be removed, thereby leading to destabilization and degradation of the polypeptide.
- the disclosure provides nucleic acids encoding a polypeptide of the disclosure, particles comprising the nucleic acids, such as viral particles, and host cells comprising a nucleic acid of the disclosure.
- nucleic acids include vectors such as viral (e.g., retroviral) genomes, plasmids, and mRNA molecules.
- exemplary particles include viral particles e.g., retroviral particles). Further exemplary features of nucleic acids, particles and host cells of the disclosure are described in Section 4.3 and specific embodiments 85 to 96, /nfra.
- the disclosure provides methods of (a) inducing mitophagy in a cell, and/or (b) increasing mitochondrial turnover in a cell, and/or (c) increasing mitochondrial mass, and/or (d) inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell by contacting the cell with a polypeptide, nucleic acid, or particle of the disclosure and a stabilizing agent.
- a cell can be transfected with a nucleic acid encoding a polypeptide, transduced with a viral particle containing a nucleic acid encoding the polypeptide, or injected with the polypeptide, and cultured in the presence of the stabilizing agent. Following a period of time (e.g., 6 hours to 5 days), the stabilizing agent can be removed to allow destabilization and degradation of the polypeptide.
- the disclosure provides cells and populations of cells obtained or obtainable by the methods of (a) inducing mitophagy in a cell, and/or (b) increasing mitochondrial turnover in a cell, and/or (c) increasing mitochondrial mass, and/or (d) inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell described herein.
- a “cell” encompasses single cells as well as populations of cells.
- the disclosure provides methods of treating subjects with cells and populations of cells of the disclosure.
- the subject can be a subject having an age- related disease, a mitochondrial disease or disorder, a neurodegenerative disease, an eye disease (e.g., a retinal disease), diabetes, a hearing disorder, a genetic disease, heart failure, an immunodeficiency, cancer, or an infectious disease.
- compositions comprising a polypeptide, nucleic acid, particle or cell (including populations of cells) of the disclosure.
- Such pharmaceutical compositions can be used, for example, in the methods of treatment described herein.
- kits comprising a polypeptide, nucleic acid, or particle of the disclosure and a stabilizing agent. Kits can be used, for example, in the methods of the disclosure.
- compositions and kits are described in Section 4.5 and specific embodiments 196 to 200, infra.
- FIG. 1 shows a retroviral vector map with an MTS-XbalR-DHFR polypeptide (Example 1).
- FIG. 2 shows a molecular model of the MTS-XbalR-DHFR polypeptide of Example 1.
- FIG. 3 shows a retroviral vector map with an EFGR-DHFR polypeptide (Example 1).
- FIG. 4 shows a molecular model of the EGFR-DHFR polypeptide of Example 1 .
- FIG. 5 shows fluorescence microscopy images of Hela cells tranduced with an EGFP- DHFR retroviral vector (Example 1).
- FIG. 6 shows FACS data for Hela cells tranduced with an EGFP-DHFR retroviral vector, showing EGFP expression when cultured in the presence of TMP (Example 1).
- FIG. 7 shows relative EGFP mRNA expression levels in Hela cells tranduced with an EGFP-DHFR retroviral vector cultured with TMP (Example 1).
- FIGS. 8A-8D show fluorescence intensity of Hela cells tranduced with an EGFP-DHFR retroviral vector by fluorescence microscopy and FACS after culturing in media with TMP for two days followed by washout of TMP at 0, 1 , 2, 4, 6, 8, 24, and 48 hours post-washout (Example 1).
- FIG. 8A Time course
- FIG. 8B Fluorescence images
- FIG. 8C FACS analysis
- FIG. 8D Mean fluorescence intensity (MFI) in FACS analysis overtime.
- MFI Mean fluorescence intensity
- FIG. 9A-9D show fluorescence intensity of Hela cells transduced with an EGFP-DHFR retroviral vector by fluorescence microscopy and FACS after culturing in media with TMP for several durations (0, 1 , 2, 4, 6, 8, 24, and 48 hours).
- FIG. 9A Time course
- FIG. 9B Fluorescence images
- FIG. 9C FACS analysis
- FIG. 9D MFI in FACS analysis over time
- FIG. 10A-10C show Xbal expression (FIG. 10A) and mtDNA copy number (CN) (FIG. 10B) for Hela cells transduced with a retroviral vector encoding an MTS-XbalR-DHFR polypeptide (Hela MXD sc20) and cultured overtime in the absence or presence of 0.5 pm TMP for two days followed by TMP washout (FIG. 10C) (Example 1).
- FIG. 11A-11D show MFIs of Mitogreen staining that is an indicator for mitochondrial mass (mtMass) (FIG. 11 A), MFIs of TMRM staining that is an indicator for global mitochondrial membrane potential (mtMP) (FIG. 11 B), and relative TMRM/Mitogreen ratios that is an indicator for mtMP per mass unit (FIG. 11 C) in Hela MXD sc20 incubated with TMP for 3 different durations (16, 20, and 48 hours) following cell culture overtime (FIG. 11 D)
- FIGS. 12A-12C show cell number (FIG. 12A) and cell viability (FIG. 12B) for Hela MXD sc20 cells cultured in the absence or presence of 0.5 pm TMP for two days followed by TMP washout (FIG. 12C). (Example 1).
- FIGS. 13A-13B show mitophagy index (FIG. 13A) by FACS analysis and mtDNA CN estimated by qPCR (FIG. 13B) for Hela MDX sc20 cells transduced with a retroviral vector encoding mtKeimaRed and PARK2 and cultured without or with TMP for several durations.
- CCCP is used for a positive control of mitophagy at 10 pM. (Example 1).
- FIGS. 14A-14D show changes in mitochondrial biogenesis in Hela_GiM cells stably expressing a genetically induced mitophagy (GiM) unit, in the presence and absence of TMP.
- Mitochondrial ROS mtROS
- PGC1a FIG. 14B
- NRF1 FIG. 14C
- TFAM FIGS. 14D
- FIGS. 15A-15F show the expression of several mitochondrial proteins overtime in Hela_GiM cells in the presence and absence of TMP.
- FIG. 15A shows western blot images of mitochondrial proteins on Days 2, 4, 6, and 8.
- FIGS. 15B-15F show the quantified levels of the same proteins. (Example 2).
- FIGS. 16A-16B show phase contrast (PhC) and fluorescence microscopy images of mtKeimaRed-expressing HeLa_GiM cells in the presence and absence of TMP (FIG. 16A) and the percentage of cells that are mitophagy positive (FIG. 16B). (Example 2).
- FIGS. 17A-17B show colocalization of the autophagosomal membrane marker LC3, mitochondrial marker TOM20, and the nuclear stain DAPI in Hela_GiM cells in the presence or absence of TMP and BafA1 (FIG. 17A) and quantified values for the areas that correspond to autophagosomes (FIG. 17B) (Example 3).
- FIGS. 18A-18B show LC3-II expression levels in HeLa_GiM cells in the absence or presence TMP or TMP + BafA1 .
- FIG. 18A shows the western blot images and
- FIG. 18B is a graphical representation of the triplicate quantification. (Example 3).
- FIGS. 19A-19C show results of respirometry, displaying OXPHOS and glycolysis changes overtime (FIG. 19A), two-dimensional expansion of OXPHOS and Glycolysis relation (FIG. 19B), and changes in ATP production, basal respiration, proton leak, and spare capacity overtime (FIG. 19C). (Example 4).
- FIGS. 20A-20G show the assay setup used in Example 5 (FIG. 20A), FACS results in untreated Alzheimer’s Disease (AD) fibroblasts and control NHDF cells (FIG. 20B) and in AD fibroblasts at 7, 14, and 21 days after genetically induced mitophagy (GiM) (FIG. 20C), mtMass and mtMP levels in untreated (FIGS. 20D and 20E, respectively) and in AD fibroblasts at 7 and 14 days after GiM, relative to controls (FIGS. 20F and 20G, respectively), which are described further in Example 5.
- FIGS. 20A-20G show the assay setup used in Example 5 (FIG. 20A), FACS results in untreated Alzheimer’s Disease (AD) fibroblasts and control NHDF cells (FIG. 20B) and in AD fibroblasts at 7, 14, and 21 days after genetically induced mitophagy (GiM) (FIG. 20C), mtMass and mtMP levels in untreated (FI
- the disclosure provides polypeptides capable of inducing double strand breaks (DSBs) in mitochondria to enable a temporary and partial reduction in the number of mitochondria in a cell.
- DSBs double strand breaks
- the mitochondrial genome encodes respiratory chain proteins that are under a strict regulation balanced with the translation of nuclear-coded respiratory proteins. Therefore, the partial loss of the mitochondrial genome directly links to insufficient proton uptake, resulting in depolarization of mitochondrial membrane potential.
- the mitochondrial membrane potential is depolarized in dysfunctional mitochondrial compartments, and a key regulator for mitophagy. Without being bound by theory, it is believed that the more depolarized portions can be preferentially subject to mitophagy following introduction of DSBs.
- mitochondria In response to the stress of DSBs, mitochondria strongly transmit signals to the nucleus (e.g., as part of the mitochondrial unfolded protein response (UPRmt)) to promote replication of the mitochondrial genome and increase production of mitochondrial component proteins.
- URRmt mitochondrial unfolded protein response
- the metabolic alterations resulting from mitochondrial genome reduction can affect the epigenomic status of the cell.
- the reduction can decrease some intermediates of TCA cycle, which are utilized for acetylation and methylation in the nuclear genome and histone.
- introducing DSBs in mitochondrial DNA can be used to efficiently promote mitochondrial turnover, thereby ameliorating mitochondrial dysfunction through the generation of new mitochondria.
- Mitochondrial DNA may possess some modifications, such as 8-oxo-7,8-dihydroguanine (8- OXOG), which is an oxidized form of guanine. With aging, damaging modifications accumulate. Since newly generated mitochondrial genomes lack these modifications, mitochondriogenesis can promote a regain of healthy mitochondrial function.
- 8- OXOG 8-oxo-7,8-dihydroguanine
- DSBs can be introduced into mitochondrial DNA through use a polypeptide comprising a mitochondrial targeting sequence (MTS) fused to an endonuclease.
- MTS mitochondrial targeting sequence
- one potential problem with this approach is excessive endonuclease activity.
- the present disclosure provides polypeptides having a destabilization domain in addition to a mitochondrial targeting sequence and endonuclease sequence. When stabilized by a stabilizing agent, the destabilization domain permits the polypeptide to retain structure and endonuclease activity; in absence of the stabilizing agent, the destabilization domain loses stability leading to degradation of the polypeptide by the proteasome.
- destabilization domain refers to a polypeptide domain which when fused to a second polypeptide domain such as an endonuclease causes the polypeptide to be degraded in the absence of a stabilizing agent which prevents or inhibits the degradation otherwise elicited by the destabilization domain.
- Exemplary destabilization domains include dihydrofolate reductase (DHFR) destabilization domains (which can be stabilized by the exemplary stabilizing agent trimethoprim), FK506-binding protein (FKBP) destabilization domains (which can be stabilized by the exemplary stabilizing agents Shield-1 (Shldl), rapamycin and FK506) and PDE5 destabilization domains (which can be stabilized by the exemplary stabilizing agents sildenafil, vardenafil, tadalafil, avanafil, lodenafil, mirodenafil, udenafil, benzamidenafil, dasantafil, and beminafil).
- DHFR dihydrofolate reductase
- FKBP FK506-binding protein
- Shield-1 Shield-1
- rapamycin and FK506 rapamycin and FK506
- PDE5 destabilization domains which can be stabilized by the exemplary stabil
- Exemplary DHFR destabilization domains are described in Iwamoto et al., 2010, Chem Biol. 17(9):981-8, Liu et al., 2014 Int. J. Parasitol. 44(10):729-735, and US 9,487,787; exemplary FKBP destabilization domains are described in Banaszynski et al., 2006, Cell 126(5):995-1104 and US 9,487,787; and exemplary PDE5 destabilization domains are described in WO 2018/237323, the contents of each of which are incorporated herein by reference in their entireties.
- Effective amount means the amount or quantity of an agent or composition that is sufficient to elicit the required or desired response, or in other words, the amount that is sufficient to elicit an appreciable biological response when administered to a subject. Said amount preferably relates to an amount that is therapeutically or in a broader sense also prophylactically effective against the progression of a disease or disorder as disclosed herein. It is understood that an “effective amount” or a “therapeutically effective amount” can vary from subject to subject, due to variation in metabolism of an agent, age, weight, general condition of the subject, the condition being treated, the severity of the condition being treated, and the judgment of the prescribing physician.
- Endonuclease refers to an enzyme that cleaves a polynucleotide chain by separating nucleotides other than nucleotides at a 5’ or 3’ end. Endonucleases differ from exonucleases, which cleave nucleotides from a 5’ or 3’ ends of a polynucleotide chain. Exemplary endonucleases include restriction endonucleases capable of cleaving double-stranded DNA at or near specific recognition sites to form double strand breaks (DSB) in the DNA.
- DSB double strand breaks
- Exemplary restriction endonucleases include XbaIR, EcoRI, Smal, Aflll, BamHI, Bell, EcoRI, Haelll, Hindll, Hindlll, Ndel, Pvull, Pstl, and Spel.
- Exemplary endonuclease amino acid sequences are described in publicly available databases such as UniProt.
- an exemplary XbaIR amino acid sequence has UniProt accession number 068567; an exemplary EcoRI amino acid sequence has UniProt accession number P00642; an exemplary Smal amino acid sequence has UniProt accession number P14229; an exemplary Afll I amino acid sequence has UniProt accession number E3VX87; an exemplary BamHI amino acid sequence has UniProt accession number P23940; an exemplary Bell amino acid sequence has UniProt accession number E5LGB8; an exemplary Haelll amino acid sequence has UniProt accession number 068584; an exemplary Hindll amino acid sequence has UniProt accession number P44413; an exemplary Hindlll amino acid sequence has UniProt accession number P43870; an exemplary Pvull amino acid sequence has UniProt accession number A0A4R7BM34; an exemplary Pstl amino acid sequence has UniProt accession number P00640; and an exemplary Spel amino acid sequence has UniProt accession number F1
- Mitochondrial Targeting Sequence refers to an amino acid sequence capable of directing the transport of a polypeptide containing the sequence to mitochondria.
- An MTS is typically 10-70 amino acids in length.
- An MTS frequently comprises an alternating pattern of hydrophobic and positively charged amino acids to form an amphipathic helix.
- an “or” conjunction is intended to be used in its correct sense as a Boolean logical operator, encompassing both the selection of features in the alternative (A or B, where the selection of A is mutually exclusive from B) and the selection of features in conjunction (A or B, where both A and B are selected).
- the term “and/or” is used for the same purpose, which shall not be construed to imply that “or” is used with reference to mutually exclusive alternatives.
- Peptide, protein, and polypeptide are used interchangeably to refer to a natural or synthetic molecule comprising two or more amino acids linked by the carboxyl group of one amino acid to the alpha amino group of another.
- the amino acids may be natural or synthetic, and can contain chemical modifications such as disulfide bridges, substitution of radioisotopes, phosphorylation, substrate chelation (e.g., chelation of iron or copper atoms), glycosylation, acetylation, formylation, amidation, biotinylation, and a wide range of other modifications.
- a polypeptide is comprised of approximately twenty standard naturally occurring amino acids, although natural and synthetic amino acids which are not members of the standard twenty amino acids may also be used.
- the standard twenty amino acids include alanine (Ala, A), arginine (Arg, R), asparagine (Asn, N), aspartic acid (Asp, D), cysteine (Cys, C), glutamine (Gin, Q), glutamic acid (Glu, E), glycine (Gly, G), histidine, (His, H), isoleucine (He, I), leucine (Leu, L), lysine (Lys, K), methionine (Met, M), phenylalanine (Phe, F), proline (Pro, P), serine (Ser, S), threonine (Thr, T), tryptophan (Trp, W), tyrosine (Tyr, Y), and valine (Vai, V).
- polypeptide sequence or “amino acid sequence” are an alphabetical representation of a polypeptide molecule.
- Percentage identity Percentage identity between two amino acid sequences is calculated by multiplying the number of matches between a pair of aligned sequences by 100, and dividing by the length of the aligned region. Identity scoring only counts perfect matches and does not consider the degree of similarity of amino acids to one another, nor does it consider substitutions or deletions as matches. Alignment for purposes of determining percent sequence identity can be achieved in various ways that are within the skill in the art, for instance, by manual alignment or using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for achieving maximum alignment.
- Subject As used herein, the term “subject” means a human.
- Treat, treating, treatment refers in one embodiment to ameliorating the disease or disorder (e.g., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms or pathological features thereof).
- “treat”, “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter or pathological features of the disease, e.g., including those, which may not be discernible by the subject.
- “treat”, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of at least one discernible or non-discernible symptom), physiologically (e.g., stabilization of a physical parameter) or both.
- “treat”, “treating” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder, or of at least one symptoms or pathological features associated thereof.
- “treat”, “treating” or “treatment” refers to preventing or delaying progression of the disease to a more advanced stage or a more serious condition.
- the benefit to a patient to be treated is either statistically significant or at least perceptible to the patient or to the physician. However, it will be appreciated that when a medicament is administered to a patient to treat a disease, the outcome may not always be an effective treatment.
- the disclosure provides polypeptides comprising a mitochondrial targeting sequence (MTS), an endonuclease sequence, and a destabilizing domain (DD) sequence.
- MTS mitochondrial targeting sequence
- DD destabilizing domain
- Exemplary features of mitochondrial targeting sequences, endonuclease and destabilizing domains that can be included in polypeptides of the disclosure are described in Sections 4.2.1 , 4.2.2, and 4.2.3, respectively.
- the MTS, endonuclease sequence, and DD can be positioned in any suitable N- terminal to C-terminal order.
- the MTS can be positioned at the N-terminal or C- terminal end of the polypeptide.
- the MTS is positioned at the N-terminal end of the polypeptide.
- the endonuclease sequence can be positioned N-terminal to the DD, or C-terminal to the DD.
- the polypeptide comprises, in N-terminal to C- terminal order, the MTS, the endonuclease sequence, and the DD sequence.
- the MTS, endonuclease sequence and the DD sequence can be directly linked, or can be separated by a spacer sequence, for example, a short amino acid sequence, for example of one, two, three, four or more amino acids.
- Mitochondria have about 1500 proteins encoded by the nuclear genome. They are translated in the cytosol, and imported to the mitochondrial inner or outer membrane, intermembrane space, or matrix, depending upon the MTS. Polypeptides of the disclosure can include a full length MTS of a mitochondrial protein, or a variant of a wild-type MTS (e.g., a truncated version of a full-length MTS and/or an MTS with one or more amino acid substitutions compared to a wild-type sequence, for example one or more conservative amino acid substitutions).
- a wild-type MTS e.g., a truncated version of a full-length MTS and/or an MTS with one or more amino acid substitutions compared to a wild-type sequence, for example one or more conservative amino acid substitutions.
- Polypeptides of the disclosure can include a human MTS or a non-human MTS (e.g., rodent such as mouse or rat or non-human primate such as cynomolgus monkey).
- an MTS of a polypeptide of the disclosure can comprise an MTS of a TCA cycle- related enzyme, a chaperone protein, a mitochondrial genome replication protein, a protease, an mRNA processing protein, a mitochondrial RNA degradation protein, a deoxynucleotide triphosphate synthesis-related protein, a mitoribosomal protein, a phospholipid metabolism- related protein, a protein involved in metabolism of toxic compounds, a disulfide relay system- related protein, an iron-sulfur protein assembly protein, a tRNA modification protein, an aminoacyl-tRNA synthetase, a release factor, or an elongation factor.
- the MTS comprises an MTS of a cytochrome c oxidase subunit (for example, either a full-length MTS or a truncated version thereof which retains mitochondrial targeting activity), for example cytochrome c oxidase subunit VIII (COX8), cytochrome c oxidase subunit X (COX10), or cytochrome c oxidase subunit IV (COX4).
- a cytochrome c oxidase subunit for example, either a full-length MTS or a truncated version thereof which retains mitochondrial targeting activity
- COX8 cytochrome c oxidase subunit VIII
- COX10 cytochrome c oxidase subunit X
- COX4 cytochrome c oxidase subunit IV
- the MTS comprises an MTS of a frataxin (FXN) protein.
- FXN frataxin
- the MTS comprises an MTS of a TCA cycle-related enzyme, for example, Pyruvate dehydrogenase, Citrate synthase, Aconitase, Isocitrate dehydrogenase, a- ketoglutarate dehydrogenase, Succinyl-CoA synthetase, Succinic dehydrogenase, Fumarase, Malate dehydrogenase, or Pyruvate carboxylase.
- the MTS comprises an MTS of a chaperone protein, for example, mtHSPIO, mtHSP60, mtHSP70, or mtHSP90.
- the MTS comprises an MTS of a mitochondrial genome replication protein, for example, TFAM, Twinkle, PolG, TFB2M, TEFM, or MTERF1 .
- a mitochondrial genome replication protein for example, TFAM, Twinkle, PolG, TFB2M, TEFM, or MTERF1 .
- the MTS comprises an MTS of a protease, for example, MPP, CLPXP, LON ATPase, or PreP.
- a protease for example, MPP, CLPXP, LON ATPase, or PreP.
- the MTS comprises an MTS of an mRNA processing protein, for example, LRPPRC, TACO1 , ELAC2, PNPT1 , HSD17B10, MTPAP, or PTCDI .
- an mRNA processing protein for example, LRPPRC, TACO1 , ELAC2, PNPT1 , HSD17B10, MTPAP, or PTCDI .
- the MTS comprises an MTS of a mitochondrial RNA degradation protein, for example, PNPasse, REX02, or SUV3.
- a mitochondrial RNA degradation protein for example, PNPasse, REX02, or SUV3.
- the MTS comprises an MTS of a deoxynucleotide triphosphate synthesis-related protein, for example, DGUOK, TK2, TYMP, MGME1 , SUCLG1 , SUCLA2, RNASEH1 , or C10orf2.
- a deoxynucleotide triphosphate synthesis-related protein for example, DGUOK, TK2, TYMP, MGME1 , SUCLG1 , SUCLA2, RNASEH1 , or C10orf2.
- the MTS comprises an MTS of a mitoribosomal protein, for example, MRPS16, MRPS22, MRPL3, MRP12, or MRPL44.
- the MTS comprises an MTS of a phospholipid metabolism-related protein, for example, AGK, SERAC1 , or TAZ.
- the MTS comprises an MTS of a protein involved in metabolism of toxic compounds, for example, HIBCH, ECHS1 , ETHE1 or MPV17.
- the MTS comprises an MTS of a disulfide relay system-related protein, for example, GFER.
- the MTS comprises an MTS of an iron-sulfur protein assembly protein, for example, ISCU, BOLA3, NFU1 , or lBA57.
- the MTS comprises an MTS of a tRNA modification protein, for example, MTO1 , GTP3BP, TRMU, PUS1 , MTFMT, TRIT1 , TRNT1 or TRMT5.
- the MTS comprises an MTS of an aminoacyl-tRNA synthetase, for example, AARS2, DARS2, EARS2, RARS2, YARS2, FARS2, HARS2, LARS2, VARS2, TARS2, IARS2, CARS2, PARS2, NARS2, KARS, GARS, SARS2 or MARS2.
- an aminoacyl-tRNA synthetase for example, AARS2, DARS2, EARS2, RARS2, YARS2, FARS2, HARS2, LARS2, VARS2, TARS2, IARS2, CARS2, PARS2, NARS2, KARS, GARS, SARS2 or MARS2.
- the MTS comprises an MTS of an elongation factor, for example, TUFM, TSFM, or GFMI .
- Exemplary mitochondrial targeting sequences are set forth in Table 1 .
- a polypeptide of the disclosure can include an MTS identified in Table 1 or a variant thereof (for example, an MTS having one or more conservative amino acid substitutions and/or a truncation).
- a truncation can be a truncation of the C-terminal sequence (for example, an MTS can correspond to a sequence set forth in Table 1 , but with a C-terminal truncation of one or more amino acids, e.g., one, two, three, four, five or more than five amino acids).
- an MTS comprises at least 15 N-terminal amino acids of a MTS sequence set forth in Table 1 .
- a variant MTS can include, for example, an MTS that is at least 80%, at least 95%, at least 90%, or at least 95% identical to an MTS listed in Table 1 .
- the endonuclease can be a restriction endonuclease, an RNA-guided endonuclease ⁇ e.g., Cas9 or Cas12), a zinc finger nuclease, or a transcription activator-like effector nuclease (TALEN).
- the endonuclease can comprise a catalytic domain (for example, from a wild-type or engineered endonuclease) and, optionally one or more additional domains, for example all domains present in a full-length wild-type or engineered endonuclease.
- Endonucleases can be bacterial in origin. Many restriction enzymes are known in the art and include, for example, XbaIR, EcoRI, Smal, Aflll, BamHI, Bell, Haelll, Hind 11, Hind I II , Ndel, Pvull, Pstl, and Spel.
- the endonuclease is XbaIR.
- An exemplary XbaIR sequence is set forth in SEQ ID NO:16:
- the endonuclease comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, more than 95%, or 100% identity with SEQ ID NO:16.
- the endonuclease is EcoRI.
- the endonuclease is Smal.
- the endonuclease is Aflll.
- the endonuclease is BamHI.
- the endonuclease is Bell.
- the endonuclease is Haelll.
- the endonuclease is Hindi I.
- the endonuclease is Hindi II .
- the endonuclease is Ndel.
- the endonuclease is Pvull.
- the endonuclease is Pstl.
- the endonuclease is Spel.
- the endonuclease sequence is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to the amino acid sequence of: UniProt accession number 068567, UniProt accession number P00642, UniProt accession number P14229, UniProt accession number E3VX87, UniProt accession number P23940, UniProt accession number E5LGB8, UniProt accession number 068584, UniProt accession number P44413, UniProt accession number P43870, UniProt accession number A0A4R7BM34, UniProt accession number P00640, or UniProt accession number F1 KM35.
- RNA-guided endonucleases such as Cas9 and Cas12 are described in US 11 ,001 ,863 B2, WO 2014/093661 and WO 2019/233990, the contents of which are incorporated herein by reference in their entireties.
- the endonuclease is SaCas9 or SpCas9.
- the polypeptide can be used in combination with one or more guide RNA molecules targeting mitochondrial DNA.
- Exemplary Zinc finger nucleases are described in WO 2001/025255, and WO 2003/066828, the contents of which are incorporated herein by reference in their entireties.
- TALEN nucleases are described in WO 2014/134412, WO 2015/013583, and WO 2013/163628, the contents of which are incorporated herein by reference in their entireties.
- the polypeptides of the disclosure include a destabilization domain (DD), which allows for ON/OFF control of the endonuclease.
- DDs include DHFR, FKBP, and PDE5 DDs.
- DHFR DDs are described in US 9,487,787, the contents of which are incorporated herein in their entirety.
- An amino acid sequence of wild-type E coli DHFR is as follows:
- a DHFR DD can comprise a wild-type DHFR sequence or can comprise one or more amino acid substitutions and/or truncations at the N and/or C terminal end.
- a DHFR DD sequence can be at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identical to SEQ ID NO:17.
- Exemplary amino acid substitutions and combinations that can be included in a DHFR DD include Y100I, G121V, N18T/A19V, F103L, H12Y/Y100I, H12L/Y100I, R98H/F103S, M42T/H114R, I61 F/T68S. Combinations of the foregoing substitutions can also be used.
- the DHFR comprises an amino acid sequence which is identical to SEQ ID NO:17 except for a Y1001, G121V, N18T/A19V, F103L, H12Y/Y100I, H12L/Y100I, R98H/F103S, M42T/H114R, or I61 F/T68S substitution(s), or a combination thereof.
- a DHFR DD lacks an N-terminal methionine.
- the DHFR comprises an amino acid sequence which is identical to SEQ ID NO:17 except for a Y1001, G121V, N18T/A19V, F103L, H12Y/Y100I, H12L/Y100I, R98H/F103S, M42T/H114R, or I61 F/T68S substitution(s), or a combination thereof, and lack of the N-terminal methionine.
- a DHFR DD has an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to ISLIAALAVDHVIGMETVMPWNLPADLAWFKRNTLNKPVIMGRHTWESIGRPLPGRKNIILSSQP STDDRVTWVKSVDEAIAACGDVPEIMVIGGGRVYEQFLPKAQKLYLTHIDAEVEGDTHFPDYEP DDWESVFSEFHDADAQNSHSYCFEILERR (SEQ ID NO:18).
- An exemplary nucleotide sequence encoding SEQ ID NO:18 is the following: atcagtctgattgcggcgttagcggtagatcacgttatcggcatggaaaccgtcatgccgtggaacctgcctgccgatctcgcctggttt aaacgcaacaccttaaataaacccgtgattatgggccgccatacctgggaatcaatcggtcgtccgttgccaggacgcaaaatatt atcctcagcagtcaaccgagtacggacgatcgcgtaacgtgggtgaagtcggtggatgaagccatcgcggcgtgtggtgacgtacc agaaatcatggttattggcggcggtcgcgtttatgaacagttcttgccaaaag
- An exemplary stabilizing agent for a DHFR DD is trimethoprim.
- Exemplary FKBP DDs are described in US 9,487,787, the contents of which are incorporated herein in their entirety.
- An amino acid sequence of an exemplary FKBP DD (having a F36V substitution compared to the wild-type sequence) is as follows:
- a FKBP DD can comprise a wild-type FKBP sequence or can comprise one or more amino acid substitutions.
- a FKBP DD sequence can be at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identical to SEQ ID NQ:20.
- Exemplary amino acid substitutions that can be included in a FKBP DD include F15S, V24A, H25R, E60G, L106P, D100G, M66T, R71G, D100N, E102G, and K105I. Combinations of the foregoing substitutions can also be used.
- the DD comprises an amino acid sequence which is identical to SEQ ID NQ:20 except for a F15S, V24A, H25R, E60G, L106P, D100G, M66T, R71G, D100N, E102G, or K105l substitution, or a combination thereof.
- Exemplary FKBP DD stabilizing agents include Shield-1 (Shldl), rapamycin and FK506.
- PDE5 DDs are described in WO 2018/237323, the contents of which are incorporated herein in their entirety.
- PDE5 DDs may be derived from PDE5A, Isoform 1 (SEQ ID NO:21); PDE5A Isoform 2 (SEQ ID NO:22) and/or PDE5A Isoform 3 (SEQ ID NO:23). These isoforms differ at their N terminal regions and have unique first exons followed by a common sequence of 823 amino acids.
- All PDE5A isoforms contain a catalytic domain that is located near the C terminus of the protein and is relatively selective for cGMP as a substrate at physiological levels.
- the substrate binding site is also the binding site for several known PDE5 inhibitors such as sildenafil, which have been utilized to treat cardiovascular diseases and erectile dysfunction.
- PDE5 inhibitors such as sildenafil
- GAF-A contains a high affinity binding site for cGMP. Occupancy of this domain by cGMP is known to cause activation of the catalytic domain.
- the affinity of this site for cGMP is increased by cGMP-dependent protein kinase-mediated phosphorylation of serine 92.
- a PDE5A DD can comprise the catalytic domain of PDE5A, spanning from amino acid position 535 to position 860 of UniProt ID: 076074 (SEQ ID NO:21), as represented in SEQ ID NO:24.
- PDE5A DDs may also comprise one or more GAF domains and/or the C terminal portion that extends beyond the catalytic domain.
- the PDE5A derived DD comprises amino acids from position 535 to position 875 of SEQ ID NO:21 .
- the PDE5 DD comprises amino acids from position 466 to 875 or position 420 to 875 of SEQ ID NO:21 .
- Exemplary PDE5 DD sequences are set forth in Table 2.
- Exemplary amino acid substitutions that can be included in PDE5 DDs include one or more amino acid substitutions selected from E535D, E536G, Q541 R, K555R, F559L S560G, F561 L, F564L, F564S, V585A, N587S, K591 E, I599V, K604E, K608E, N609H, K630R, K633E, N636S, I648V, N661 S, S663P, L675P, Y676D, Y676N, C677R, H678R, D687A, T711A, T712S, D724N, L738H, N742S, F744L, L746S, F755L, A762S, D764V, D764N, D764G, S766F, K795E, L797F, I799T, L804P.
- a PDE5 DD sequence comprises a sequence selected from the group of amino acid sequences identified by SEQ ID NOs. 19-35 of WO 2018/237323 and SEQ ID NOs.66-69 of WO 2018/237323.
- Exemplary stabilizing agents for PDE5 DDs include sildenafil, vardenafil, tadalafil, avanafil, lodenafil, mirodenafil, udenafil, benzamidenafil, dasantafil, and beminafil.
- the disclosure provides nucleic acids encoding a polypeptide of the disclosure, e.g., as described in Section 4.2.
- the nucleic acid can be, for example, a vector such as a viral genome or a plasmid, or an mRNA molecule.
- Exemplary vectors include viral expression vectors (e.g., viral vectors based on vaccinia virus; poliovirus; adenovirus (see, e.g., Li et al., 1994, Invest Opthalmol Vis Sci 35:2543-2549; Borras et al, 1999, Gene Ther 6:515-524; Li and Davidson, 1995, PNAS 92:7700-7704;
- viral expression vectors e.g., viral vectors based on vaccinia virus; poliovirus; adenovirus (see, e.g., Li et al., 1994, Invest Opthalmol Vis Sci 35:2543-2549; Borras et al, 1999, Gene Ther 6:515-524; Li and Davidson, 1995, PNAS 92:7700-7704;
- a recombinant expression vector of the present disclosure is a recombinant lentivirus vector. In some cases, a recombinant expression vector of the present disclosure is a recombinant retroviral vector.
- the vector comprises a retroviral genome. Nucleic acids, such as retroviral genomes, can be provided in the form of a particle, for example a viral particle (e.g., retroviral particle).
- Nucleic acids encoding a polypeptide of the disclosure can further include one or more regulatory sequences, for example a promoter, for example a SV40, CMV, or CAG promoter.
- a promoter for example a SV40, CMV, or CAG promoter.
- An exemplary SV40 promoter sequence is as follows:
- the disclosure provides host cells comprising a nucleic acid of the disclosure.
- Host cells can be prokaryotic (e.g., bacterial such as E. coli) or eukaryotic (e.g., a human cell line such as HEK293 or 293T).
- Host cells can be used, for example, to propagate a nucleic acid such as a retroviral genome or plasmid, or to propagate and package a particle, for example a retroviral particle.
- the disclosure provides methods of inducing mitophagy in a cell, and/or increasing mitochondrial turnover in a cell, and/or increasing mitochondrial mass, and/or inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell using the polypeptides, nucleic acids, and particles of the disclosure, e.g., polypeptides, nucleic acids, and particles as described in Sections 4.2 and 4.3.
- the methods typically comprise contacting a cell with a polypeptide, nucleic acid, or particle and a stabilizing agent capable of stabilizing the DD.
- a polypeptide can be introduced to the cell by electroporation, injection, or a carrier (e.g., lipid-based carrier such as a liposome), or any other means known in the art for delivering polypeptides to cells.
- a nucleic acid can be introduced to a cell by transfection, electroporation, injection, a carrier, or any other means known in the art for delivering nucleic acids to cells.
- a viral particle can be introduced to a cell by transduction.
- the cell can be contacted with a stabilizing agent, for example, by culturing the cell in a medium comprising the stabilizing agent.
- the cell can be cultured in the medium with the stabilizing agent for a period of time to allow the endonuclease to introduce DSBs in mitochondrial DNA.
- the cell is cultured in a medium with the stabilizing agent for at least 8 hours (e.g., at least 12 hours, at least 1 day, at least 2 days, or more) and/or up to 5 days (e.g., up to 4 days, up to 3 days, or up to 2 days).
- the stabilizing agent can be removed, for example by culturing the cell in a culture medium without the stabilizing agent. Once the stabilizing agent is removed, the polypeptide will be destabilized, leading to degradation of the polypeptide.
- the cell can be cultured for a period of time without the stabilizing agent, during which the cell can produce new mitochondria.
- the cell is cultured for at least 6 hours in a medium that does not include the stabilizing agent (e.g., at least 12 hours, at least 1 day, at least 2 days, at least 3 days, at least 4 days) and/or up to 10 days (e.g., up to 8 days, up to 6 days, or up to 4 days).
- the methods of the disclosure result in the induction of mitophagy in the cell. In some embodiments, the methods of the disclosure result in an increase in mitochondrial turnover in the cell. In some embodiments, the methods of the disclosure result in an increase in mitochondrial mass in the cell. In some embodiments, the methods result in an induction of DSBs in mitochondrial DNA in the cell. In some embodiments, the methods result in epigenomic modifications in the cell, e.g., induced by mitochondrial depletion. It has been previously reported that rhoO cells, which are completely depleted of mitochondrial genome, show significant levels of epigenomic changes (see, e.g., Hertzog Santos, 2021 Free Radio Biol Med.
- compositions of the disclosure can be used to induce epigenomic modifications.
- the methods of the disclosure result in one, two, three, four, or all five of the following in the cell: (a) induction of mitophagy, (b) increased mitochondrial turnover, (c) increased mitochondrial mass, (d) DSBs in mitochondrial DNA, and (e) epigenomic modifications.
- Exemplary cells that can be used in the methods include mammalian cells, preferably human cells, more preferably human somatic cells.
- Cell types that can be used include bone marrow cells, stem cells such as hematopoietic stem cells (HSCs) or a mesenchymal stem cells (MSCs), immune cells such as T cells, phagocytes, microglial cells, and macrophages.
- the cell is a T cell such as a CD4+ and/or CD8+ T cell.
- Primary cells obtained from a subject, as well as progeny thereof can be used.
- the cells be normal cells (e.g., from a healthy donor) or have dysfunctional mitochondria (e.g., from a subject having a disease or disorder).
- cells can be from a subject having an age-related disease or disorder, such as an autoimmune disease, a metabolic disease, a genetic disease, cancer, a neurodegenerative disease, or immunosenescence.
- cells can be from a subject having a mitochondrial disease or disorder such as chronic progressive external ophthalmoplegia (CPEO), Pearson syndrome, Kearns-Sayre syndrome (KSS), diabetes and deafness (DAD), mitochondrial diabetes, Leber hereditary optic neuropathy (LHON), LHON-plus, neuropathy, ataxia, retinitis pigmentosa syndrome (NARP), maternally inherited Leigh syndrome (MILS), mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), myoclonic epilepsy and ragged-red fiber disease (MERRF), familial bilateral striatal necrosis/striatonigral degeneration (FBSN), Lucas disease, aminoglycoside-induced Deafness (AID), or multiple deletions of mitochondrial DNA syndrome.
- CPEO chronic progressive external ophthalmoplegia
- KSS Kearns-Sayre syndrome
- DAD diabetes and deafness
- mitochondrial diabetes Leber hereditary
- Mitochondrial DNA depletion syndrome-4A mitochondrial recessive ataxia syndrome (MIRAS), mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), mitochondrial DNA depletion syndrome (MTDPS), DNA polymerase gamma (POLG)-related disorders, sensory ataxia neuropathy dysarthria ophthalmoplegia (SANDO), leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), co-enzyme Q10 deficiency, Leigh syndrome, mitochondrial complex abnormalities, fumarase deficiency, a-ketoglutarate dehydrogenase complex (KGDHC) deficiency, succinyl-CoA ligase deficiency, pyruvate dehydrogenase complex deficiency (PDHC), pyruvate carboxylase deficiency (PCD), carnitine palmitoyltransferase I (CPT I) deficiency,
- MIRAS mitochondrial
- cells can be from a subject having a neurodegenerative disease such as amyotrophic lateral sclerosis (ALS), Huntington’s disease, Alzheimer's disease, Parkinson's disease, Friedreich' s ataxia, Charcot Marie Tooth disease or leukodystrophy.
- a neurodegenerative disease such as amyotrophic lateral sclerosis (ALS), Huntington’s disease, Alzheimer's disease, Parkinson's disease, Friedreich' s ataxia, Charcot Marie Tooth disease or leukodystrophy.
- ALS amyotrophic lateral sclerosis
- APOE4 allele e.g., an E3/E4 or E4/E4 genotype.
- cells can be from a subject having an eye disease (e.g., a retinal disease) such as age-related macular degeneration, macular edema or glaucoma.
- eye disease e.g., a retinal disease
- age-related macular degeneration e.g., macular edema or glaucoma.
- cells can be from a subject having diabetes, a hearing disorder, a genetic disease (such as Hutchinson-Gilford Progeria Syndrome, Werner Syndrome, or Huntington's disease), heart failure, immunodeficiency, cancer, or an infectious disease.
- a genetic disease such as Hutchinson-Gilford Progeria Syndrome, Werner Syndrome, or Huntington's disease
- Cells obtained or obtainable by the methods described herein can be administered to a subject, for example, administered to the subject from which the cells originate or in the case of cells from a healthy donor, administered to a different subject.
- the disclosure provides a method of treating a subject having an age-related disease, mitochondrial disease or disorder, neurodegenerative disease, retinal disease, diabetes, hearing disorder, genetic disease, heart failure, immunodeficiency, cancer, or infectious disease by administering a therapeutically effective amount of cells obtained or obtainable by the methods described herein.
- the subject can have a disease or disorder described in this Section.
- the disclosure provides a pharmaceutical composition
- a pharmaceutical composition comprising a polypeptide of the disclosure (e.g., as described in Section 4.2), a nucleic acid of the disclosure, (e.g., as described in Section 4.3), a particle of the disclosure (e.g., as described in Section 4.3, or a cell of the disclosure (e.g., a obtained by a method described in 4.4) and a pharmaceutically acceptable excipient.
- compositions can be prepared by mixing a polypeptide, nucleic acid, particle, or cell with one or more physiologically acceptable carriers, excipients, or stabilizers in the form of, e.g., aqueous solutions or suspensions (see, e.g., Hardman et al., 2001 , Goodman and Gilman’s The Pharmacological Basis of Therapeutics, McGraw-Hill, New York, N.Y.; Gennaro, 2000, Remington: The Science and Practice of Pharmacy, Lippincott, Williams, and Wilkins, New York, N.Y.;Weiner and Kotkoskie, 2000, Excipient Toxicity and Safety, Marcel Dekker, Inc., New York, N.Y.).
- kits comprising a polypeptide of the disclosure (e.g., as described in Section 4.2), a nucleic acid of the disclosure, (e.g., as described in Section 4.3), or a particle of the disclosure and a stabilizing agent.
- the kit can include trimethoprim (TMP) when the DD sequence of the polypeptide is a DHFR DD sequence, Shield-1 , rapamycin, or FK506 when the DD sequence of the polypeptide is a FKBP DD sequence, or sildenafil, vardenafil, tadalafil, avanafil, lodenafil, mirodenafil, udenafil, benzamidenafil, dasantafil, or beminafil when the DD sequence of the polypeptide is a PDE5 DD sequence.
- TMP trimethoprim
- Example 1 Polypeptides for inducing mitophagy, biogenesis, and acceleration of mitochondria turnover
- This Example describes compositions and methods for simultaneously activating not only mitophagy, but also biogenesis, and for accelerating mitochondria turnover by eliminating dysfunctional mitochondria and generating new mitochondria.
- XbaIR was selected to induce DSBs in the mitochondrial genome.
- XbaIR has five cleavage sites in the mitochondrial genome consensus sequence.
- a Cox8a post-mitochondrial signal was placed on the N- terminal side of XbaIR. Inducing DSBs in the mitochondrial genome leads to a strong temporary energy depletion and, to control endonuclease activity, a DHFR destabilization domain (Liu et al., 2014 Int. J. Parasitol.
- TMP trimethoprim
- a retroviral vector having the MTS-XbalR-DHFR coding sequences was constructed (FIG. 1).
- the nucleotide sequence of the vector is shown in Table 3.
- the EGFP-DHFR retroviral vector was transduced into Hela cells. Infection efficiency was high, and TMP exposure was performed for two days at various concentrations without enriching infected cells, followed by fluorescent expression of EGFP using fluorescence microscopy and FACS (FIG. 5 and FIG. 6). EGFP mRNA was observed to be transcribed and present (FIG. 7), but the protein was not observed in the absence of TMP (FIG. 5 and FIG. 6).
- Hela transfectants with MTS-XbalR-DHFR were cloned by limiting dilution, named as Hela MXD sc20.
- Hela MXD sc20 was subject to 0.5pM TMP exposure for several time durations (16, 20, and 48 hours) to examine mitochondrial mass (mtMass), which was measured by Mito Green staining, global mitochondrial membrane potential (mtMP), which was measured by TMRM staining, and corrected mtMP with mtMass, which was calculated as the ratio of mtMP to mtMass (FIGS. 11 A-11C).
- the introduced transgene appears to have caused DSB in the mitochondrial genome and decreased ON and, in addition, MM increased very responsively to this stress despite the need for protein from the nucleus. Based on these two factors, the density of the respiratory chain complex is thought to decrease, and MMP is thought to decrease as a phenotype. The mitochondrial genome appeared to return to normal by Day 6, with MM increased slightly. Without being bound by theory, it is believed that this indicates that mitochondrial biogenesis was enhanced in the MTS-XbalR-DHFR/TMP system and that mitochondrial capacity as an abundance of mitochondria was increased.
- mtKeima-Red-transfected Hela MXD sc20 cells with PARK2 overexpression were generated in order to quantify mitophagy in a more sophisticated fashion.
- Various durations of TMP exposure to the transfectants were examined to measure mitophagy (16, 20, 24, 40, 44, and 48 hours).
- Mitophagy inducer carbonyl cyanide 3-chlorophenylhydrazone (COOP) was used as a positive control. More mitophagy was observed with increasing duration of TMP exposure (FIG. 13A). Mitophagy reached a plateau at 40 hours of TMP exposure (FIG.13A).
- measured mtDNA ON was observed to return to the initial value regardless of the duration of TMP exposure (FIG.13B).
- the results indicate that mitochondrial turnover was accelerated by MTS-XbalR-DHFR/TMP.
- This Example describes mitochondrial biogenesis and functional changes associated with genetically induced mitophagy (GiM).
- MTS-Xbal-ecDHFR was cloned into Hela cells after gene transfer using a retrovirus, and stable transfectant that constitutively expresses the GiM unit (Hela_GiM) was generated.
- the endonuclease, Xbal was transiently present in the mitochondrial matrix during treatment of Hela_GiM cells with trimethoprim (TMP) for 2 days. Since TMP was dissolved in DMSO, the control group was treated with the same amount of DMSO.
- the reactive oxygen species (ROS) over time were assessed by staining cells with mitoSox and measuring fluorescence intensity with FACS, setting a threshold line compared to stress-free cells, and measuring the percentage of positive cells.
- ROS reactive oxygen species
- the mitochondrial biogenesis- related peroxisome proliferator-activated receptor gamma coactivator 1-a PPC1a
- NRF1 nuclear factor receptor 1
- TFAM mitochondrial transcription factor A
- Mitophagy occurs when mitochondria are incorporated into the phagophore and become autolysosomes by fusion with lysosomes.
- This Example describes compositions and methods for detecting autolysosome formation and autophagy following GiM-induced transient mitochondrial genome reduction.
- Hela_GiM cells described in Section 5.2.1 were retrovirally engineered with a sequence encoding mKeimaRed. The percentage of cells undergoing mitophagy was quantified every two days for two weeks using fluorescence and phase contrast microscopy.
- the final step of autophagy flux depends on lysosomal V-ATPase activity. Therefore, the final step of flux was suppressed after GiM-induced transient mitochondrial genome reduction using a lysosomal V-ATPase inhibitor, bafilomycinAI (BafA1), to assess the mitochondria-targeted autophagy flux.
- Antibody staining for LC3 MAP1 LC3: Microtubule- associated protein 1 light chain 3
- TOM20 staining as a mitochondrial membrane marker was used to quantify autophagosomes.
- LC3-II was quantified by western blotting in Hela_GiM cells treated with BafA1 , wherein protein was extracted from the cells on Day 8 after 48 h exposure to TMP. 5.3.2. Results
- AD Alzheimer’s Disease
- This example describes how transient mitochondrial genome reduction by gene transfer can transform the cellular phenotype by enhancing mitochondrial turnover in fibroblasts obtained from a patient with AD. 5.5.1. Materials and Methods
- NHDF Normal Human Dermal Fibroblasts
- AD fibroblasts fibroblasts obtained from forearm skin samples of a patient with Alzheimer's disease whose APOE genotype is E3/E45 (AD fibroblasts) were utilized as target cells.
- Transient mitochondrial genome reduction was performed by transferring the plasmid carrying the gene encoding endonuclease XbaIR downstream of the mitochondrial transfer signal derived from human Cox8, and expressing the puromycin resistance as a selection marker under a different promoter (pCAGGS-MTS-XbaIR).
- Electroporation was used as the gene transfer method, and transfected target cells were enriched by exposure to puromycin at a concentration of 3 pg/mL for 24 hours on day 2 after electroporation. This condition was set by the criteria of 70 ⁇ 80% GFP expression and viability of more than 90%, using a plasmid with recombinant GFP gene instead of XbaIR. After gene transfer, the copy number of the mitochondrial genome was assessed on days 7, 14, and 21 to confirm genome reduction and subsequent biogenesis (FIG. 20A). In addition, mitochondrial phenotype was evaluated by measuring mitochondrial volume (mtMass) and mitochondrial membrane potential (mtMP) using MitoTracker Green and TMRM, respectively.
- mtMass mitochondrial volume
- mtMP mitochondrial membrane potential
- Both mtMass and mtMP were quantified using mean fluorescent intensity (MFI) as an indicator of fluorescence intensity. Both mtMass and mtMP were lower in untreated AD fibroblast cells relative to NHDF cells. In AD fibroblasts, mtMass and mtMP levels were even lower on day 7 after GIM induction, but increased to a level comparable to NHDF on Day 14 (FIGS. 20F-20G), suggesting that both the mitochondrial volume and mitochondrial membrane potential in AD fibroblasts were restored to the same levels as those of healthy NHDF cells by GiM.
- MFI mean fluorescent intensity
- a polypeptide comprising:
- a mitochondrial targeting sequence (a) a mitochondrial targeting sequence (MTS);
- the MTS comprises an MTS of a TCA cycle-related enzyme, a chaperone protein, a mitochondrial genome replication protein, a protease, an mRNA processing protein, a mitochondrial RNA degradation protein, a deoxynucleotide triphosphate synthesis-related protein, a mitoribosomal protein, a phospholipid metabolism-related protein, a protein involved in metabolism of toxic compounds, a disulfide relay system-related protein, an iron-sulfur protein assembly protein, a tRNA modification protein, an aminoacyl-tRNA synthetase, a release factor, or an elongation factor.
- a TCA cycle-related enzyme comprises an MTS of a TCA cycle-related enzyme, a chaperone protein, a mitochondrial genome replication protein, a protease, an mRNA processing protein, a mitochondrial RNA degradation protein, a deoxynucleotide triphosphate synthesis-related protein, a mitoribosomal protein, a
- the MTS comprises an MTS of a TCA cycle-related enzyme, optionally which is Pyruvate dehydrogenase, Citrate synthase, Aconitase, Isocitrate dehydrogenase, a-ketoglutarate dehydrogenase, Succinyl-CoA synthetase, Succinic dehydrogenase, Fumarase, Malate dehydrogenase, or Pyruvate carboxylase.
- a TCA cycle-related enzyme optionally which is Pyruvate dehydrogenase, Citrate synthase, Aconitase, Isocitrate dehydrogenase, a-ketoglutarate dehydrogenase, Succinyl-CoA synthetase, Succinic dehydrogenase, Fumarase,
- MTS comprises an MTS of a chaperone protein, optionally which is mtHSPIO, mtHSP60, mtHSP70, or mtHSP90.
- MTS comprises an MTS of a mitochondrial genome replication protein, optionally which is TFAM, Twinkle, PolG, TFB2M, TEFM, or MTERF1 .
- MTS comprises an MTS of a protease, optionally which is MPP, CLPXP, LON ATPase, or PreP.
- MTS comprises an MTS of an mRNA processing protein, optionally which is LRPPRC, TACO1 , ELAC2, PNPT1 , HSD17B10, MTPAP, or PTCD1 .
- MTS comprises an MTS of a mitochondrial RNA degradation protein, optionally which is PNPasse, REX02, or SUV3.
- MTS comprises an MTS of a deoxynucleotide triphosphate synthesis-related protein, optionally which is DGUOK, TK2, TYMP, MGME1 , SUCLG1 , SUCLA2, RNASEH1 , or C10orf2.
- a deoxynucleotide triphosphate synthesis-related protein optionally which is DGUOK, TK2, TYMP, MGME1 , SUCLG1 , SUCLA2, RNASEH1 , or C10orf2.
- MTS comprises an MTS of a mitoribosomal protein, optionally which is MRPS16, MRPS22, MRPL3, MRP12, or MRPL44.
- MTS comprises an MTS of a phospholipid metabolism-related protein, optionally which is AGK, SERAC1 , or TAZ.
- MTS comprises an MTS of a protein involved in metabolism of toxic compounds, optionally which is HIBCH, ECHS1 , ETHE1 or MPV17.
- MTS comprises an MTS of a disulfide relay system- related protein, optionally which is GFER.
- MTS comprises an MTS of an iron-sulfur protein assembly protein, optionally which is ISCU, BOLA3, NFU1 , or IBA57.
- MTS comprises an MTS of a tRNA modification protein, optionally which is MTO1 , GTP3BP, TRMU, PUS1 , MTFMT, TRIT1 , TRNT1 or TRMT5.
- MTS comprises an MTS of an aminoacyl-tRNA synthetase, optionally which is AARS2, DARS2, EARS2, RARS2, YARS2, FARS2, HARS2, LARS2, VARS2, TARS2, IARS2, CARS2, PARS2, NARS2, KARS, GARS, SARS2 or MARS2.
- aminoacyl-tRNA synthetase optionally which is AARS2, DARS2, EARS2, RARS2, YARS2, FARS2, HARS2, LARS2, VARS2, TARS2, IARS2, CARS2, PARS2, NARS2, KARS, GARS, SARS2 or MARS2.
- polypeptide of embodiment 27, wherein the MTS comprises a sequence that is at least 95% identical to MSVLTPLLLRGLTGSARR (SEQ ID NO:1).
- polypeptide of embodiment 27, wherein the MTS comprises a sequence that is 100% identical to MSVLTPLLLRGLTGSARR (SEQ ID NO:1).
- MTS comprises a sequence that is at least 80% identical to MSVLTPLLLRGLTGSARRLPVPRAKIHSL (SEQ ID NO:2).
- polypeptide of embodiment 47 wherein the MTS comprises a sequence that is at least 90% identical to MWTLGRRAVAGLLASPSPAQ (SEQ ID NO:5).
- polypeptide of embodiment 47, wherein the MTS comprises a sequence that is at least 95% identical to MWTLGRRAVAGLLASPSPAQ (SEQ ID NO:5).
- polypeptide of embodiment 47, wherein the MTS comprises a sequence that is 100% identical to MWTLGRRAVAGLLASPSPAQ (SEQ ID NO:5).
- polypeptide of embodiment 1 wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MAPYSLLVTRLQKALG (SEQ ID NO:6).
- polypeptide of embodiment 1 wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MALLTAAARLLGTKNASCLVLAARHASA (SEQ ID NOT).
- polypeptide of embodiment 1 wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MVKQIESKTAFQEALDAAGDKLVVVDFSATWC (SEQ ID NO:8).
- polypeptide of embodiment 1 wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MATNWGSLLQDKQQLEELARQAVDRALAEGVLLRTSQ (SEQ ID NO:9).
- polypeptide of embodiment 1 wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MAFLRSMWGVLSALGRSGA (SEQ ID NQ:10).
- polypeptide of embodiment 1 wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MWVLLRSGYPLRILLPLRG (SEQ ID NO:11).
- MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MSRLLWRKVAGATVGPGPVPAPG (SEQ ID NO:12).
- MSRLLWRKVAGATVGPGPVPAPG SEQ ID NO:12
- MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MKRNTLVELLTFWKNWHFRLL (SEQ ID NO:13).
- MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MISASRAAAARLVGAAASRGPTAA (SEQ ID NO:14).
- polypeptide of embodiment 1 wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MEALIPVINKLQDVFNTVGA (SEQ ID NO:15).
- polypeptide of any one of embodiments 1 to 61 wherein the endonuclease is a restriction endonuclease, an RNA-guided endonuclease (e.g., Cas9 or Cas12), a zinc finger nuclease, or a transcription activator-like effector nuclease (TALEN).
- endonuclease is a restriction endonuclease, an RNA-guided endonuclease (e.g., Cas9 or Cas12), a zinc finger nuclease, or a transcription activator-like effector nuclease (TALEN).
- polypeptide of embodiment 66, wherein the endonuclease sequence comprises a sequence that is at least 85% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16). 68.
- polypeptide of embodiment 66 wherein the endonuclease sequence comprises a sequence that is at least 90% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
- polypeptide of embodiment 66, wherein the endonuclease sequence comprises a sequence that is at least 95% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
- polypeptide of embodiment 66, wherein the endonuclease sequence comprises a sequence that is 100% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
- the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MISLIAALAVDHVIGMENAMPWNLPADLAWFKRNTLNKPVIMGRHTWESIGRPLPGRKNIILSS QPSTDDRVTWVKSVDEAIAACGDVPEIMVIGGGRVYEQFLPKAQKLYLTHIDAEVEGDTHFPD YEPDDWESVFSEFHDADAQNSHSYCFEILERR (SEQ ID NO:17).
- the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to ISLIAALAVDHVIGMETVMPWNLPADLAWFKRNTLNKPVIMGRHTWESIGRPLPGRKNIILSSQP STDDRVTWVKSVDEAIAACGDVPEIMVIGGGRVYEQFLPKAQKLYLTHIDAEVEGDTHFPDYEP DDWESVFSEFHDADAQNSHSYCFEILERR (SEQ ID NO:18).
- polypeptide of embodiment 74, wherein the destabilization domain sequence has one or more of the following amino acid substitutions: N18T/A19V, F103L, Y100I, G121V, H12Y/Y100I, H12L/Y100I, R98H/F103S, M42T/H114R, and I61 F/T68S.
- the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to GVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKVDSSRDRNKPFKFMLGKQEVIRGWEE GVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLE (SEQ ID NO:20).
- polypeptide of embodiment 77, wherein the destabilization domain sequence has one or more of the following amino acid substitutions: F15S, V24A, H25R, E60G, L106P, D100G, M66T, R71G, D100N, E102G, and K105I.
- polypeptide of any one of embodiments 1 to 71 , wherein the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to any one of SEQ ID NOs: 19-35 and 66-69 of WO 2018/237323.
- polypeptide of any one of embodiments 1 to 81 wherein the endonuclease sequence is positioned N-terminal to the destabilization domain sequence.
- polypeptide of any one of embodiments 1 to 81 wherein the endonuclease sequence is positioned C-terminal to the destabilization domain sequence.
- the nucleic acid of embodiment 85 which comprises a promoter operably linked to the nucleotide sequence encoding the polypeptide.
- nucleic acid of embodiment 86 wherein the promoter is a SV40 promoter, a CMV promoter or a CAG promoter.
- nucleic acid of embodiment 87, wherein the promoter is a SV40 promoter.
- nucleic acid of embodiment 88, wherein the SV40 promoter comprises a nucleotide sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to SEQ ID NO:25.
- nucleic acid of any one of embodiments 85 to 89 which is a vector.
- nucleic acid of embodiment 90 wherein the vector is a retroviral genome.
- nucleic acid of embodiment 91 wherein the vector is a murine retroviral genome.
- nucleic acid of embodiment 90 wherein the vector is a plasmid.
- nucleic acid of embodiment 85 which is an mRNA molecule.
- a particle comprising the nucleic acid of any one of embodiments 85 to 94, optionally wherein the particle is a retroviral particle.
- a host cell comprising the nucleic acid of any one of embodiments 85 to 94.
- a method of (a) inducing mitophagy in a cell, and/or (b) increasing mitochondrial turnover in a cell, and/or (c) increasing mitochondrial mass, and/or (d) inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell comprising contacting the cell with (i) the polypeptide of any one of embodiments 1 to 84, the nucleic acid of any one of embodiments 85 to 94, or the particle of embodiment 95 and (ii) a stabilizing agent.
- the stabilizing agent is Shield-1 , rapamycin, or FK506 when the destabilizing domain sequence is a FKBP destabilization domain sequence.
- the contacting the cell with the stabilizing agent comprises culturing the cell in a medium comprising the stabilizing agent.
- invention 101 The method of embodiment 100, which comprises culturing the cell for at least 8 hours in a medium comprising the stabilizing agent.
- invention 100 which comprises culturing the cell for at least 12 hours in a medium comprising the stabilizing agent.
- invention 100 which comprises culturing the cell for at least 1 day in a medium comprising the stabilizing agent.
- invention 100 which comprises culturing the cell for at least 2 days in a medium comprising the stabilizing agent.
- removing the stabilizing agent from the cell comprises culturing the cell in a medium that does not include the stabilizing agent.
- invention 110 which comprises culturing the cell for at 3 hours in a medium that does not include the stabilizing agent.
- the method of embodiment 110 which comprises culturing the cell for at least 6 hours in a medium that does not include the stabilizing agent. 113. The method of embodiment 110, which comprises culturing the cell for at least 12 hours in a medium that does not include the stabilizing agent.
- invention 110 which comprises culturing the cell for at least 1 day in a medium that does not include the stabilizing agent.
- invention 110 which comprises culturing the cell for at least 2 days in a medium that does not include the stabilizing agent.
- a cell comprising the polypeptide of any one of embodiments 1 to 84, the nucleic acid of any one of embodiments 85 to 94, or particle of embodiment 95.
- the cell of embodiment 134 further comprising a stabilizing agent.
- TMP trimethoprim
- HSC hematopoietic stem cell
- MSC mesenchymal stem cell
- the method or cell of embodiment 150, wherein the age-related disease is an autoimmune disease, a metabolic disease, a genetic disease, cancer, a neurodegenerative disease, or immunosenescence.
- mitochondrial disease or disorder is caused by mitochondrial DNA abnormalities, nuclear DNA abnormalities, or both.
- mitochondrial disease or disorder is chronic progressive external ophthalmoplegia (CPEO), Pearson syndrome, Kearns-Sayre syndrome (KSS), diabetes and deafness (DAD), mitochondrial diabetes, Leber hereditary optic neuropathy (LHON), LHON-plus, neuropathy, ataxia, retinitis pigmentosa syndrome (NARP), maternally inherited Leigh syndrome (MILS), mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), myoclonic epilepsy and ragged-red fiber disease (MERRF), familial bilateral striatal necrosis/striatonigral degeneration (FBSN), Lucas disease, aminoglycoside-induced Deafness (AID), or multiple deletions of mitochondrial DNA syndrome.
- CPEO chronic progressive external ophthalmoplegia
- KSS Kearns-Sayre syndrome
- DAD diabetes and deafness
- mitochondrial diabetes Leber hereditary optic neuropathy
- LHON Leber her
- mitochondrial disease or disorder is Mitochondrial DNA depletion syndrome-4A, mitochondrial recessive ataxia syndrome (MIRAS), mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), mitochondrial DNA depletion syndrome (MTDPS), DNA polymerase gamma (POLG)- related disorders, sensory ataxia neuropathy dysarthria ophthalmoplegia (SANDO), leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), co-enzyme Q10 deficiency, Leigh syndrome, mitochondrial complex abnormalities, fumarase deficiency, a- ketoglutarate dehydrogenase complex (KGDHC) deficiency, succinyl-CoA ligase deficiency, pyruvate dehydrogenase complex deficiency (PDHC), pyruvate carboxylase deficiency (PCD), carnitine palmitoy
- 157 The method or cell of embodiment 156, wherein the neurodegenerative disease is amyotrophic lateral sclerosis (ALS), Huntington’s disease, Alzheimer's disease, Parkinson's disease, Friedreich' s ataxia, Charcot Marie Tooth disease or leukodystrophy.
- ALS amyotrophic lateral sclerosis
- Huntington s disease
- Alzheimer's disease Parkinson's disease
- Friedreich' s ataxia Charcot Marie Tooth disease or leukodystrophy.
- ALS amyotrophic lateral sclerosis
- the method or cell of embodiment 156, wherein the neurodegenerative disease is Alzheimer's disease, optionally wherein the cell is from a subject having an APOE4 allele, for example an E3/E4 or E4/E4 genotype. 161 .
- the method or cell of embodiment 156, wherein the neurodegenerative disease is Parkinson's disease.
- retinal disease is age- related macular degeneration, macular edema or glaucoma.
- a method of treating a subject having an age-related disease, mitochondrial disease or disorder, neurodegenerative disease, retinal disease, diabetes, hearing disorder, genetic disease, heart failure, immunodeficiency, cancer, or infectious disease comprising administering a therapeutically effective amount of cells according to any one of embodiments 133 to 175 to the subject.
- the age-related disease is an autoimmune disease, a metabolic disease, a genetic disease, cancer, a neurodegenerative disease, or immunosenescence.
- mitochondrial disease or disorder is caused by mitochondrial DNA abnormalities, nuclear DNA abnormalities, or both.
- the mitochondrial disease or disorder is chronic progressive external ophthalmoplegia (CPEO), Pearson syndrome, Kearns-Sayre syndrome (KSS), diabetes and deafness (DAD), mitochondrial diabetes, Leber hereditary optic neuropathy (LHON), LHON-plus, neuropathy, ataxia, and retinitis pigmentosa syndrome (NARP), maternally inherited Leigh syndrome (MILS), mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), myoclonic epilepsy and ragged-red fiber disease (MERRF), familial bilateral striatal necrosis/striatonigral degeneration (FBSN), Lucas disease, aminoglycoside-induced Deafness (AID), and multiple deletions of mitochondrial DNA syndrome.
- CPEO chronic progressive external ophthalmoplegia
- KSS Kearns-Sayre syndrome
- DAD diabetes and deafness
- mitochondrial diabetes Leber hereditary optic neuropathy
- LHON Le
- mitochondrial disease or disorder is Mitochondrial DNA depletion syndrome-4A, mitochondrial recessive ataxia syndrome (MIRAS), mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), mitochondrial DNA depletion syndrome (MTDPS), DNA polymerase gamma (POLG)-related disorders, sensory ataxia neuropathy dysarthria ophthalmoplegia (SANDO), leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), co-enzyme Q10 deficiency, Leigh syndrome, mitochondrial complex abnormalities, fumarase deficiency, a- ketoglutarate dehydrogenase complex (KGDHC) deficiency, succinyl-CoA ligase deficiency, pyruvate dehydrogenase complex deficiency (PDHC), pyruvate carboxylase deficiency (PCD), carnitine palmitoyltrans
- MIRAS mitochondrial recessive ataxia syndrome
- ALS amyotrophic lateral sclerosis
- Huntington s disease
- Alzheimer's disease Parkinson's disease
- Friedreich' s ataxia Charcot Marie Tooth disease or leukodystrophy.
- retinal disease is age-related macular degeneration, macular edema or glaucoma.
- a pharmaceutical composition comprising the polypeptide of any one of embodiments 1 to 84, the nucleic acid of any one of embodiments 85 to 94, the particle of embodiment 95, or the cell of any one of embodiments 133 to 175 and a pharmaceutically acceptable excipient.
- a kit comprising (a) the polypeptide of any one of embodiments 1 to 84, the nucleic acid of any one of embodiments 85 to 94, or the particle of embodiment 95 and (b) a stabilizing agent.
- the stabilizing agent is sildenafil, vardenafil, tadalafil, avanafil, lodenafil, mirodenafil, udenafil, benzamidenafil, dasantafil, or beminafil when the destabilizing domain sequence is a PDE5 destabilization domain sequence.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Wood Science & Technology (AREA)
- Zoology (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- General Health & Medical Sciences (AREA)
- General Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Biotechnology (AREA)
- Biomedical Technology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Polypeptides having a mitochondrial targeting sequence (MTS), an endonuclease sequence, and a destabilizing domain sequence; nucleic acids encoding same; uses of the polypeptides and nucleic acids to induce mitophagy, increase mitochondrial turnover, and/or induce double strand breaks in mitochondrial DNA in a cell; and therapeutic applications of the foregoing, for example in the treatment of mitochondrial diseases and disorders.
Description
COMPOSITIONS FOR MITOPHAGY INDUCTION AND USES THEREOF
1. CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the priority benefit of U.S. provisional application no. 63/426,424, filed November 18, 2022, the contents of which are incorporated herein in their entireties by reference thereto.
2. SEQUENCE LISTING
[0002] The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML Sequence Listing, created on November 8, 2023, is named RMG-004WO_SL.xml and is 42,441 bytes in size.
1. BACKGROUND
[0003] Mitochondrial dysfunction is caused by a variety of factors, including mutations in genes encoding mitochondrial proteins, tRNA, and rRNA, accumulation of mutations in the mitochondrial genome, mismanagement of mitochondrial proteins, mismanagement of intracellular organelles such as the endoplasmic reticulum and lysosome, and mitochondrial protein quality control defects. Mitochondrial protein quality control defects can be caused by a variety of factors, such as mismanagement of intracellular organelles such as the endoplasmic reticulum and the lysosome. Among these, quality control by mitochondrial biogenesis and autophagy (mitophagy) and dynamics by fusion/fission is where various mitochondrial dysfunctions converge. Mitochondrial quality control defects associated with various diseases continue to be intensively studied as targets for disease therapy. Target diseases include not only mitochondrial diseases, but also neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and ALS, heart failure, diabetes, and immunodeficiency for cancer, infectious diseases, and autoimmune diseases.
Small molecule compounds have been developed to enhance biogenesis, to induce mitophagy, and to suppress excessive fission, etc. For example, coenzyme Q10, idebenone, and metformin have been shown to induce mitophagy.
[0004] However, no compound with a clear mode of action has yet reached the clinic. Accordingly, there remains a need for compositions and methods for ameliorating mitochondrial dysfunction.
2. SUMMARY
[0005] The disclosure provides polypeptides capable of inducing double strand breaks (DSBs) in mitochondria to enable a temporary and partial reduction in the number of mitochondria in a cell. Compared to the nucleus, mitochondria havp noor gene repair mechanisms. In response
to the stress of DSBs, mitochondria strongly transmit signals to the nucleus to promote replication of the mitochondrial genome and increase production of mitochondrial component proteins. Without being bound by theory, it is believed that introducing DSBs in mitochondrial DNA can be used to efficiently promote mitochondrial turnover, thereby ameliorating mitochondrial dysfunction through the generation of new mitochondria. DSBs can be introduced into mitochondrial DNA through use of a polypeptide comprising a mitochondrial targeting sequence (MTS) fused to an endonuclease. However, one potential problem with this approach is excessive endonuclease activity. To provide sensitive ON/OFF control of the endonuclease, the present disclosure provides polypeptides having a destabilization domain in addition to a mitochondrial targeting sequence and endonuclease sequence. When stabilized by a stabilizing agent, the destabilization domain permits the polypeptide to retain structure and endonuclease activity; in absence of the stabilizing agent, the destabilization domain loses stability leading to degradation of the polypeptide by the proteasome.
[0006] Accordingly, in one aspect, the disclosure provides polypeptides comprising a mitochondrial targeting sequence (MTS), an endonuclease sequence, for example XbaIR, and a destabilizing domain sequence. Inclusion of an MTS is useful for directing the polypeptide to the desired site of endonuclease activity, namely the mitochondrial genome. Inclusion of a destabilizing domain sequence, which is stabilized by a stabilizing agent, enables sensitive ON/OFF control of the endonuclease. For example, a cell can be contacted with the polypeptide in the presence of the stabilizing agent for a period of time (during which the polypeptide is active) and, subsequently, the stabilizing agent can be removed, thereby leading to destabilization and degradation of the polypeptide.
[0007] Exemplary features of polypeptides of the disclosure are described in Sections 4.2 and specific embodiments 1 to 84, infra.
[0008] In another aspect, the disclosure provides nucleic acids encoding a polypeptide of the disclosure, particles comprising the nucleic acids, such as viral particles, and host cells comprising a nucleic acid of the disclosure. Exemplary nucleic acids include vectors such as viral (e.g., retroviral) genomes, plasmids, and mRNA molecules. Exemplary particles include viral particles e.g., retroviral particles). Further exemplary features of nucleic acids, particles and host cells of the disclosure are described in Section 4.3 and specific embodiments 85 to 96, /nfra.
[0009] In another aspect, the disclosure provides methods of (a) inducing mitophagy in a cell, and/or (b) increasing mitochondrial turnover in a cell, and/or (c) increasing mitochondrial mass, and/or (d) inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell by contacting the cell with a polypeptide, nucleic acid, or particle of the disclosure and a stabilizing agent. For example, a cell can be transfected with a nucleic acid
encoding a polypeptide, transduced with a viral particle containing a nucleic acid encoding the polypeptide, or injected with the polypeptide, and cultured in the presence of the stabilizing agent. Following a period of time (e.g., 6 hours to 5 days), the stabilizing agent can be removed to allow destabilization and degradation of the polypeptide.
[0010] In another aspect, the disclosure provides cells and populations of cells obtained or obtainable by the methods of (a) inducing mitophagy in a cell, and/or (b) increasing mitochondrial turnover in a cell, and/or (c) increasing mitochondrial mass, and/or (d) inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell described herein. Unless required otherwise by context, reference herein to a “cell” encompasses single cells as well as populations of cells.
[0011] In another aspect, the disclosure provides methods of treating subjects with cells and populations of cells of the disclosure. For example, the subject can be a subject having an age- related disease, a mitochondrial disease or disorder, a neurodegenerative disease, an eye disease (e.g., a retinal disease), diabetes, a hearing disorder, a genetic disease, heart failure, an immunodeficiency, cancer, or an infectious disease.
[0012] Further exemplary features of methods and cells of the disclosure are described in Section 4.4 and specific embodiments 97 to 195, /nfra.
[0013] In further aspects, the disclosure provides pharmaceutical compositions comprising a polypeptide, nucleic acid, particle or cell (including populations of cells) of the disclosure. Such pharmaceutical compositions can be used, for example, in the methods of treatment described herein.
[0014] In yet another aspect, the disclosure provides a kit comprising a polypeptide, nucleic acid, or particle of the disclosure and a stabilizing agent. Kits can be used, for example, in the methods of the disclosure.
[0015] Further exemplary features of pharmaceutical compositions and kits are described in Section 4.5 and specific embodiments 196 to 200, infra.
3. BRIEF DESCRIPTION OF THE FIGURES
[0016] FIG. 1 shows a retroviral vector map with an MTS-XbalR-DHFR polypeptide (Example 1).
[0017] FIG. 2 shows a molecular model of the MTS-XbalR-DHFR polypeptide of Example 1.
[0018] FIG. 3 shows a retroviral vector map with an EFGR-DHFR polypeptide (Example 1).
[0019] FIG. 4 shows a molecular model of the EGFR-DHFR polypeptide of Example 1 .
[0020] FIG. 5 shows fluorescence microscopy images of Hela cells tranduced with an EGFP- DHFR retroviral vector (Example 1).
[0021] FIG. 6 shows FACS data for Hela cells tranduced with an EGFP-DHFR retroviral vector, showing EGFP expression when cultured in the presence of TMP (Example 1).
[0022] FIG. 7 shows relative EGFP mRNA expression levels in Hela cells tranduced with an EGFP-DHFR retroviral vector cultured with TMP (Example 1).
[0023] FIGS. 8A-8D show fluorescence intensity of Hela cells tranduced with an EGFP-DHFR retroviral vector by fluorescence microscopy and FACS after culturing in media with TMP for two days followed by washout of TMP at 0, 1 , 2, 4, 6, 8, 24, and 48 hours post-washout (Example 1). FIG. 8A: Time course; FIG. 8B: Fluorescence images; FIG. 8C: FACS analysis, FIG. 8D: Mean fluorescence intensity (MFI) in FACS analysis overtime.
[0024] FIG. 9A-9D show fluorescence intensity of Hela cells transduced with an EGFP-DHFR retroviral vector by fluorescence microscopy and FACS after culturing in media with TMP for several durations (0, 1 , 2, 4, 6, 8, 24, and 48 hours). FIG. 9A: Time course; FIG. 9B: Fluorescence images; FIG. 9C: FACS analysis; FIG. 9D: MFI in FACS analysis over time
[0025] FIG. 10A-10C show Xbal expression (FIG. 10A) and mtDNA copy number (CN) (FIG. 10B) for Hela cells transduced with a retroviral vector encoding an MTS-XbalR-DHFR polypeptide (Hela MXD sc20) and cultured overtime in the absence or presence of 0.5 pm TMP for two days followed by TMP washout (FIG. 10C) (Example 1).
[0026] FIG. 11A-11D show MFIs of Mitogreen staining that is an indicator for mitochondrial mass (mtMass) (FIG. 11 A), MFIs of TMRM staining that is an indicator for global mitochondrial membrane potential (mtMP) (FIG. 11 B), and relative TMRM/Mitogreen ratios that is an indicator for mtMP per mass unit (FIG. 11 C) in Hela MXD sc20 incubated with TMP for 3 different durations (16, 20, and 48 hours) following cell culture overtime (FIG. 11 D)
[0027] FIGS. 12A-12C show cell number (FIG. 12A) and cell viability (FIG. 12B) for Hela MXD sc20 cells cultured in the absence or presence of 0.5 pm TMP for two days followed by TMP washout (FIG. 12C). (Example 1).
[0028] FIGS. 13A-13B show mitophagy index (FIG. 13A) by FACS analysis and mtDNA CN estimated by qPCR (FIG. 13B) for Hela MDX sc20 cells transduced with a retroviral vector encoding mtKeimaRed and PARK2 and cultured without or with TMP for several durations. CCCP is used for a positive control of mitophagy at 10 pM. (Example 1).
[0029] FIGS. 14A-14D show changes in mitochondrial biogenesis in Hela_GiM cells stably expressing a genetically induced mitophagy (GiM) unit, in the presence and absence of TMP. Mitochondrial ROS (mtROS) (FIG. 14A), PGC1a (FIG. 14B), NRF1 (FIG. 14C), and TFAM (FIG. 14D) were assessed over time using FACS. (Example 2).
[0030] FIGS. 15A-15F show the expression of several mitochondrial proteins overtime in Hela_GiM cells in the presence and absence of TMP. FIG. 15A shows western blot images of mitochondrial proteins on Days 2, 4, 6, and 8. FIGS. 15B-15F show the quantified levels of the same proteins. (Example 2).
[0031] FIGS. 16A-16B show phase contrast (PhC) and fluorescence microscopy images of mtKeimaRed-expressing HeLa_GiM cells in the presence and absence of TMP (FIG. 16A) and the percentage of cells that are mitophagy positive (FIG. 16B). (Example 2).
[0032] FIGS. 17A-17B show colocalization of the autophagosomal membrane marker LC3, mitochondrial marker TOM20, and the nuclear stain DAPI in Hela_GiM cells in the presence or absence of TMP and BafA1 (FIG. 17A) and quantified values for the areas that correspond to autophagosomes (FIG. 17B) (Example 3).
[0033] FIGS. 18A-18B show LC3-II expression levels in HeLa_GiM cells in the absence or presence TMP or TMP + BafA1 . FIG. 18A shows the western blot images and FIG. 18B is a graphical representation of the triplicate quantification. (Example 3).
[0034] FIGS. 19A-19C show results of respirometry, displaying OXPHOS and glycolysis changes overtime (FIG. 19A), two-dimensional expansion of OXPHOS and Glycolysis relation (FIG. 19B), and changes in ATP production, basal respiration, proton leak, and spare capacity overtime (FIG. 19C). (Example 4).
[0035] FIGS. 20A-20G show the assay setup used in Example 5 (FIG. 20A), FACS results in untreated Alzheimer’s Disease (AD) fibroblasts and control NHDF cells (FIG. 20B) and in AD fibroblasts at 7, 14, and 21 days after genetically induced mitophagy (GiM) (FIG. 20C), mtMass and mtMP levels in untreated (FIGS. 20D and 20E, respectively) and in AD fibroblasts at 7 and 14 days after GiM, relative to controls (FIGS. 20F and 20G, respectively), which are described further in Example 5.
4. DETAILED DESCRIPTION
[0036] The disclosure provides polypeptides capable of inducing double strand breaks (DSBs) in mitochondria to enable a temporary and partial reduction in the number of mitochondria in a cell.
[0037] The mitochondrial genome encodes respiratory chain proteins that are under a strict regulation balanced with the translation of nuclear-coded respiratory proteins. Therefore, the partial loss of the mitochondrial genome directly links to insufficient proton uptake, resulting in depolarization of mitochondrial membrane potential. The mitochondrial membrane potential is depolarized in dysfunctional mitochondrial compartments, and a key regulator for mitophagy. Without being bound by theory, it is believed that the more depolarized portions can be preferentially subject to mitophagy following introduction of DSBs.
[0038] In response to the stress of DSBs, mitochondria strongly transmit signals to the nucleus (e.g., as part of the mitochondrial unfolded protein response (UPRmt)) to promote replication of the mitochondrial genome and increase production of mitochondrial component proteins. In addition to signals of the UPRmt, the metabolic alterations resulting from mitochondrial genome reduction can affect the epigenomic status of the cell. For example, the reduction can decrease some intermediates of TCA cycle, which are utilized for acetylation and methylation in the nuclear genome and histone. Without being bound by theory, it is believed that introducing DSBs in mitochondrial DNA can be used to efficiently promote mitochondrial turnover, thereby ameliorating mitochondrial dysfunction through the generation of new mitochondria.
Mitochondrial DNA may possess some modifications, such as 8-oxo-7,8-dihydroguanine (8- OXOG), which is an oxidized form of guanine. With aging, damaging modifications accumulate. Since newly generated mitochondrial genomes lack these modifications, mitochondriogenesis can promote a regain of healthy mitochondrial function.
[0039] DSBs can be introduced into mitochondrial DNA through use a polypeptide comprising a mitochondrial targeting sequence (MTS) fused to an endonuclease. However, one potential problem with this approach is excessive endonuclease activity. To provide sensitive ON/OFF control of the endonuclease, the present disclosure provides polypeptides having a destabilization domain in addition to a mitochondrial targeting sequence and endonuclease sequence. When stabilized by a stabilizing agent, the destabilization domain permits the polypeptide to retain structure and endonuclease activity; in absence of the stabilizing agent, the destabilization domain loses stability leading to degradation of the polypeptide by the proteasome.
4.1. Definitions
[0040] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. The following definitions are provided for the full understanding of terms used in this specification.
[0041] As used herein, the following terms are intended to have the following meanings:
[0042] A, An, The: As used herein, the term "a", "an", "the" and similar terms used in the context of the present disclosure (especially in the context of the claims) are to be construed to cover both the singular and plural unless otherwise indicated herein or clearly contradicted by the context. As such, the terms "a" (or "an"), "one or more", and "at least one" can be used interchangeably herein.
[0043] And/or: The term "and/or" means that each one or both or all the components or features of a list are possible variants, especially two or more thereof in an alternative or cumulative way.
[0044] Destabilization Domain (DP): The term destabilization domain refers to a polypeptide domain which when fused to a second polypeptide domain such as an endonuclease causes the polypeptide to be degraded in the absence of a stabilizing agent which prevents or inhibits the degradation otherwise elicited by the destabilization domain. Exemplary destabilization domains include dihydrofolate reductase (DHFR) destabilization domains (which can be stabilized by the exemplary stabilizing agent trimethoprim), FK506-binding protein (FKBP) destabilization domains (which can be stabilized by the exemplary stabilizing agents Shield-1 (Shldl), rapamycin and FK506) and PDE5 destabilization domains (which can be stabilized by the exemplary stabilizing agents sildenafil, vardenafil, tadalafil, avanafil, lodenafil, mirodenafil, udenafil, benzamidenafil, dasantafil, and beminafil). Exemplary DHFR destabilization domains are described in Iwamoto et al., 2010, Chem Biol. 17(9):981-8, Liu et al., 2014 Int. J. Parasitol. 44(10):729-735, and US 9,487,787; exemplary FKBP destabilization domains are described in Banaszynski et al., 2006, Cell 126(5):995-1104 and US 9,487,787; and exemplary PDE5 destabilization domains are described in WO 2018/237323, the contents of each of which are incorporated herein by reference in their entireties.
[0045] Effective amount: The term "effective amount" or "therapeutically effective amount" means the amount or quantity of an agent or composition that is sufficient to elicit the required or desired response, or in other words, the amount that is sufficient to elicit an appreciable biological response when administered to a subject. Said amount preferably relates to an amount that is therapeutically or in a broader sense also prophylactically effective against the progression of a disease or disorder as disclosed herein. It is understood that an “effective amount" or a “therapeutically effective amount" can vary from subject to subject, due to variation in metabolism of an agent, age, weight, general condition of the subject, the condition being treated, the severity of the condition being treated, and the judgment of the prescribing physician.
[0046] Endonuclease: The term “endonuclease” refers to an enzyme that cleaves a polynucleotide chain by separating nucleotides other than nucleotides at a 5’ or 3’ end. Endonucleases differ from exonucleases, which cleave nucleotides from a 5’ or 3’ ends of a polynucleotide chain. Exemplary endonucleases include restriction endonucleases capable of cleaving double-stranded DNA at or near specific recognition sites to form double strand breaks (DSB) in the DNA. Exemplary restriction endonucleases include XbaIR, EcoRI, Smal, Aflll, BamHI, Bell, EcoRI, Haelll, Hindll, Hindlll, Ndel, Pvull, Pstl, and Spel. Exemplary endonuclease amino acid sequences are described in publicly available databases such as
UniProt. For example, an exemplary XbaIR amino acid sequence has UniProt accession number 068567; an exemplary EcoRI amino acid sequence has UniProt accession number P00642; an exemplary Smal amino acid sequence has UniProt accession number P14229; an exemplary Afll I amino acid sequence has UniProt accession number E3VX87; an exemplary BamHI amino acid sequence has UniProt accession number P23940; an exemplary Bell amino acid sequence has UniProt accession number E5LGB8; an exemplary Haelll amino acid sequence has UniProt accession number 068584; an exemplary Hindll amino acid sequence has UniProt accession number P44413; an exemplary Hindlll amino acid sequence has UniProt accession number P43870; an exemplary Pvull amino acid sequence has UniProt accession number A0A4R7BM34; an exemplary Pstl amino acid sequence has UniProt accession number P00640; and an exemplary Spel amino acid sequence has UniProt accession number F1 KM35.
[0047] Mitochondrial Targeting Sequence (MTS): The term “mitochondrial targeting sequence” refers to an amino acid sequence capable of directing the transport of a polypeptide containing the sequence to mitochondria. An MTS is typically 10-70 amino acids in length. An MTS frequently comprises an alternating pattern of hydrophobic and positively charged amino acids to form an amphipathic helix.
[0048] Or: Unless indicated otherwise, an “or” conjunction is intended to be used in its correct sense as a Boolean logical operator, encompassing both the selection of features in the alternative (A or B, where the selection of A is mutually exclusive from B) and the selection of features in conjunction (A or B, where both A and B are selected). In some places in the text, the term “and/or” is used for the same purpose, which shall not be construed to imply that “or” is used with reference to mutually exclusive alternatives.
[0049] Peptide, protein, and polypeptide: The terms peptide, protein, and polypeptide are used interchangeably to refer to a natural or synthetic molecule comprising two or more amino acids linked by the carboxyl group of one amino acid to the alpha amino group of another. The amino acids may be natural or synthetic, and can contain chemical modifications such as disulfide bridges, substitution of radioisotopes, phosphorylation, substrate chelation (e.g., chelation of iron or copper atoms), glycosylation, acetylation, formylation, amidation, biotinylation, and a wide range of other modifications. There is expressly no requirement that a polypeptide must contain an intended function; a polypeptide can be functional, non-functional, function for unexpected/unintended purposes, or have unknown function. A polypeptide is comprised of approximately twenty standard naturally occurring amino acids, although natural and synthetic amino acids which are not members of the standard twenty amino acids may also be used. The standard twenty amino acids include alanine (Ala, A), arginine (Arg, R), asparagine (Asn, N), aspartic acid (Asp, D), cysteine (Cys, C), glutamine (Gin, Q), glutamic acid (Glu, E), glycine (Gly, G), histidine, (His, H), isoleucine (He, I), leucine (Leu, L), lysine (Lys, K),
methionine (Met, M), phenylalanine (Phe, F), proline (Pro, P), serine (Ser, S), threonine (Thr, T), tryptophan (Trp, W), tyrosine (Tyr, Y), and valine (Vai, V). The terms “polypeptide sequence” or “amino acid sequence” are an alphabetical representation of a polypeptide molecule.
[0050] Percentage identity: Percentage identity between two amino acid sequences is calculated by multiplying the number of matches between a pair of aligned sequences by 100, and dividing by the length of the aligned region. Identity scoring only counts perfect matches and does not consider the degree of similarity of amino acids to one another, nor does it consider substitutions or deletions as matches. Alignment for purposes of determining percent sequence identity can be achieved in various ways that are within the skill in the art, for instance, by manual alignment or using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for achieving maximum alignment.
[0051] Subject: As used herein, the term “subject" means a human.
[0052] Treat, treating, treatment: As used herein, the term “treat”, “treating" or "treatment" of any disease or disorder refers in one embodiment to ameliorating the disease or disorder (e.g., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms or pathological features thereof). In another embodiment “treat”, "treating" or "treatment" refers to alleviating or ameliorating at least one physical parameter or pathological features of the disease, e.g., including those, which may not be discernible by the subject. In yet another embodiment, “treat”, "treating" or "treatment" refers to modulating the disease or disorder, either physically, (e.g., stabilization of at least one discernible or non-discernible symptom), physiologically (e.g., stabilization of a physical parameter) or both. In yet another embodiment, “treat”, "treating" or "treatment" refers to preventing or delaying the onset or development or progression of the disease or disorder, or of at least one symptoms or pathological features associated thereof. In yet another embodiment, “treat”, "treating" or "treatment" refers to preventing or delaying progression of the disease to a more advanced stage or a more serious condition. The benefit to a patient to be treated is either statistically significant or at least perceptible to the patient or to the physician. However, it will be appreciated that when a medicament is administered to a patient to treat a disease, the outcome may not always be an effective treatment.
4.2. Polypeptides
[0053] In one aspect, the disclosure provides polypeptides comprising a mitochondrial targeting sequence (MTS), an endonuclease sequence, and a destabilizing domain (DD) sequence. Exemplary features of mitochondrial targeting sequences, endonuclease and destabilizing domains that can be included in polypeptides of the disclosure are described in Sections 4.2.1 , 4.2.2, and 4.2.3, respectively.
[0054] The MTS, endonuclease sequence, and DD can be positioned in any suitable N- terminal to C-terminal order. For example, the MTS can be positioned at the N-terminal or C- terminal end of the polypeptide. In some embodiments, the MTS is positioned at the N-terminal end of the polypeptide. The endonuclease sequence can be positioned N-terminal to the DD, or C-terminal to the DD. In some embodiments, the polypeptide comprises, in N-terminal to C- terminal order, the MTS, the endonuclease sequence, and the DD sequence. The MTS, endonuclease sequence and the DD sequence can be directly linked, or can be separated by a spacer sequence, for example, a short amino acid sequence, for example of one, two, three, four or more amino acids.
4.2.1. Mitochondrial Targeting Sequences
[0055] Mitochondria have about 1500 proteins encoded by the nuclear genome. They are translated in the cytosol, and imported to the mitochondrial inner or outer membrane, intermembrane space, or matrix, depending upon the MTS. Polypeptides of the disclosure can include a full length MTS of a mitochondrial protein, or a variant of a wild-type MTS (e.g., a truncated version of a full-length MTS and/or an MTS with one or more amino acid substitutions compared to a wild-type sequence, for example one or more conservative amino acid substitutions).
[0056] Polypeptides of the disclosure can include a human MTS or a non-human MTS (e.g., rodent such as mouse or rat or non-human primate such as cynomolgus monkey). For example, an MTS of a polypeptide of the disclosure can comprise an MTS of a TCA cycle- related enzyme, a chaperone protein, a mitochondrial genome replication protein, a protease, an mRNA processing protein, a mitochondrial RNA degradation protein, a deoxynucleotide triphosphate synthesis-related protein, a mitoribosomal protein, a phospholipid metabolism- related protein, a protein involved in metabolism of toxic compounds, a disulfide relay system- related protein, an iron-sulfur protein assembly protein, a tRNA modification protein, an aminoacyl-tRNA synthetase, a release factor, or an elongation factor.
[0057] In some embodiments, the MTS comprises an MTS of a cytochrome c oxidase subunit (for example, either a full-length MTS or a truncated version thereof which retains mitochondrial targeting activity), for example cytochrome c oxidase subunit VIII (COX8), cytochrome c oxidase subunit X (COX10), or cytochrome c oxidase subunit IV (COX4).
[0058] In some embodiments, the MTS comprises an MTS of a frataxin (FXN) protein.
[0059] In some embodiments, the MTS comprises an MTS of a TCA cycle-related enzyme, for example, Pyruvate dehydrogenase, Citrate synthase, Aconitase, Isocitrate dehydrogenase, a- ketoglutarate dehydrogenase, Succinyl-CoA synthetase, Succinic dehydrogenase, Fumarase, Malate dehydrogenase, or Pyruvate carboxylase.
[0060] In other embodiments, the MTS comprises an MTS of a chaperone protein, for example, mtHSPIO, mtHSP60, mtHSP70, or mtHSP90.
[0061] In other embodiments, the MTS comprises an MTS of a mitochondrial genome replication protein, for example, TFAM, Twinkle, PolG, TFB2M, TEFM, or MTERF1 .
[0062] In other embodiments, the MTS comprises an MTS of a protease, for example, MPP, CLPXP, LON ATPase, or PreP.
[0063] In other embodiments, the MTS comprises an MTS of an mRNA processing protein, for example, LRPPRC, TACO1 , ELAC2, PNPT1 , HSD17B10, MTPAP, or PTCDI .
[0064] In other embodiments, the MTS comprises an MTS of a mitochondrial RNA degradation protein, for example, PNPasse, REX02, or SUV3.
[0065] In other embodiments, the MTS comprises an MTS of a deoxynucleotide triphosphate synthesis-related protein, for example, DGUOK, TK2, TYMP, MGME1 , SUCLG1 , SUCLA2, RNASEH1 , or C10orf2.
[0066] In other embodiments, the MTS comprises an MTS of a mitoribosomal protein, for example, MRPS16, MRPS22, MRPL3, MRP12, or MRPL44.
[0067] In other embodiments, the MTS comprises an MTS of a phospholipid metabolism- related protein, for example, AGK, SERAC1 , or TAZ.
[0068] In other embodiments, the MTS comprises an MTS of a protein involved in metabolism of toxic compounds, for example, HIBCH, ECHS1 , ETHE1 or MPV17.
[0069] In other embodiments, the MTS comprises an MTS of a disulfide relay system-related protein, for example, GFER.
[0070] In other embodiments, the MTS comprises an MTS of an iron-sulfur protein assembly protein, for example, ISCU, BOLA3, NFU1 , or lBA57.
[0071] In other embodiments, the MTS comprises an MTS of a tRNA modification protein, for example, MTO1 , GTP3BP, TRMU, PUS1 , MTFMT, TRIT1 , TRNT1 or TRMT5.
[0072] In other embodiments, the MTS comprises an MTS of an aminoacyl-tRNA synthetase, for example, AARS2, DARS2, EARS2, RARS2, YARS2, FARS2, HARS2, LARS2, VARS2, TARS2, IARS2, CARS2, PARS2, NARS2, KARS, GARS, SARS2 or MARS2.
[0073] In other embodiments, the MTS comprises an MTS of an elongation factor, for example, TUFM, TSFM, or GFMI .
[0075] A polypeptide of the disclosure can include an MTS identified in Table 1 or a variant thereof (for example, an MTS having one or more conservative amino acid substitutions and/or a truncation). A truncation can be a truncation of the C-terminal sequence (for example, an MTS can correspond to a sequence set forth in Table 1 , but with a C-terminal truncation of one or more amino acids, e.g., one, two, three, four, five or more than five amino acids). In some embodiments, an MTS comprises at least 15 N-terminal amino acids of a MTS sequence set forth in Table 1 . A variant MTS can include, for example, an MTS that is at least 80%, at least 95%, at least 90%, or at least 95% identical to an MTS listed in Table 1 .
[0076] Those of skill in the art will appreciate that additional mitochondrial targeting sequences beyond those identified in this section can be used. Various tools for predicting MTS can be used to identify additional mitochondrial targeting sequences, including SignalP (Bendtsen et al., 2004, J. Mol. Biol. 340:783-795; Teufel et al., 2022 Nat Biotechnol. doi.org/10.1038/s41587- 021-01156-3), MitoFates (Fukasawa et al., 2015 Mol Cell Proteomics 14(4):1113-1126), and MitoProt (Claros, 1995, Comput Apl Biosci. 11 (4):441 -7).
4.2.2. Endonuclease
[0077] Various endonucleases can be used in the polypeptides of the disclosure. For example, the endonuclease can be a restriction endonuclease, an RNA-guided endonuclease {e.g., Cas9 or Cas12), a zinc finger nuclease, or a transcription activator-like effector nuclease (TALEN).The endonuclease can comprise a catalytic domain (for example, from a wild-type or
engineered endonuclease) and, optionally one or more additional domains, for example all domains present in a full-length wild-type or engineered endonuclease.
[0078] Endonucleases can be bacterial in origin. Many restriction enzymes are known in the art and include, for example, XbaIR, EcoRI, Smal, Aflll, BamHI, Bell, Haelll, Hind 11, Hind I II , Ndel, Pvull, Pstl, and Spel.
[0079] In some embodiments, the endonuclease is XbaIR. An exemplary XbaIR sequence is set forth in SEQ ID NO:16:
MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
[0080] In some embodiments, the endonuclease comprises an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, more than 95%, or 100% identity with SEQ ID NO:16.
[0081] In some embodiments, the endonuclease is EcoRI.
[0082] In some embodiments, the endonuclease is Smal.
[0083] In some embodiments, the endonuclease is Aflll.
[0084] In some embodiments, the endonuclease is BamHI.
[0085] In some embodiments, the endonuclease is Bell.
[0086] In some embodiments, the endonuclease is Haelll.
[0087] In some embodiments, the endonuclease is Hindi I.
[0088] In some embodiments, the endonuclease is Hindi II .
[0089] In some embodiments, the endonuclease is Ndel.
[0090] In some embodiments, the endonuclease is Pvull.
[0091] In some embodiments, the endonuclease is Pstl.
[0092] In some embodiments, the endonuclease is Spel.
[0093] In some embodiments, the endonuclease sequence is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to the amino acid sequence of: UniProt accession number 068567, UniProt accession number P00642, UniProt accession number P14229, UniProt accession number E3VX87, UniProt accession number P23940, UniProt accession number E5LGB8, UniProt accession number 068584, UniProt accession number P44413,
UniProt accession number P43870, UniProt accession number A0A4R7BM34, UniProt accession number P00640, or UniProt accession number F1 KM35.
[0094] Exemplary RNA-guided endonucleases such as Cas9 and Cas12 are described in US 11 ,001 ,863 B2, WO 2014/093661 and WO 2019/233990, the contents of which are incorporated herein by reference in their entireties. In some embodiments, the endonuclease is SaCas9 or SpCas9. When an RNA-guided endonuclease is used, the polypeptide can be used in combination with one or more guide RNA molecules targeting mitochondrial DNA.
[0095] Exemplary Zinc finger nucleases are described in WO 2001/025255, and WO 2003/066828, the contents of which are incorporated herein by reference in their entireties.
[0096] Exemplary TALEN nucleases are described in WO 2014/134412, WO 2015/013583, and WO 2013/163628, the contents of which are incorporated herein by reference in their entireties.
4.2.3. Destabilizing Domain
[0097] The polypeptides of the disclosure include a destabilization domain (DD), which allows for ON/OFF control of the endonuclease. Exemplary DDs include DHFR, FKBP, and PDE5 DDs.
[0098] Exemplary DHFR DDs are described in US 9,487,787, the contents of which are incorporated herein in their entirety. An amino acid sequence of wild-type E coli DHFR is as follows:
MISLIAALAVDHVIGMENAMPWNLPADLAWFKRNTLNKPVIMGRHTWESIGRPLPGRKNIILSS QPSTDDRVTWVKSVDEAIAACGDVPEIMVIGGGRVYEQFLPKAQKLYLTHIDAEVEGDTHFPD YEPDDWESVFSEFHDADAQNSHSYCFEILERR (SEQ ID NO:17)
[0099] A DHFR DD can comprise a wild-type DHFR sequence or can comprise one or more amino acid substitutions and/or truncations at the N and/or C terminal end. For example, a DHFR DD sequence can be at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identical to SEQ ID NO:17. Exemplary amino acid substitutions and combinations that can be included in a DHFR DD include Y100I, G121V, N18T/A19V, F103L, H12Y/Y100I, H12L/Y100I, R98H/F103S, M42T/H114R, I61 F/T68S. Combinations of the foregoing substitutions can also be used. In some embodiments, the DHFR comprises an amino acid sequence which is identical to SEQ ID NO:17 except for a Y1001, G121V, N18T/A19V, F103L, H12Y/Y100I, H12L/Y100I, R98H/F103S, M42T/H114R, or I61 F/T68S substitution(s), or a combination thereof. In some embodiments, a DHFR DD lacks an N-terminal methionine. For example, in some embodiments, the DHFR comprises an amino acid sequence which is identical to SEQ ID NO:17 except for a Y1001, G121V, N18T/A19V, F103L, H12Y/Y100I, H12L/Y100I,
R98H/F103S, M42T/H114R, or I61 F/T68S substitution(s), or a combination thereof, and lack of the N-terminal methionine.
[0100] In some embodiments, a DHFR DD has an amino acid sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to ISLIAALAVDHVIGMETVMPWNLPADLAWFKRNTLNKPVIMGRHTWESIGRPLPGRKNIILSSQP STDDRVTWVKSVDEAIAACGDVPEIMVIGGGRVYEQFLPKAQKLYLTHIDAEVEGDTHFPDYEP DDWESVFSEFHDADAQNSHSYCFEILERR (SEQ ID NO:18). An exemplary nucleotide sequence encoding SEQ ID NO:18 is the following: atcagtctgattgcggcgttagcggtagatcacgttatcggcatggaaaccgtcatgccgtggaacctgcctgccgatctcgcctggttt aaacgcaacaccttaaataaacccgtgattatgggccgccatacctgggaatcaatcggtcgtccgttgccaggacgcaaaaatatt atcctcagcagtcaaccgagtacggacgatcgcgtaacgtgggtgaagtcggtggatgaagccatcgcggcgtgtggtgacgtacc agaaatcatggttattggcggcggtcgcgtttatgaacagttcttgccaaaagcgcaaaaactgtatctgacgcatatcgacgcagaa gtggaaggcgacacccatttcccggattacgagccggatgactgggaatcggtattcagcgaattccacgatgctgatgcgcagaa ctctcacagctattgctttgagattctggagcggcgataa (SEQ ID NO:19).
[0101] An exemplary stabilizing agent for a DHFR DD is trimethoprim.
[0102] Exemplary FKBP DDs are described in US 9,487,787, the contents of which are incorporated herein in their entirety. An amino acid sequence of an exemplary FKBP DD (having a F36V substitution compared to the wild-type sequence) is as follows:
GVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKVDSSRDRNKPFKFMLGKQEVIRGWEE GVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLE (SEQ ID NQ:20)
[0103] A FKBP DD can comprise a wild-type FKBP sequence or can comprise one or more amino acid substitutions. For example, a FKBP DD sequence can be at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identical to SEQ ID NQ:20. Exemplary amino acid substitutions that can be included in a FKBP DD include F15S, V24A, H25R, E60G, L106P, D100G, M66T, R71G, D100N, E102G, and K105I. Combinations of the foregoing substitutions can also be used. In some embodiments, the DD comprises an amino acid sequence which is identical to SEQ ID NQ:20 except for a F15S, V24A, H25R, E60G, L106P, D100G, M66T, R71G, D100N, E102G, or K105l substitution, or a combination thereof.
[0104] Exemplary FKBP DD stabilizing agents include Shield-1 (Shldl), rapamycin and FK506.
[0105] Exemplary PDE5 DDs are described in WO 2018/237323, the contents of which are incorporated herein in their entirety. PDE5 DDs may be derived from PDE5A, Isoform 1 (SEQ ID NO:21); PDE5A Isoform 2 (SEQ ID NO:22) and/or PDE5A Isoform 3 (SEQ ID NO:23). These isoforms differ at their N terminal regions and have unique first exons followed by a common sequence of 823 amino acids.
[0106] All PDE5A isoforms contain a catalytic domain that is located near the C terminus of the protein and is relatively selective for cGMP as a substrate at physiological levels. The substrate binding site is also the binding site for several known PDE5 inhibitors such as sildenafil, which have been utilized to treat cardiovascular diseases and erectile dysfunction. Towards the N terminus, two homologous GAF domains are located. One of the GAF domains, GAF-A contains a high affinity binding site for cGMP. Occupancy of this domain by cGMP is known to cause activation of the catalytic domain. Moreover, the affinity of this site for cGMP is increased by cGMP-dependent protein kinase-mediated phosphorylation of serine 92. In another embodiment, a PDE5A DD can comprise the catalytic domain of PDE5A, spanning from amino acid position 535 to position 860 of UniProt ID: 076074 (SEQ ID NO:21), as represented in SEQ ID NO:24. In addition to the catalytic domain, PDE5A DDs may also comprise one or more GAF domains and/or the C terminal portion that extends beyond the catalytic domain. In one embodiment, the PDE5A derived DD comprises amino acids from position 535 to position 875 of SEQ ID NO:21 . In another embodiment, the PDE5 DD comprises amino acids from position 466 to 875 or position 420 to 875 of SEQ ID NO:21 . Exemplary PDE5 DD sequences are set forth in Table 2.
[0107] Exemplary amino acid substitutions that can be included in PDE5 DDs include one or more amino acid substitutions selected from E535D, E536G, Q541 R, K555R, F559L S560G, F561 L, F564L, F564S, V585A, N587S, K591 E, I599V, K604E, K608E, N609H, K630R, K633E, N636S, I648V, N661 S, S663P, L675P, Y676D, Y676N, C677R, H678R, D687A, T711A, T712S, D724N, L738H, N742S, F744L, L746S, F755L, A762S, D764V, D764N, D764G, S766F, K795E, L797F, I799T, L804P.T802P, S815C, M816A, M816T, I824T, C839S, F840S, and K852E. The PDE5 DDs can also contain additional substitutions such as Q589R. In some embodiments, a PDE5 DD sequence comprises a sequence selected from the group of amino acid sequences identified by SEQ ID NOs. 19-35 of WO 2018/237323 and SEQ ID NOs.66-69 of WO 2018/237323.
[0108] Exemplary stabilizing agents for PDE5 DDs include sildenafil, vardenafil, tadalafil, avanafil, lodenafil, mirodenafil, udenafil, benzamidenafil, dasantafil, and beminafil.
4.3. Nucleic acids, particles and host cells
[0109] In another aspect, the disclosure provides nucleic acids encoding a polypeptide of the disclosure, e.g., as described in Section 4.2. The nucleic acid can be, for example, a vector such as a viral genome or a plasmid, or an mRNA molecule.
[0110] Exemplary vectors include viral expression vectors (e.g., viral vectors based on vaccinia virus; poliovirus; adenovirus (see, e.g., Li et al., 1994, Invest Opthalmol Vis Sci 35:2543-2549; Borras et al, 1999, Gene Ther 6:515-524; Li and Davidson, 1995, PNAS 92:7700-7704;
Sakamoto et al., 1999, H Gene Ther 5:1088-1097, 1999; WO 94/12649, WO 93/03769; WO 93/19191 ; WO 94/28938; WO 95/11984 and WO 95/00655); adeno-associated virus (AAV) (see, e.g., Ali et al., 1998, Hum Gene Ther 9:81 86; Flannery et al., 1997, PNAS 94:6916-6921 ; Bennett et al., 1997, Invest Opthalmol Vis Sci 38:2857-2863; Jomary et al., 1997, Gene Ther 4:683 690; Rolling et al., 1999, Hum Gene Ther 10:641-648; Ali et al., 1996, Hum Mol Genet 5:591-594; WO 93/09239); SV40; herpes simplex virus; human immunodeficiency virus (see, e.g., Miyoshi et al., 1997, PNAS 94:10319-23; Takahashi et al., 1999, J Virol 73:7812-7816); a retroviral vector (e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, a lentivirus, human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumor virus); and the like. In some cases, a recombinant expression vector of the present disclosure is a recombinant lentivirus vector. In some cases, a recombinant expression vector of the present disclosure is a recombinant retroviral vector.
[0111] In some embodiments, the vector comprises a retroviral genome. Nucleic acids, such as retroviral genomes, can be provided in the form of a particle, for example a viral particle (e.g., retroviral particle).
[0112] Nucleic acids encoding a polypeptide of the disclosure can further include one or more regulatory sequences, for example a promoter, for example a SV40, CMV, or CAG promoter. An exemplary SV40 promoter sequence is as follows:
GTGTGTCAGTTAGGGTGTGGAAAGTCCCCAGGCTCCCCAGCAGGCAGAAGTATGCAAAG CATGCATCTCAATTAGTCAGCAACCAGGTGTGGAAAGTCCCCAGGCTCCCCAGCAGGCAG AAGTATGCAAAGCATGCATCTCAATTAGTCAGCAACCATAGTCCCGCCCCTAACTCCGCCC ATCCCGCCCCTAACTCCGCCCAGTTCCGCCCATTCTCCGCCCCATGGCTGACTAATTTTTT TTATTTATGCAGAGGCCGAGGCCGCCTCGGCCTCTGAGCTATTCCAGAAGTAGTGAGGAG GCTTTTTTGGAGGCCTAGGCTTTTGCAAA (SEQ ID NO:25).
[0113] In another aspect, the disclosure provides host cells comprising a nucleic acid of the disclosure. Host cells can be prokaryotic (e.g., bacterial such as E. coli) or eukaryotic (e.g., a human cell line such as HEK293 or 293T). Host cells can be used, for example, to propagate a nucleic acid such as a retroviral genome or plasmid, or to propagate and package a particle, for example a retroviral particle.
4.4. Methods of inducing mitophagy and methods of treatment
[0114] In further aspects, the disclosure provides methods of inducing mitophagy in a cell, and/or increasing mitochondrial turnover in a cell, and/or increasing mitochondrial mass, and/or inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell using the polypeptides, nucleic acids, and particles of the disclosure, e.g., polypeptides, nucleic acids, and particles as described in Sections 4.2 and 4.3.
[0115] The methods typically comprise contacting a cell with a polypeptide, nucleic acid, or particle and a stabilizing agent capable of stabilizing the DD. A polypeptide can be introduced to the cell by electroporation, injection, or a carrier (e.g., lipid-based carrier such as a liposome), or any other means known in the art for delivering polypeptides to cells. A nucleic acid can be introduced to a cell by transfection, electroporation, injection, a carrier, or any other means known in the art for delivering nucleic acids to cells. A viral particle can be introduced to a cell by transduction.
[0116] The cell can be contacted with a stabilizing agent, for example, by culturing the cell in a medium comprising the stabilizing agent. The cell can be cultured in the medium with the stabilizing agent for a period of time to allow the endonuclease to introduce DSBs in mitochondrial DNA. In some embodiments, the cell is cultured in a medium with the stabilizing agent for at least 8 hours (e.g., at least 12 hours, at least 1 day, at least 2 days, or more) and/or
up to 5 days (e.g., up to 4 days, up to 3 days, or up to 2 days). Subsequently, the stabilizing agent can be removed, for example by culturing the cell in a culture medium without the stabilizing agent. Once the stabilizing agent is removed, the polypeptide will be destabilized, leading to degradation of the polypeptide.
[0117] Following removal of the stabilizing agent, the cell can be cultured for a period of time without the stabilizing agent, during which the cell can produce new mitochondria. In some embodiments, the cell is cultured for at least 6 hours in a medium that does not include the stabilizing agent (e.g., at least 12 hours, at least 1 day, at least 2 days, at least 3 days, at least 4 days) and/or up to 10 days (e.g., up to 8 days, up to 6 days, or up to 4 days).
[0118] In some embodiments, the methods of the disclosure result in the induction of mitophagy in the cell. In some embodiments, the methods of the disclosure result in an increase in mitochondrial turnover in the cell. In some embodiments, the methods of the disclosure result in an increase in mitochondrial mass in the cell. In some embodiments, the methods result in an induction of DSBs in mitochondrial DNA in the cell. In some embodiments, the methods result in epigenomic modifications in the cell, e.g., induced by mitochondrial depletion. It has been previously reported that rhoO cells, which are completely depleted of mitochondrial genome, show significant levels of epigenomic changes (see, e.g., Hertzog Santos, 2021 Free Radio Biol Med. 170:69-69). Thus, it is believed that compositions of the disclosure can be used to induce epigenomic modifications. In some embodiments, the methods of the disclosure result in one, two, three, four, or all five of the following in the cell: (a) induction of mitophagy, (b) increased mitochondrial turnover, (c) increased mitochondrial mass, (d) DSBs in mitochondrial DNA, and (e) epigenomic modifications.
[0119] Exemplary cells that can be used in the methods include mammalian cells, preferably human cells, more preferably human somatic cells. Cell types that can be used include bone marrow cells, stem cells such as hematopoietic stem cells (HSCs) or a mesenchymal stem cells (MSCs), immune cells such as T cells, phagocytes, microglial cells, and macrophages. In some embodiment, the cell is a T cell such as a CD4+ and/or CD8+ T cell. Primary cells obtained from a subject, as well as progeny thereof can be used.
[0120] The cells be normal cells (e.g., from a healthy donor) or have dysfunctional mitochondria (e.g., from a subject having a disease or disorder). For example, cells can be from a subject having an age-related disease or disorder, such as an autoimmune disease, a metabolic disease, a genetic disease, cancer, a neurodegenerative disease, or immunosenescence.
[0121] As another example, cells can be from a subject having a mitochondrial disease or disorder such as chronic progressive external ophthalmoplegia (CPEO), Pearson syndrome, Kearns-Sayre syndrome (KSS), diabetes and deafness (DAD), mitochondrial diabetes, Leber hereditary optic neuropathy (LHON), LHON-plus, neuropathy, ataxia, retinitis pigmentosa
syndrome (NARP), maternally inherited Leigh syndrome (MILS), mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), myoclonic epilepsy and ragged-red fiber disease (MERRF), familial bilateral striatal necrosis/striatonigral degeneration (FBSN), Luft disease, aminoglycoside-induced Deafness (AID), or multiple deletions of mitochondrial DNA syndrome. Additional mitochondrial diseases and disorders include Mitochondrial DNA depletion syndrome-4A, mitochondrial recessive ataxia syndrome (MIRAS), mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), mitochondrial DNA depletion syndrome (MTDPS), DNA polymerase gamma (POLG)-related disorders, sensory ataxia neuropathy dysarthria ophthalmoplegia (SANDO), leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), co-enzyme Q10 deficiency, Leigh syndrome, mitochondrial complex abnormalities, fumarase deficiency, a-ketoglutarate dehydrogenase complex (KGDHC) deficiency, succinyl-CoA ligase deficiency, pyruvate dehydrogenase complex deficiency (PDHC), pyruvate carboxylase deficiency (PCD), carnitine palmitoyltransferase I (CPT I) deficiency, carnitine palmitoyltransferase II (CPT IT) deficiency, carnitine-acyl-carnitine (CACT) deficiency, autosomal dominant-/ autosomal recessive- progressive external ophthalmoplegia (ad-Zar-PEO), infantile onset spinal cerebellar atrophy (IOSCA), mitochondrial myopathy (MM) spinal muscular atrophy (SMA), growth retardation, aminoaciduria, cholestasis, iron overload, early death (GRACILE), and Charcot-Marie-Tooth disease type 2 A (CMT2A).
[0122] As another example, cells can be from a subject having a neurodegenerative disease such as amyotrophic lateral sclerosis (ALS), Huntington’s disease, Alzheimer's disease, Parkinson's disease, Friedreich' s ataxia, Charcot Marie Tooth disease or leukodystrophy. In some embodiments, cells are from a subject having Alzheimer's disease, for example a subject having an APOE4 allele, e.g., an E3/E4 or E4/E4 genotype.
[0123] As yet another example, cells can be from a subject having an eye disease (e.g., a retinal disease) such as age-related macular degeneration, macular edema or glaucoma.
[0124] In yet further examples, cells can be from a subject having diabetes, a hearing disorder, a genetic disease (such as Hutchinson-Gilford Progeria Syndrome, Werner Syndrome, or Huntington's disease), heart failure, immunodeficiency, cancer, or an infectious disease.
[0125] Cells obtained or obtainable by the methods described herein can be administered to a subject, for example, administered to the subject from which the cells originate or in the case of cells from a healthy donor, administered to a different subject.
[0126] Accordingly, in another aspect, the disclosure provides a method of treating a subject having an age-related disease, mitochondrial disease or disorder, neurodegenerative disease, retinal disease, diabetes, hearing disorder, genetic disease, heart failure, immunodeficiency, cancer, or infectious disease by administering a therapeutically effective amount of cells
obtained or obtainable by the methods described herein. For example, the subject can have a disease or disorder described in this Section.
4.5. Pharmaceutical compositions and kits
[0100] In another aspect, the disclosure provides a pharmaceutical composition comprising a polypeptide of the disclosure (e.g., as described in Section 4.2), a nucleic acid of the disclosure, (e.g., as described in Section 4.3), a particle of the disclosure (e.g., as described in Section 4.3, or a cell of the disclosure (e.g., a obtained by a method described in 4.4) and a pharmaceutically acceptable excipient. For example, pharmaceutical compositions can be prepared by mixing a polypeptide, nucleic acid, particle, or cell with one or more physiologically acceptable carriers, excipients, or stabilizers in the form of, e.g., aqueous solutions or suspensions (see, e.g., Hardman et al., 2001 , Goodman and Gilman’s The Pharmacological Basis of Therapeutics, McGraw-Hill, New York, N.Y.; Gennaro, 2000, Remington: The Science and Practice of Pharmacy, Lippincott, Williams, and Wilkins, New York, N.Y.;Weiner and Kotkoskie, 2000, Excipient Toxicity and Safety, Marcel Dekker, Inc., New York, N.Y.).
[0127] In another aspect, the disclosure provides kits comprising a polypeptide of the disclosure (e.g., as described in Section 4.2), a nucleic acid of the disclosure, (e.g., as described in Section 4.3), or a particle of the disclosure and a stabilizing agent. For example, the kit can include trimethoprim (TMP) when the DD sequence of the polypeptide is a DHFR DD sequence, Shield-1 , rapamycin, or FK506 when the DD sequence of the polypeptide is a FKBP DD sequence, or sildenafil, vardenafil, tadalafil, avanafil, lodenafil, mirodenafil, udenafil, benzamidenafil, dasantafil, or beminafil when the DD sequence of the polypeptide is a PDE5 DD sequence.
5. EXAMPLES
5.1. Example 1 : Polypeptides for inducing mitophagy, biogenesis, and acceleration of mitochondria turnover
This Example describes compositions and methods for simultaneously activating not only mitophagy, but also biogenesis, and for accelerating mitochondria turnover by eliminating dysfunctional mitochondria and generating new mitochondria.
5.1.1. Design of Transgenes
[0128] XbaIR was selected to induce DSBs in the mitochondrial genome. XbaIR has five cleavage sites in the mitochondrial genome consensus sequence. To direct the XbaIR endonuclease to the mitochondria, a Cox8a post-mitochondrial signal was placed on the N- terminal side of XbaIR. Inducing DSBs in the mitochondrial genome leads to a strong temporary energy depletion and, to control endonuclease activity, a DHFR destabilization domain (Liu et al., 2014 Int. J. Parasitol. 44(10):729-735) was fused to the C-terminal end of XbaIR to allow
sensitive ON/OFF control of the endonuclease. The DHFR destabilization domain is stabilized by the antibiotic trimethoprim (TMP).
[0129] A retroviral vector having the MTS-XbalR-DHFR coding sequences was constructed (FIG. 1). The nucleotide sequence of the vector is shown in Table 3.
[0130] Molecular simulation was used to confirm that the three functional domains of the polypeptide construct (MTS, Xbal, and DHFR) adopt a three-dimensional structure without interfering with each other's structure (FIG. 2). In addition, a retroviral vector with EGFP-DHFR as the transgene was created to evaluate the responsiveness of the DHFR/TMP system (FIG. 3 and FIG. 4). The nucleotide sequence of the vector is shown in Table 4.
5.1.2. Responsiveness of DHFR/TMP system
[0131] The EGFP-DHFR retroviral vector was transduced into Hela cells. Infection efficiency was high, and TMP exposure was performed for two days at various concentrations without enriching infected cells, followed by fluorescent expression of EGFP using fluorescence microscopy and FACS (FIG. 5 and FIG. 6). EGFP mRNA was observed to be transcribed and present (FIG. 7), but the protein was not observed in the absence of TMP (FIG. 5 and FIG. 6).
[0132] To confirm the OFF control of the construct, medium washout was performed after two days of TMP exposure, and the fluorescence intensity over time was checked by fluorescence microscopy and FACS (FIG. 8A). The EGFP fluorescence abruptly declined even one hour after removing TMP in the culture medium, then four hours later, the fluorescence dissipated (FIG. 8B and FIG 8C). Additionally, thereafter expression of the transgene product was not observed to leak out in the 48 hours following TMP OFF (FIG. 8D).
[0133] To confirm the ON control of the construct, addition of TMP was performed, and the fluorescence intensity overtime was checked by fluorescence microscopy and FACS (FIG. 9A). The EGFP fluorescence abruptly turned on even one hour after adding TMP in the culture medium, then six hours later, the fluorescence reached to over 80% of the maximum intensity (FIG. 9B and FIG. 9C). From eight hours later, the intensity plateaued until the 48 hour time point (FIG. 9D).
5.1.3. Mitochondria Characteristics following Genetically Induced Mitophagy
[0134] Hela cells were transfected with the MTS-XbalR-DHFR vector and exposed to 0.5 pM TMP for two days followed by washout. XbaRI RNA expression was measured following TMP exposure. The transcript level of the transgene was not significantly changed (FIG. 10A). On Day 2, CN decreased to less than half of the initial value (FIG. 10B).
[0135] Hela transfectants with MTS-XbalR-DHFR were cloned by limiting dilution, named as Hela MXD sc20. Hela MXD sc20 was subject to 0.5pM TMP exposure for several time durations (16, 20, and 48 hours) to examine mitochondrial mass (mtMass), which was measured by Mito Green staining, global mitochondrial membrane potential (mtMP), which was measured by TMRM staining, and corrected mtMP with mtMass, which was calculated as the ratio of mtMP to mtMass (FIGS. 11 A-11C). Regardless of the duration of TMP exposure, the mtMass temporally increased about to two-fold compared to the resting state, then returned to the initial volume at 10 hours following TMP ON, suggesting that mitochondrial biogenesis was temporarily and strongly activated (FIG. 11 A). Both the global and corrected mtMP
demonstrated a sharp decline and then increase, indicating poor pumping up of hydrogen ions through either respiratory chain complex I, III, and IV or counter clock-wise rotated complex V (FIG. 11 B and FIG. 11C).
[0136] The introduced transgene appears to have caused DSB in the mitochondrial genome and decreased ON and, in addition, MM increased very responsively to this stress despite the need for protein from the nucleus. Based on these two factors, the density of the respiratory chain complex is thought to decrease, and MMP is thought to decrease as a phenotype. The mitochondrial genome appeared to return to normal by Day 6, with MM increased slightly. Without being bound by theory, it is believed that this indicates that mitochondrial biogenesis was enhanced in the MTS-XbalR-DHFR/TMP system and that mitochondrial capacity as an abundance of mitochondria was increased.
[0137] Next, it was investigated whether changes in cell proliferation and viability occurred in this system and affected the above-described changes. No significant difference in cell number or viability in this system with or without TMP was observed (FIGS. 12A-12B). Thus, the main benefit of this system was determined to be the intervention on mitochondria by the MTS- XbalR-DHFR polypeptide.
5.1.4. Effects on Mitophagy following MTS-XbalR-DHFR/TMP
[0138] mtKeima-Red-transfected Hela MXD sc20 cells with PARK2 overexpression were generated in order to quantify mitophagy in a more sophisticated fashion. Various durations of TMP exposure to the transfectants were examined to measure mitophagy (16, 20, 24, 40, 44, and 48 hours). Mitophagy inducer carbonyl cyanide 3-chlorophenylhydrazone (COOP) was used as a positive control. More mitophagy was observed with increasing duration of TMP exposure (FIG. 13A). Mitophagy reached a plateau at 40 hours of TMP exposure (FIG.13A). Simultaneously, measured mtDNA ON was observed to return to the initial value regardless of the duration of TMP exposure (FIG.13B). In total, the results indicate that mitochondrial turnover was accelerated by MTS-XbalR-DHFR/TMP.
5.2. Example 2: Changes in mitochondrial functioning and biogenesis due to genetically induced mitophagy
[0139] This Example describes mitochondrial biogenesis and functional changes associated with genetically induced mitophagy (GiM).
5.2.1. Materials and Methods
[0140] MTS-Xbal-ecDHFR was cloned into Hela cells after gene transfer using a retrovirus, and stable transfectant that constitutively expresses the GiM unit (Hela_GiM) was generated. The endonuclease, Xbal, was transiently present in the mitochondrial matrix during treatment of Hela_GiM cells with trimethoprim (TMP) for 2 days. Since TMP was dissolved in DMSO, the control group was treated with the same amount of DMSO.
[0141] The reactive oxygen species (ROS) over time were assessed by staining cells with mitoSox and measuring fluorescence intensity with FACS, setting a threshold line compared to stress-free cells, and measuring the percentage of positive cells. The mitochondrial biogenesis- related peroxisome proliferator-activated receptor gamma coactivator 1-a (PGC1a), nuclear factor receptor 1 (NRF1), which is involved in mitochondriogenesis, and mitochondrial transcription factor A (TFAM), which forms nucleoids with mtDNA and is deeply involved in mtDNA transcription, replication, and maintenance, were assessed with qPCR. Expression levels of several nuclear-encoded and mtDNA-encoded proteins were quantified via Western Blotting.
5.2.2. Results
[0142] The level of ROS increased initially in the TMP-treated group but was essentially unchanged in the control group throughout the assessments. On Day 2, the percentage of mitoSox-positive cells in the TMP-treated group was higher than in the control group. On Day 4 and onward, the percentage of mitoSox-positive cells were comparable for the two groups (FIG. 14A). Mitochondriogenesis-promoting transcription factors PGC1a and NRF1 were also found to be elevated in the TMP-treated group relative to the control group at all observation points (FIGS. 14B and 14C) and TFAM was largely maintained on Days 2, 4, and 6 in the TMP-treated group (FIG. 14D).
[0143] Without being bound by theory, it is believed that these results indicate that ROS production is temporarily enhanced by transient mitochondrial genome reduction, whereby mitochondrial biogenesis is brought about by amplification of transcription factors such as PGC1a and NRF1 . It is further believed, again without being bound by theory, that the maintenance of the elevated transcript levels of TFAM, a major constituent protein of nucleoids, indicates that mitochondrial biogenesis continues for some time after mitochondrial genome reduction has been triggered.
[0144] Next, the effect of GiM on the expression of nuclear-encoded and mtDNA-encoded proteins was assessed. The expression level of ATP5A, a respiratory chain complex encoded in the nucleus, was essentially unchanged by mitochondrial genome reduction (FIGS. 15A and 15B). The expression of translocase of the outer membrane (TOM20), also encoded in the nucleus, was reduced immediately after mitochondrial genome reduction but returned to a level comparable to that observed in the DMSO-treated group by Day 6 (FIGS. 15A and 15C). On the other hand, ATP6, which is encoded by mtDNA, was greatly reduced immediately after mitochondrial genome reduction in the TMP-treated group, then increased on Day 4, and later returned to a comparable level as the control group (FIGS. 15A and 15D). This trend was also observed with the ATP6/ATP5A ratio (FIGS. 15A and 15E). These results indicate that GiM
causes a transient surge of mitochondrial proteins derived from the mitochondrial genome, but not from the nuclear genome.
[0145] Changes in the level of the activated form of AMPK, p-AMPK, were used to estimate changes in mitochondrial energy production after mitochondrial genome reduction. Mitochondrial energy production was confirmed by an increase in p-AMPK on Day 4 (FIGS. 15A and 15F). Transient mitochondrial genome reduction was associated with a reduced competence of the respiratory chain complex on Day 2, and the effect was directly linked to cellular energy depletion, leading to a marked increase in p-AMPK on Day 4.
5.3. Example 3: Autophagy associated with GiM-induced transient mitochondrial genome reduction
[0146] Mitophagy occurs when mitochondria are incorporated into the phagophore and become autolysosomes by fusion with lysosomes. This Example describes compositions and methods for detecting autolysosome formation and autophagy following GiM-induced transient mitochondrial genome reduction.
5.3.1. Materials and Methods
[0147] Autolysosomes have lower pH values than mitochondria that are not fused with lysosomes. Using a pH-sensitive mitochondrial reporter, mtKeimaRed, autolysosome formation was assessed in cells after GiM-induced transient mitochondrial genome reduction. mtKeimaRed emits fluorescence with a peak at 440 nm (green) at pH > 6 and a peak at 620 nm (red) at pH < 5 and has a transport signal that allows its transport into mitochondria. Cells that stably express mKeimaRed emit red light when the environmental pH is below 5. To quantify autolysosomes upon GiM-induced transient mitochondrial genome reduction, Hela_GiM cells described in Section 5.2.1 were retrovirally engineered with a sequence encoding mKeimaRed. The percentage of cells undergoing mitophagy was quantified every two days for two weeks using fluorescence and phase contrast microscopy.
[0148] The final step of autophagy flux depends on lysosomal V-ATPase activity. Therefore, the final step of flux was suppressed after GiM-induced transient mitochondrial genome reduction using a lysosomal V-ATPase inhibitor, bafilomycinAI (BafA1), to assess the mitochondria-targeted autophagy flux. Antibody staining for LC3 (MAP1 LC3: Microtubule- associated protein 1 light chain 3), a representative marker for autophagosome formation, along with TOM20 staining as a mitochondrial membrane marker, was used to quantify autophagosomes. Given that LC3 is present on autophagosome membranes as LC3-II with PE, LC3-II was quantified by western blotting in Hela_GiM cells treated with BafA1 , wherein protein was extracted from the cells on Day 8 after 48 h exposure to TMP.
5.3.2. Results
[0149] The peak of mtKeimaRed signal associated with GiM-induced autolysosome formation was on Day 8 (FIG. 16A), with mitophagy occurring in about 20% of cells (FIG. 16B). The time course analysis suggested that kinetics of mitophagy increased on Day 6 and decreased to control levels by Day 14, with only a few percent of cells undergoing mitophagy (FIG. 16B). This transient surge in induced mitophagy was not associated with cell death or a decrease in viability. Without being bound by theory, these results suggest that the adverse reactions of excess mitophagy, such as mitophagy-induced cell death, can be suppressed by controlling GiM.
[0150] Autophagosomes formed upon fusion of mitochondria and lysosomes were detected as spots where LC3 staining and TOM20 staining overlap (FIG. 17A). In the absence of TMP, the spots with overlapping LC3 and TOM20 staining were about 80 pm2 in size and were not significantly affected by the use of BafA1 .On the other hand, in the TMP-exposed group, the area of spots with overlapping LC3 and TOM20 staining significantly increased to about 120 pm2 with the addition of BafA1 (FIG. 17B). Although LC3-II was found to be significantly increased by GiM, the increase was more pronounced and significant when BafA1 was used (FIGS. 18A and 18B). Taken together, these results suggest that GiM significantly promotes autophagy.
5.4. Example 4: Metabolic Effects of Mitochondrial Genome Reduction
[0151] Mitochondrial genome reduction was performed using Hela_GiM cells described in Section 5.2.1 with 2 days of TMP exposure. Respirometry using SeaHorse overtime was performed to evaluate oxidative phosphorylation (OXPHOS) and glycolysis. OXPHOS (FIG. 19A, left panel) decreased until day 4 and gradually increased from day 6 until approaching the starting level on day 10. Glycolysis, on the other hand, increased until day 8 and then decreased by day 10 (FIG. 19A, right panel). The two-dimensional plot depicting OXPHOS and Glycolysis relationship, showed a circular change, indicating that the metabolic change was transient (FIG. 19B). Individual assessments of ATP production, basal respiration, proton leak, and spare capacity further supported the transient nature of GiM-associated metabolic changes (FIG. 19C). Even in terms of oxygen consumption, the metabolic effects of GiM indicate that the temporary mitochondrial genome reduction is a reversible change.
5.5. Example 5: Genetically Induced Mitophagy in AD Fibroblasts
[0152] Alzheimer’s Disease (AD) is associated with mitochondrial dysfunction. This example describes how transient mitochondrial genome reduction by gene transfer can transform the cellular phenotype by enhancing mitochondrial turnover in fibroblasts obtained from a patient with AD.
5.5.1. Materials and Methods
[0153] Normal Human Dermal Fibroblasts (NHDF) and fibroblasts obtained from forearm skin samples of a patient with Alzheimer's disease whose APOE genotype is E3/E45 (AD fibroblasts) were utilized as target cells. Transient mitochondrial genome reduction was performed by transferring the plasmid carrying the gene encoding endonuclease XbaIR downstream of the mitochondrial transfer signal derived from human Cox8, and expressing the puromycin resistance as a selection marker under a different promoter (pCAGGS-MTS-XbaIR). Electroporation was used as the gene transfer method, and transfected target cells were enriched by exposure to puromycin at a concentration of 3 pg/mL for 24 hours on day 2 after electroporation. This condition was set by the criteria of 70~80% GFP expression and viability of more than 90%, using a plasmid with recombinant GFP gene instead of XbaIR. After gene transfer, the copy number of the mitochondrial genome was assessed on days 7, 14, and 21 to confirm genome reduction and subsequent biogenesis (FIG. 20A). In addition, mitochondrial phenotype was evaluated by measuring mitochondrial volume (mtMass) and mitochondrial membrane potential (mtMP) using MitoTracker Green and TMRM, respectively.
5.5.2. Results
[0154] In untreated AD fibroblasts mtMP was significantly decreased relative to untreated NHDF (FIGS. 20B and 20E). Similarly, mtMass was decreased in untreated AD fibroblasts relative to untreated NHDF (FIG. 20B and 20D). The two fluorescent signals were expanded in two dimensions, and a quadrant analysis was performed using NHDFs as a positive control to set a threshold line. The fraction ratio is used as a biomarker of aging in lymphocytes for detecting mitochondrial dysfunction. This double positive rate was found to be about half of that of NHDF. The percentage of double positives did not change significantly on Day 7 after genetically induced mitophagy (GiM), but increased to 60.3% on Day 14 and furthermore to 87.3% on Day 21 , comparable to the control NHDF (FIG. 20C).
[0155] Both mtMass and mtMP were quantified using mean fluorescent intensity (MFI) as an indicator of fluorescence intensity. Both mtMass and mtMP were lower in untreated AD fibroblast cells relative to NHDF cells. In AD fibroblasts, mtMass and mtMP levels were even lower on day 7 after GIM induction, but increased to a level comparable to NHDF on Day 14 (FIGS. 20F-20G), suggesting that both the mitochondrial volume and mitochondrial membrane potential in AD fibroblasts were restored to the same levels as those of healthy NHDF cells by GiM.
[0156] These results indicate that the promotion of mitochondrial turnover by GiM is qualitatively favorable for nascent generated mitochondria. Given that accumulation of dysfunctional mitochondria is a common phenomenon in various neurogenerative diseases and
in aging, GiM can be employed as a therapeutic strategy for the treatment of neurodegenerative diseases and also for the reacquisition of normal function in aging cells.
6. SPECIFIC EMBODIMENTS
[0157] The present disclosure is exemplified by the specific embodiments below.
1 . A polypeptide comprising:
(a) a mitochondrial targeting sequence (MTS);
(b) an endonuclease sequence; and
(c) a destabilizing domain sequence.
2. The polypeptide of embodiment 1 , wherein the MTS comprises a human MTS.
3. The polypeptide of embodiment 1 , wherein the MTS comprises a non-human MTS.
4. The polypeptide of any one of embodiments 1 to 3, wherein the MTS comprises an MTS of a mitochondrial protein.
5. The polypeptide of any one of embodiments 1 to 4, wherein the MTS comprises an MTS of a TCA cycle-related enzyme, a chaperone protein, a mitochondrial genome replication protein, a protease, an mRNA processing protein, a mitochondrial RNA degradation protein, a deoxynucleotide triphosphate synthesis-related protein, a mitoribosomal protein, a phospholipid metabolism-related protein, a protein involved in metabolism of toxic compounds, a disulfide relay system-related protein, an iron-sulfur protein assembly protein, a tRNA modification protein, an aminoacyl-tRNA synthetase, a release factor, or an elongation factor.
6. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a cytochrome c oxidase subunit.
7. The polypeptide of embodiment 6, wherein the MTS comprises an MTS of a cytochrome c oxidase subunit VIII (COX8).
8. The polypeptide of embodiment 6, wherein the MTS comprises an MTS of a cytochrome c oxidase subunit X (COX10).
9. The polypeptide of embodiment 6, wherein the MTS comprises an MTS of a cytochrome c oxidase subunit IV (COX4).
10. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a frataxin (FXN) protein.
11 . The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a TCA cycle-related enzyme, optionally which is Pyruvate dehydrogenase, Citrate synthase, Aconitase, Isocitrate dehydrogenase, a-ketoglutarate dehydrogenase, Succinyl-CoA synthetase, Succinic dehydrogenase, Fumarase, Malate dehydrogenase, or Pyruvate carboxylase.
12. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a chaperone protein, optionally which is mtHSPIO, mtHSP60, mtHSP70, or mtHSP90.
13. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a mitochondrial genome replication protein, optionally which is TFAM, Twinkle, PolG, TFB2M, TEFM, or MTERF1 .
14. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a protease, optionally which is MPP, CLPXP, LON ATPase, or PreP.
15. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of an mRNA processing protein, optionally which is LRPPRC, TACO1 , ELAC2, PNPT1 , HSD17B10, MTPAP, or PTCD1 .
16. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a mitochondrial RNA degradation protein, optionally which is PNPasse, REX02, or SUV3.
17. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a deoxynucleotide triphosphate synthesis-related protein, optionally which is DGUOK, TK2, TYMP, MGME1 , SUCLG1 , SUCLA2, RNASEH1 , or C10orf2.
18. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a mitoribosomal protein, optionally which is MRPS16, MRPS22, MRPL3, MRP12, or MRPL44.
19. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a phospholipid metabolism-related protein, optionally which is AGK, SERAC1 , or TAZ.
20. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a protein involved in metabolism of toxic compounds, optionally which is HIBCH, ECHS1 , ETHE1 or MPV17.
21 . The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a disulfide relay system- related protein, optionally which is GFER.
22. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of an iron-sulfur protein assembly protein, optionally which is ISCU, BOLA3, NFU1 , or IBA57.
23. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a tRNA modification protein, optionally which is MTO1 , GTP3BP, TRMU, PUS1 , MTFMT, TRIT1 , TRNT1 or TRMT5.
24. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of an aminoacyl-tRNA synthetase, optionally which is AARS2, DARS2, EARS2, RARS2, YARS2, FARS2, HARS2, LARS2, VARS2, TARS2, IARS2, CARS2, PARS2, NARS2, KARS, GARS, SARS2 or MARS2.
25. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of a release factor, optionally which is a C12orf65.
26. The polypeptide of any one of embodiments 1 to 5, wherein the MTS comprises an MTS of an elongation factor, optionally which is TUFM, TSFM, or GFM1 .
27. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80% identical to MSVLTPLLLRGLTGSARR (SEQ ID NO:1).
28. The polypeptide of embodiment 27, wherein the MTS comprises a sequence that is at least 85% identical to MSVLTPLLLRGLTGSARR (SEQ ID NO:1).
29. The polypeptide of embodiment 27, wherein the MTS comprises a sequence that is at least 90% identical to MSVLTPLLLRGLTGSARR (SEQ ID NO:1).
30. The polypeptide of embodiment 27, wherein the MTS comprises a sequence that is at least 95% identical to MSVLTPLLLRGLTGSARR (SEQ ID NO:1).
31 . The polypeptide of embodiment 27, wherein the MTS comprises a sequence that is 100% identical to MSVLTPLLLRGLTGSARR (SEQ ID NO:1).
32. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80% identical to MSVLTPLLLRGLTGSARRLPVPRAKIHSL (SEQ ID NO:2).
33. The polypeptide of embodiment 32, wherein the MTS comprises a sequence that is at least 85% identical to MSVLTPLLLRGLTGSARRLPVPRAKIHSL (SEQ ID NO:2).
48. The polypeptide of embodiment 47, wherein the MTS comprises a sequence that is at least 85% identical to MWTLGRRAVAGLLASPSPAQ (SEQ ID NO:5).
49. The polypeptide of embodiment 47, wherein the MTS comprises a sequence that is at least 90% identical to MWTLGRRAVAGLLASPSPAQ (SEQ ID NO:5).
50. The polypeptide of embodiment 47, wherein the MTS comprises a sequence that is at least 95% identical to MWTLGRRAVAGLLASPSPAQ (SEQ ID NO:5).
51 . The polypeptide of embodiment 47, wherein the MTS comprises a sequence that is 100% identical to MWTLGRRAVAGLLASPSPAQ (SEQ ID NO:5).
52. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MAPYSLLVTRLQKALG (SEQ ID NO:6).
53. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MALLTAAARLLGTKNASCLVLAARHASA (SEQ ID NOT).
54. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MVKQIESKTAFQEALDAAGDKLVVVDFSATWC (SEQ ID NO:8).
55. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MATNWGSLLQDKQQLEELARQAVDRALAEGVLLRTSQ (SEQ ID NO:9).
56. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MAFLRSMWGVLSALGRSGA (SEQ ID NQ:10).
57. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MWVLLRSGYPLRILLPLRG (SEQ ID NO:11).
58. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MSRLLWRKVAGATVGPGPVPAPG (SEQ ID NO:12).
59. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MKRNTLVELLTFWKNWHFRLL (SEQ ID NO:13).
60. The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MISASRAAAARLVGAAASRGPTAA (SEQ ID NO:14).
61 . The polypeptide of embodiment 1 , wherein the MTS comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MEALIPVINKLQDVFNTVGA (SEQ ID NO:15).
62. The polypeptide of any one of embodiments 1 to 61 , wherein the endonuclease is a restriction endonuclease, an RNA-guided endonuclease (e.g., Cas9 or Cas12), a zinc finger nuclease, or a transcription activator-like effector nuclease (TALEN).
63. The polypeptide of embodiment 62, wherein the endonuclease is a restriction endonuclease.
64. The polypeptide of embodiment 63, wherein the restriction endonuclease is an XbaIR, EcoRI, Smal, Aflll, BamHI, Bell, Haelll, Hindi I, Hind II I , Ndel, Pvull, Pstl, or Spel endonuclease.
65. The polypeptide of embodiment 64, wherein the restriction endonuclease is an XbaIR endonuclease.
66. The polypeptide of any one of embodiments 1 to 63, wherein the endonuclease comprises a sequence that is at least 80% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
67. The polypeptide of embodiment 66, wherein the endonuclease sequence comprises a sequence that is at least 85% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
68. The polypeptide of embodiment 66, wherein the endonuclease sequence comprises a sequence that is at least 90% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
69. The polypeptide of embodiment 66, wherein the endonuclease sequence comprises a sequence that is at least 95% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
70. The polypeptide of embodiment 66, wherein the endonuclease sequence comprises a sequence that is 100% identical to MTTLEKIKLLADGYADRLKLAIDGRVLEMQGDDVSHYLIYRVLGVAQEEGRLIDVYQNKGRFLY KYAGSFLEAATKLCFKEAFPDSASLRLPNTQGQRPRTVEIDCLVGNDALEIKWKDATTDGDHIT KEHTRIKVISDAGYKPIRIMFYYPHRTQAIRIQETLETLYNGVHGEYHYGEAAWDYVLQRTSVNL KVALEQIADSRTNEAA (SEQ ID NO:16).
71. The polypeptide of any one of embodiments 1 to 70, wherein the destabilizing domain sequence is a DHFR, FKBP, or PDE5 destabilization domain sequence.
72. The polypeptide of embodiment 71 , wherein the destabilization domain sequence is a DHFR destabilization domain sequence.
73. The polypeptide of embodiment 72, wherein the destabilization domain sequence is an E. coli DHFR (ecDHFR) destabilization domain sequence.
74. The polypeptide of any one of embodiments 1 to 73, wherein the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to MISLIAALAVDHVIGMENAMPWNLPADLAWFKRNTLNKPVIMGRHTWESIGRPLPGRKNIILSS QPSTDDRVTWVKSVDEAIAACGDVPEIMVIGGGRVYEQFLPKAQKLYLTHIDAEVEGDTHFPD YEPDDWESVFSEFHDADAQNSHSYCFEILERR (SEQ ID NO:17).
75. The polypeptide of any one of embodiments 1 to 73, wherein the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to
ISLIAALAVDHVIGMETVMPWNLPADLAWFKRNTLNKPVIMGRHTWESIGRPLPGRKNIILSSQP STDDRVTWVKSVDEAIAACGDVPEIMVIGGGRVYEQFLPKAQKLYLTHIDAEVEGDTHFPDYEP DDWESVFSEFHDADAQNSHSYCFEILERR (SEQ ID NO:18).
76. The polypeptide of embodiment 74, wherein the destabilization domain sequence has one or more of the following amino acid substitutions: N18T/A19V, F103L, Y100I, G121V, H12Y/Y100I, H12L/Y100I, R98H/F103S, M42T/H114R, and I61 F/T68S.
77. The polypeptide of any one of embodiments 1 to 71 , wherein the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to GVQVETISPGDGRTFPKRGQTCVVHYTGMLEDGKKVDSSRDRNKPFKFMLGKQEVIRGWEE GVAQMSVGQRAKLTISPDYAYGATGHPGIIPPHATLVFDVELLKLE (SEQ ID NO:20).
78. The polypeptide of embodiment 77, wherein the destabilization domain sequence has one or more of the following amino acid substitutions: F15S, V24A, H25R, E60G, L106P, D100G, M66T, R71G, D100N, E102G, and K105I.
79. The polypeptide of any one of embodiments 1 to 71 , wherein the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to any one of SEQ ID NOs: 19-35 and 66-69 of WO 2018/237323.
80. The polypeptide of any one of embodiments 1 to 79, wherein the MTS is positioned N-terminal to the endonuclease sequence and the destabilizing domain sequence.
81 . The polypeptide of any one of embodiments 1 to 79, wherein the MTS is positioned C-terminal to the endonuclease sequence and the destabilizing domain sequence.
82. The polypeptide of any one of embodiments 1 to 81 , wherein the endonuclease sequence is positioned N-terminal to the destabilization domain sequence.
83. The polypeptide of any one of embodiments 1 to 81 , wherein the endonuclease sequence is positioned C-terminal to the destabilization domain sequence.
84. The polypeptide of any one of embodiments 1 to 79, wherein the MTS is positioned N-terminal to the endonuclease sequence and the endonuclease sequence is positioned N-terminal to the destabilizing domain sequence.
85. A nucleic acid encoding the polypeptide of any one of embodiments 1 to 84.
86. The nucleic acid of embodiment 85, which comprises a promoter operably linked to the nucleotide sequence encoding the polypeptide.
87. The nucleic acid of embodiment 86, wherein the promoter is a SV40 promoter, a CMV promoter or a CAG promoter.
88. The nucleic acid of embodiment 87, wherein the promoter is a SV40 promoter.
89. The nucleic acid of embodiment 88, wherein the SV40 promoter comprises a nucleotide sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to SEQ ID NO:25.
90. The nucleic acid of any one of embodiments 85 to 89, which is a vector.
91 . The nucleic acid of embodiment 90, wherein the vector is a retroviral genome.
92. The nucleic acid of embodiment 91 , wherein the vector is a murine retroviral genome.
93. The nucleic acid of embodiment 90, wherein the vector is a plasmid.
94. The nucleic acid of embodiment 85, which is an mRNA molecule.
95. A particle comprising the nucleic acid of any one of embodiments 85 to 94, optionally wherein the particle is a retroviral particle.
96. A host cell comprising the nucleic acid of any one of embodiments 85 to 94.
97. A method of (a) inducing mitophagy in a cell, and/or (b) increasing mitochondrial turnover in a cell, and/or (c) increasing mitochondrial mass, and/or (d) inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell, the method comprising contacting the cell with (i) the polypeptide of any one of embodiments 1 to 84, the nucleic acid of any one of embodiments 85 to 94, or the particle of embodiment 95 and (ii) a stabilizing agent.
98. The method of embodiment 97, wherein the stabilizing agent is trimethoprim (TMP) when the destabilizing domain sequence is a DHFR destabilization domain sequence.
99. The method of embodiment 97, wherein the stabilizing agent is Shield-1 , rapamycin, or FK506 when the destabilizing domain sequence is a FKBP destabilization domain sequence.
100. The method of any one of embodiments 97 to 99, wherein the contacting the cell with the stabilizing agent comprises culturing the cell in a medium comprising the stabilizing agent.
101 . The method of embodiment 100, which comprises culturing the cell for at least 8 hours in a medium comprising the stabilizing agent.
102. The method of embodiment 100, which comprises culturing the cell for at least 12 hours in a medium comprising the stabilizing agent.
103. The method of embodiment 100, which comprises culturing the cell for at least 1 day in a medium comprising the stabilizing agent.
104. The method of embodiment 100, which comprises culturing the cell for at least 2 days in a medium comprising the stabilizing agent.
105. The method of any one of embodiments 100 to 104, which comprises culturing the cell for up to 5 days in a medium comprising the stabilizing agent.
106. The method of any one of embodiments 100 to 104, which comprises culturing the cell for up to 4 days in a medium comprising the stabilizing agent.
107. The method of any one of embodiments 100 to 104, which comprises culturing the cell for up to 3 days in a medium comprising the stabilizing agent.
108. The method of any one of embodiments 100 to 104, which comprises culturing the cell for up to 2 days in a medium comprising the stabilizing agent.
109. The method of any one of embodiments 97 to 108, which further comprises, subsequent to contacting the cell with the stabilizing agent, removing the stabilizing agent from the cell.
110. The method of embodiment 109, wherein removing the stabilizing agent from the cell comprises culturing the cell in a medium that does not include the stabilizing agent.
111. The method of embodiment 110, which comprises culturing the cell for at 3 hours in a medium that does not include the stabilizing agent.
112. The method of embodiment 110, which comprises culturing the cell for at least 6 hours in a medium that does not include the stabilizing agent.
113. The method of embodiment 110, which comprises culturing the cell for at least 12 hours in a medium that does not include the stabilizing agent.
114. The method of embodiment 110, which comprises culturing the cell for at least 1 day in a medium that does not include the stabilizing agent.
115. The method of embodiment 110, which comprises culturing the cell for at least 2 days in a medium that does not include the stabilizing agent.
116. The method of embodiment 110, which comprises culturing the cell for at least 3 days in a medium that does not include the stabilizing agent.
117. The method of embodiment 110, which comprises culturing the cell for at least 4 days in a medium that does not include the stabilizing agent.
118. The method of any one of embodiments 110 to 117, which comprises culturing the cell for up to 10 days in a medium that does not include the stabilizing agent.
119. The method of any one of embodiments 100 to 117, which comprises culturing the cell for up to 8 days in a medium that does not include the stabilizing agent.
120. The method of any one of embodiments 100 to 117, which comprises culturing the cell for up to 6 days in a medium that does not include the stabilizing agent.
121 . The method of any one of embodiments 100 to 117, which comprises culturing the cell for up to 4 days in a medium that does not include the stabilizing agent.
122. The method of any one of embodiments 97 to 121 , which comprises contacting the cell with the polypeptide of any one of embodiments 1 to 84.
123. The method of embodiment 122, wherein the contacting comprises introducing the polypeptide to the cell via electroporation, injection, or a carrier.
124. The method of any one of embodiments 97 to 121 , which comprises contacting the cell with the nucleic acid of any one of embodiments 85 to 94.
125. The method of embodiment 124, wherein the contacting comprises introducing the nucleic acid to the cell via transfection, electroporation, injection, or a carrier.
126. The method of embodiment 124 or embodiment 125, wherein the polypeptide is transiently expressed in the cell.
127. The method of any one of embodiments 97 to 126, further comprising administering the cell to a subject.
128. The method of any one of embodiments 97 to 127, which induces mitophagy in the cell.
129. The method of any one of embodiments 97 to 128, which increases mitochondrial turnover in the cell.
130. The method of any one of embodiments 97 to 129, which increases mitochondrial mass.
131 . The method of any one of embodiments 97 to 130, which induces double strand breaks in mitochondrial DNA.
132. The method of any one of embodiments 97 to 131 , which induces epigenomic modifications in the cell.
133. A cell obtained or obtainable by the method of any one of embodiments 97 to 132.
134. A cell comprising the polypeptide of any one of embodiments 1 to 84, the nucleic acid of any one of embodiments 85 to 94, or particle of embodiment 95.
135. The cell of embodiment 134, further comprising a stabilizing agent.
136. The cell of embodiment 135, wherein the stabilizing agent is trimethoprim (TMP) when the destabilizing domain sequence is a DHFR destabilization domain sequence.
137. The cell of embodiment 135, wherein the stabilizing agent is Shield-1 , rapamycin, or FK506 when the destabilizing domain sequence is a FKBP destabilization domain sequence.
138. The method of any one of embodiments 97 to 132 or cell of any one of embodiments 133 to 137, wherein the cell is a mammalian cell.
139. The method or cell of embodiment 138, wherein the cell is a human cell.
140. The method or cell of any one of embodiments 138 to 139, wherein the cell is a somatic cell.
141 . The method or cell of any one of embodiments 138 to 140, wherein the cell is a bone marrow cell.
142. The method or cell of any one of embodiments 138 to 141 , wherein the cell is a hematopoietic stem cell (HSC) or a mesenchymal stem cell (MSC).
143. The method or cell of any one of embodiments 138 to 140, wherein the cell is an immune cell.
144. The method or cell of embodiment 143, wherein the cell is a T cell, a phagocyte, a microglial cell, or a macrophage.
145. The method or cell of embodiment 144, wherein the cell is CD4+ T cell.
146. The method or cell of embodiment 144 or embodiment 145, wherein the cell is CD8+ T cell.
147. The method or cell of any one of embodiments 138 to 146, wherein the cell is a primary cell.
148. The method or cell of any one of embodiments 138 to 146, wherein the cell is a progeny of a primary cell.
149. The method or cell of any one of embodiments 138 to 148, wherein the cell has dysfunctional mitochondria.
150. The method or cell of any one of embodiments 138 to 149, wherein the cell is from a subject having an age-related disease.
151 . The method or cell of embodiment 150, wherein the age-related disease is an autoimmune disease, a metabolic disease, a genetic disease, cancer, a neurodegenerative disease, or immunosenescence.
152. The method or cell of any one of embodiments 138 to 151 , wherein the cell is from a subject having a mitochondrial disease or disorder.
153. The method or cell of embodiment 152, wherein the mitochondrial disease or disorder is caused by mitochondrial DNA abnormalities, nuclear DNA abnormalities, or both.
154. The method or cell of embodiment 152 or embodiment 153, wherein the mitochondrial disease or disorder is chronic progressive external ophthalmoplegia (CPEO), Pearson syndrome, Kearns-Sayre syndrome (KSS), diabetes and deafness (DAD),
mitochondrial diabetes, Leber hereditary optic neuropathy (LHON), LHON-plus, neuropathy, ataxia, retinitis pigmentosa syndrome (NARP), maternally inherited Leigh syndrome (MILS), mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), myoclonic epilepsy and ragged-red fiber disease (MERRF), familial bilateral striatal necrosis/striatonigral degeneration (FBSN), Luft disease, aminoglycoside-induced Deafness (AID), or multiple deletions of mitochondrial DNA syndrome.
155. The method or cell of embodiment 152 or embodiment 153, wherein the mitochondrial disease or disorder is Mitochondrial DNA depletion syndrome-4A, mitochondrial recessive ataxia syndrome (MIRAS), mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), mitochondrial DNA depletion syndrome (MTDPS), DNA polymerase gamma (POLG)- related disorders, sensory ataxia neuropathy dysarthria ophthalmoplegia (SANDO), leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), co-enzyme Q10 deficiency, Leigh syndrome, mitochondrial complex abnormalities, fumarase deficiency, a- ketoglutarate dehydrogenase complex (KGDHC) deficiency, succinyl-CoA ligase deficiency, pyruvate dehydrogenase complex deficiency (PDHC), pyruvate carboxylase deficiency (PCD), carnitine palmitoyltransferase I (CPT I) deficiency, carnitine palmitoyltransferase II (CPT IT) deficiency, carnitine-acyl-carnitine (CACT) deficiency, autosomal dominant-/ autosomal recessive- progressive external ophthalmoplegia (ad-Zar- PEO), infantile onset spinal cerebellar atrophy (IOSCA), mitochondrial myopathy (MM) spinal muscular atrophy (SMA), growth retardation, aminoaciduria, cholestasis, iron overload, early death (GRACILE), or Charcot-Marie-Tooth disease type 2 A (CMT2A).
156. The method or cell of any one of embodiments 138 to 155, wherein the cell is from a subject having a neurodegenerative disease.
157. The method or cell of embodiment 156, wherein the neurodegenerative disease is amyotrophic lateral sclerosis (ALS), Huntington’s disease, Alzheimer's disease, Parkinson's disease, Friedreich' s ataxia, Charcot Marie Tooth disease or leukodystrophy.
158. The method or cell of embodiment 156, wherein the neurodegenerative disease is amyotrophic lateral sclerosis (ALS).
159. The method or cell of embodiment 156, wherein the neurodegenerative disease is Huntington’s disease.
160. The method or cell of embodiment 156, wherein the neurodegenerative disease is Alzheimer's disease, optionally wherein the cell is from a subject having an APOE4 allele, for example an E3/E4 or E4/E4 genotype.
161 . The method or cell of embodiment 156, wherein the neurodegenerative disease is Parkinson's disease.
162. The method or cell of embodiment 156, wherein the neurodegenerative disease is Friedreich' s ataxia. Charcot Marie Tooth disease.
163. The method or cell of embodiment 156, wherein the neurodegenerative disease is Leukodystrophy.
164. The method or cell of any one of embodiments 138 to 163, wherein the cell is from a subject having a retinal disease.
165. The method or cell of embodiment 164, wherein the retinal disease is age- related macular degeneration, macular edema or glaucoma.
166. The method or cell of any one of embodiments 138 to 165, wherein the cell is from a subject having diabetes.
167. The method or cell of any one of embodiments 138 to 166, wherein the cell is from a subject having a hearing disorder.
168. The method or cell of any one of embodiments 138 to 167, wherein the cell is from a subject having a genetic disease.
169. The method or cell of embodiment 168, wherein the genetic disease is Hutchinson-Gilford Progeria Syndrome, Werner Syndrome, or Huntington's disease.
170. The method or cell of any one of embodiments 138 to 169, wherein the cell is from a subject having heart failure.
171 . The method or cell of any one of embodiments 138 to 170, wherein the cell is from a subject having immunodeficiency.
172. The method or cell of any one of embodiments 138 to 171 , wherein the cell is from a subject having cancer.
173. The method or cell of any one of embodiments 138 to 172, wherein the cell is from a subject having an infectious disease.
174. The method or cell of any one of embodiments 138 to 148, wherein the cell is from a healthy donor.
175. The method or cell of any one of embodiments 138 to 174, which is an ex vivo cell.
176. The method of embodiment 175, further comprising administering the cell to a subject, optionally wherein the subject is the same subject from which the cell originated.
177. A method of treating a subject having an age-related disease, mitochondrial disease or disorder, neurodegenerative disease, retinal disease, diabetes, hearing disorder, genetic disease, heart failure, immunodeficiency, cancer, or infectious disease, the method comprising administering a therapeutically effective amount of cells according to any one of embodiments 133 to 175 to the subject.
178. The method of embodiment 177, wherein the subject has an age-related disease.
179. The method of embodiment 178, wherein the age-related disease is an autoimmune disease, a metabolic disease, a genetic disease, cancer, a neurodegenerative disease, or immunosenescence.
180. The method of embodiment 177, wherein the subject has a mitochondrial disease or disorder.
181 . The method of embodiment 180, wherein the mitochondrial disease or disorder is caused by mitochondrial DNA abnormalities, nuclear DNA abnormalities, or both.
182. The method of embodiment 180 or embodiment 181 , wherein the mitochondrial disease or disorder is chronic progressive external ophthalmoplegia (CPEO), Pearson syndrome, Kearns-Sayre syndrome (KSS), diabetes and deafness (DAD), mitochondrial diabetes, Leber hereditary optic neuropathy (LHON), LHON-plus, neuropathy, ataxia, and retinitis pigmentosa syndrome (NARP), maternally inherited Leigh syndrome (MILS), mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), myoclonic epilepsy and ragged-red fiber disease (MERRF), familial bilateral striatal necrosis/striatonigral degeneration (FBSN), Luft disease, aminoglycoside-induced Deafness (AID), and multiple deletions of mitochondrial DNA syndrome.
183. The method of embodiment 180 or embodiment 181 , wherein the mitochondrial disease or disorder is Mitochondrial DNA depletion syndrome-4A, mitochondrial recessive ataxia syndrome (MIRAS), mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), mitochondrial DNA depletion syndrome (MTDPS), DNA polymerase gamma (POLG)-related disorders, sensory ataxia neuropathy dysarthria ophthalmoplegia (SANDO),
leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), co-enzyme Q10 deficiency, Leigh syndrome, mitochondrial complex abnormalities, fumarase deficiency, a- ketoglutarate dehydrogenase complex (KGDHC) deficiency, succinyl-CoA ligase deficiency, pyruvate dehydrogenase complex deficiency (PDHC), pyruvate carboxylase deficiency (PCD), carnitine palmitoyltransferase I (CPT I) deficiency, carnitine palmitoyltransferase II (CPT IT) deficiency, carnitine-acyl-carnitine (CACT) deficiency, autosomal dominant-/ autosomal recessive- progressive external ophthalmoplegia (ad-Zar- PEO), infantile onset spinal cerebellar atrophy (IOSCA), mitochondrial myopathy (MM) spinal muscular atrophy (SMA), growth retardation, aminoaciduria, cholestasis, iron overload, early death (GRACILE), or Charcot-Marie-Tooth disease type 2 A (CMT2A).
184. The method of embodiment 177, wherein the subject has a neurodegenerative disease.
185. The method of embodiment 184, wherein the neurodegenerative disease is amyotrophic lateral sclerosis (ALS), Huntington’s disease, Alzheimer's disease, Parkinson's disease, Friedreich' s ataxia, Charcot Marie Tooth disease or leukodystrophy.
186. The method of embodiment 177, wherein the subject has a retinal disease.
187. The method of embodiment 186, wherein the retinal disease is age-related macular degeneration, macular edema or glaucoma.
188. The method of embodiment 177, wherein the subject has diabetes.
189. The method of embodiment 177, wherein the subject has a hearing disorder.
190. The method of embodiment 177, wherein the subject has a genetic disease.
191 . The method of embodiment 190, wherein the genetic disease is Hutchinson-
Gilford Progeria Syndrome, Werner Syndrome, or Huntington's disease.
192. The method of embodiment 177, wherein the subject has heart failure.
193. The method of embodiment 177, wherein the subject has immunodeficiency.
194. The method of embodiment 177, wherein the subject has cancer.
195. The method of embodiment 177, wherein the subject has an infectious disease.
196. A pharmaceutical composition comprising the polypeptide of any one of embodiments 1 to 84, the nucleic acid of any one of embodiments 85 to 94, the particle of
embodiment 95, or the cell of any one of embodiments 133 to 175 and a pharmaceutically acceptable excipient.
197. A kit comprising (a) the polypeptide of any one of embodiments 1 to 84, the nucleic acid of any one of embodiments 85 to 94, or the particle of embodiment 95 and (b) a stabilizing agent.
198. The kit of embodiment 197, wherein the stabilizing agent is trimethoprim (TMP) when the destabilizing domain sequence is a DHFR destabilization domain sequence.
199. The kit of embodiment 197, wherein the stabilizing agent is Shield-1 , rapamycin, or FK506 when the destabilizing domain sequence is a FKBP destabilization domain sequence.
200. The kit of embodiment 197, wherein the stabilizing agent is sildenafil, vardenafil, tadalafil, avanafil, lodenafil, mirodenafil, udenafil, benzamidenafil, dasantafil, or beminafil when the destabilizing domain sequence is a PDE5 destabilization domain sequence.
7. CITATION OF REFERENCES
[0158] All publications, patents, patent applications and other documents cited in this application are hereby incorporated by reference in their entireties for all purposes to the same extent as if each individual publication, patent, patent application or other document were individually indicated to be incorporated by reference for all purposes. In the event that there is an inconsistency between the teachings of one or more of the references incorporated herein and the present disclosure, the teachings of the present specification are intended
Claims
1 . A polypeptide comprising:
(a) a mitochondrial targeting sequence (MTS);
(b) an endonuclease sequence; and
(c) a destabilizing domain sequence.
2. The polypeptide of claim 1 , wherein the MTS comprises a human MTS.
3. The polypeptide of claim 1 , wherein the MTS comprises a non-human MTS.
4. The polypeptide of any one of claims 1 to 3, wherein the MTS comprises an MTS of a mitochondrial protein.
5. The polypeptide of any one of claims 1 to 4, wherein the MTS comprises an MTS of a TCA cycle-related enzyme, a chaperone protein, a mitochondrial genome replication protein, a protease, an mRNA processing protein, a mitochondrial RNA degradation protein, a deoxy nucleotide triphosphate synthesis-related protein, a mitoribosomal protein, a phospholipid metabolism-related protein, a protein involved in metabolism of toxic compounds, a disulfide relay system-related protein, an iron-sulfur protein assembly protein, a tRNA modification protein, an aminoacyl-tRNA synthetase, a release factor, or an elongation factor.
6. The polypeptide of any one of claims 1 to 5, wherein the MTS comprises an MTS of a cytochrome c oxidase subunit.
7. The polypeptide of claim 6, wherein the MTS comprises an MTS of a cytochrome c oxidase subunit VIII (COX8), cytochrome c oxidase subunit X (COX10), cytochrome c oxidase subunit IV (COX4).
8. The polypeptide of any one of claims 1 to 5, wherein the MTS comprises:
(a) an MTS of a frataxin (FXN) protein;
(b) an MTS of a TCA cycle-related enzyme, optionally which is Pyruvate dehydrogenase, Citrate synthase, Aconitase, Isocitrate dehydrogenase, a- ketoglutarate dehydrogenase, Succinyl-CoA synthetase, Succinic dehydrogenase, Fumarase, Malate dehydrogenase, or Pyruvate carboxylase;
(c) an MTS of a chaperone protein, optionally which is mtHSPIO, mtHSP60, mtHSP70, or mtHSP90;
(d) an MTS of a mitochondrial genome replication protein, optionally which is TFAM, Twinkle, PolG, TFB2M, TEFM, or MTERF1 ;
(e) an MTS of a protease, optionally which is MPP, CLPXP, LON ATPase, or PreP;
(f) an MTS of an mRNA processing protein, optionally which is LRPPRC, TACO1 , ELAC2, PNPT1 , HSD17B10, MTPAP, or PTCDI ;
(g) an MTS of a mitochondrial RNA degradation protein, optionally which is PNPasse, REX02, or SUV3;
(h) an MTS of a deoxynucleotide triphosphate synthesis-related protein, optionally which is DGUOK, TK2, TYMP, MGME1 , SUCLG1 , SUCLA2, RNASEH1 , or C10orf2;
(i) an MTS of a mitoribosomal protein, optionally which is MRPS16, MRPS22, MRPL3, MRP12, or MRPL44;
(j) an MTS of a phospholipid metabolism-related protein, optionally which is AGK, SERAC1 , or TAZ;
(k) an MTS of a protein involved in metabolism of toxic compounds, optionally which is HIBCH, ECHS1 , ETHE1 or MPV17;
(l) an MTS of a disulfide relay system-related protein, optionally which is GFER;
(m) an MTS of an iron-sulfur protein assembly protein, optionally which is ISCU, BOLA3, NFU1 , or IBA57;
(n) an MTS of a tRNA modification protein, optionally which is MTO1 , GTP3BP, TRMU, PUS1 , MTFMT, TRIT1 , TRNT1 or TRMT5;
(o) an MTS of an aminoacyl-tRNA synthetase, optionally which is AARS2, DARS2, EARS2, RARS2, YARS2, FARS2, HARS2, LARS2, VARS2, TARS2, IARS2, CARS2, PARS2, NARS2, KARS, GARS, SARS2 or MARS2;
(p) an MTS of a release factor, optionally which is a C12orf65; or
(q) an MTS of an elongation factor, optionally which is TUFM, TSFM, or GFM1 .
9. The polypeptide of claim 1 , wherein the MTS comprises a sequence that is at least 80% identical to any one of SEQ ID NOs. 1-15.
10. The polypeptide of claim 9, wherein the MTS comprises a sequence that is 100% identical to SEQ ID NO:1.
11 . The polypeptide of claim 9, wherein the MTS comprises a sequence that is 100% identical to SEQ ID NO:2.
12. The polypeptide of any one of claims 1 to 11 , wherein the endonuclease is a restriction endonuclease, an RNA-guided endonuclease (e.g., Cas9 or Cas12), a zinc finger nuclease, or a transcription activator-like effector nuclease (TALEN).
13. The polypeptide of claim 12, wherein the endonuclease is a restriction endonuclease.
14. The polypeptide of claim 13, wherein the restriction endonuclease is an XbaIR, EcoRI, Smal, Aflll, BamHI, Bell, Haelll, Hindll, Hindlll, Ndel, Pvull, Pstl, or Spel endonuclease.
15. The polypeptide of claim 14, wherein the restriction endonuclease is an XbaIR endonuclease.
16. The polypeptide of any one of claims 1 to 13, wherein the endonuclease comprises a sequence that is at least 80% identical to SEQ ID NO:16.
17. The polypeptide of claim 16, wherein the endonuclease sequence comprises a sequence that is 100% identical to SEQ ID NO:16.
18. The polypeptide of any one of claims 1 to 17, wherein the destabilizing domain sequence is a DHFR, FKBP, or PDE5 destabilization domain sequence.
19. The polypeptide of claim 18, wherein the destabilization domain sequence is a DHFR destabilization domain sequence.
20. The polypeptide of claim 19, wherein the destabilization domain sequence is an E. coli DHFR (ecDHFR) destabilization domain sequence.
21 . The polypeptide of any one of claims 1 to 20, wherein the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to SEQ ID NO:18.
22. The polypeptide of any one of claims 1 to 18, wherein the destabilization domain sequence comprises a sequence that is at least 80%, at least 85%, at least 90%, at least 95%, or 100% identical to SEQ ID NQ:20.
23. The polypeptide of any one of claims 1 to 22, wherein the MTS is positioned N- terminal to the endonuclease sequence and the destabilizing domain sequence.
24. The polypeptide of any one of claims 1 to 22, wherein the MTS is positioned C- terminal to the endonuclease sequence and the destabilizing domain sequence.
25. The polypeptide of any one of claims 1 to 24, wherein the endonuclease sequence is positioned N-terminal to the destabilization domain sequence.
26. The polypeptide of any one of claims 1 to 24, wherein the endonuclease sequence is positioned C-terminal to the destabilization domain sequence.
27. The polypeptide of any one of claims 1 to 22, wherein the MTS is positioned N- terminal to the endonuclease sequence and the endonuclease sequence is positioned N- terminal to the destabilizing domain sequence.
28. A nucleic acid encoding the polypeptide of any one of claims 1 to 27.
29. A particle comprising the nucleic acid of claim 28, optionally wherein the particle is a retroviral particle.
30. A host cell comprising the nucleic acid of claim 28.
31 . A method of (a) inducing mitophagy in a cell, and/or (b) increasing mitochondrial turnover in a cell, and/or (c) increasing mitochondrial mass, and/or (d) inducing double strand breaks in mitochondrial DNA and/or (e) inducing epigenomic modifications in a cell, the method comprising contacting the cell with (i) the polypeptide of any one of claims 1 to 27, the nucleic acid of claim 28, or the particle of claim 29 and (ii) a stabilizing agent.
32. The method of claim 31 , wherein the stabilizing agent is trimethoprim (TMP) when the destabilizing domain sequence is a DHFR destabilization domain sequence.
33. The method of claim 31 , wherein the stabilizing agent is Shield-1 , rapamycin, or FK506 when the destabilizing domain sequence is a FKBP destabilization domain sequence.
34. The method of any one of claims 31 to 33, which further comprises, subsequent to contacting the cell with the stabilizing agent, removing the stabilizing agent from the cell.
35. A cell obtained or obtainable by the method of any one of claims 31 to 34.
36. A cell comprising the polypeptide of any one of claims 1 to 27, the nucleic acid of claim 28, or particle of claim 29.
37. The method of any one of claims 31 to 34 or cell of claim 35 or claim 36, wherein the cell is a mammalian cell.
38. The method or cell of claim 37, wherein the cell is a human cell.
39. The method or cell of claim 37 or claim 38, wherein the cell is a somatic cell, bone marrow cell, hematopoietic stem cell (HSC) or a mesenchymal stem cell (MSC), or an immune cell.
40. The method or cell of any one of claims 37 to 39, wherein the cell is from a subject having an age-related disease.
41 . The method or cell of claim 40, wherein the age-related disease is an autoimmune disease, a metabolic disease, a genetic disease, cancer, a neurodegenerative disease, or immunosenescence.
42. The method or cell of any one of claims 37 to 41 , wherein the cell is from a subject having a mitochondrial disease or disorder.
43. The method or cell of claim 42, wherein the mitochondrial disease or disorder is chronic progressive external ophthalmoplegia (CPEO), Pearson syndrome, Kearns-Sayre syndrome (KSS), diabetes and deafness (DAD), mitochondrial diabetes, Leber hereditary optic neuropathy (LHON), LHON-plus, neuropathy, ataxia, retinitis pigmentosa syndrome (NARP), maternally inherited Leigh syndrome (MILS), mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), myoclonic epilepsy and ragged-red fiber disease (MERRF), familial bilateral striatal necrosis/striatonigral degeneration (FBSN), Luft disease, aminoglycoside-induced Deafness (AID), or multiple deletions of mitochondrial DNA syndrome.
44. The method or cell of claim 42, wherein the mitochondrial disease or disorder is Mitochondrial DNA depletion syndrome-4A, mitochondrial recessive ataxia syndrome (MIRAS), mitochondrial neurogastrointestinal encephalomyopathy (MNGIE), mitochondrial DNA depletion syndrome (MTDPS), DNA polymerase gamma (POLG)-related disorders, sensory ataxia neuropathy dysarthria ophthalmoplegia (SANDO), leukoencephalopathy with brainstem and spinal cord involvement and lactate elevation (LBSL), co-enzyme Q10 deficiency, Leigh syndrome, mitochondrial complex abnormalities, fumarase deficiency, a- ketoglutarate dehydrogenase complex (KGDHC) deficiency, succinyl-CoA ligase deficiency, pyruvate dehydrogenase complex deficiency (PDHC), pyruvate carboxylase deficiency (PCD), carnitine palmitoyltransferase I (CPT I) deficiency, carnitine palmitoyltransferase II (CPT IT) deficiency, carnitine-acyl-carnitine (CACT) deficiency, autosomal dominant-/ autosomal recessive-
progressive external ophthalmoplegia (ad-/ar-PEO), infantile onset spinal cerebellar atrophy (IOSCA), mitochondrial myopathy (MM) spinal muscular atrophy (SMA), growth retardation, aminoaciduria, cholestasis, iron overload, early death (GRACILE), or Charcot-Marie-Tooth disease type 2 A (CMT2A).
45. The method or cell of any one of claims 37 to 44, wherein the cell is from a subject having a neurodegenerative disease.
46. The method or cell of claim 45, wherein the neurodegenerative disease is amyotrophic lateral sclerosis (ALS), Huntington’s disease, Alzheimer's disease, Parkinson's disease, Friedreich' s ataxia, Charcot Marie Tooth disease or leukodystrophy.
47. The method or cell of any one of claims 37 to 46, wherein the cell is from a subject having a retinal disease, diabetes, a hearing disorder, a genetic diseae, heart failure, immunodeficiency, cancer, or infectious disease.
48. The method or cell of any one of claims 37 to 47, wherein the cell is an ex vivo cell.
49. The method of claim 48, further comprising administering the cell to a subject, optionally wherein the subject is the same subject from which the cell originated.
50. A method of treating a subject having an age-related disease, mitochondrial disease or disorder, neurodegenerative disease, retinal disease, diabetes, hearing disorder, genetic disease, heart failure, immunodeficiency, cancer, or infectious disease, the method comprising administering a therapeutically effective amount of cells according to any one of claims 35 to 48 to the subject.
51 . A pharmaceutical composition comprising the polypeptide of any one of claims 1 to 27, the nucleic acid of claim 28, the particle of claim 29, or the cell of any one of claims 35 to 48 and a pharmaceutically acceptable excipient.
52. A kit comprising (a) the polypeptide of any one of claims 1 to 27, the nucleic acid of claim 28, or the particle of claim 29 and (b) a stabilizing agent.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202263426424P | 2022-11-18 | 2022-11-18 | |
US63/426,424 | 2022-11-18 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024105633A1 true WO2024105633A1 (en) | 2024-05-23 |
Family
ID=88965224
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/IB2023/061649 WO2024105633A1 (en) | 2022-11-18 | 2023-11-17 | Compositions for mitophagy induction and uses thereof |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024105633A1 (en) |
Citations (21)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1993003769A1 (en) | 1991-08-20 | 1993-03-04 | THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES | Adenovirus mediated transfer of genes to the gastrointestinal tract |
WO1993009239A1 (en) | 1991-11-08 | 1993-05-13 | Research Corporation Technologies, Inc. | Adeno-associated virus-2 basal vectors |
WO1993019191A1 (en) | 1992-03-16 | 1993-09-30 | Centre National De La Recherche Scientifique | Defective recombinant adenoviruses expressing cytokines for use in antitumoral treatment |
WO1994012649A2 (en) | 1992-12-03 | 1994-06-09 | Genzyme Corporation | Gene therapy for cystic fibrosis |
WO1994028938A1 (en) | 1993-06-07 | 1994-12-22 | The Regents Of The University Of Michigan | Adenovirus vectors for gene therapy sponsorship |
WO1995000655A1 (en) | 1993-06-24 | 1995-01-05 | Mc Master University | Adenovirus vectors for gene therapy |
WO1995011984A2 (en) | 1993-10-25 | 1995-05-04 | Canji, Inc. | Recombinant adenoviral vector and methods of use |
WO2001025255A2 (en) | 1999-10-07 | 2001-04-12 | Baylor College Of Medicine | Novel zinc finger protein |
WO2003066828A2 (en) | 2002-02-07 | 2003-08-14 | The Scripps Research Institute | Zinc finger libraries |
WO2013163628A2 (en) | 2012-04-27 | 2013-10-31 | Duke University | Genetic correction of mutated genes |
WO2014093661A2 (en) | 2012-12-12 | 2014-06-19 | The Broad Institute, Inc. | Crispr-cas systems and methods for altering expression of gene products |
WO2014134412A1 (en) | 2013-03-01 | 2014-09-04 | Regents Of The University Of Minnesota | Talen-based gene correction |
WO2015013583A2 (en) | 2013-07-26 | 2015-01-29 | President And Fellows Of Harvard College | Genome engineering |
WO2016148994A1 (en) * | 2015-03-13 | 2016-09-22 | The Jackson Laboratory | A three-component crispr/cas complex system and uses thereof |
US9487787B2 (en) | 2007-02-09 | 2016-11-08 | The Board Of Trustees Of The Leland Stanford Junior University | Method for regulating protein function in cells using synthetic small molecules |
US20180002682A1 (en) * | 2015-01-12 | 2018-01-04 | The Regents Of The University Of California | Heterodimeric cas9 and methods of use thereof |
WO2018237323A1 (en) | 2017-06-23 | 2018-12-27 | The Board Of Trustees Of The Leland Stanford Junior University | Pde5a destabilizing domains |
WO2019060297A1 (en) * | 2017-09-19 | 2019-03-28 | Unity Biotechnology, Inc. | Mitochondrial rejuvenation as a treatment for adverse age-related conditions and neurodegeneration |
WO2019233990A1 (en) | 2018-06-04 | 2019-12-12 | University Of Copenhagen | Mutant cpf1 endonucleases |
US20200054682A1 (en) * | 2018-08-14 | 2020-02-20 | Imel Biotherapeutics, Inc. | Methods and compositions for treating mitochondrial disease or disorders and heteroplasmy |
US11001863B2 (en) | 2012-05-25 | 2021-05-11 | The Regents Of The University Of California | Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription |
-
2023
- 2023-11-17 WO PCT/IB2023/061649 patent/WO2024105633A1/en unknown
Patent Citations (21)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1993003769A1 (en) | 1991-08-20 | 1993-03-04 | THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES | Adenovirus mediated transfer of genes to the gastrointestinal tract |
WO1993009239A1 (en) | 1991-11-08 | 1993-05-13 | Research Corporation Technologies, Inc. | Adeno-associated virus-2 basal vectors |
WO1993019191A1 (en) | 1992-03-16 | 1993-09-30 | Centre National De La Recherche Scientifique | Defective recombinant adenoviruses expressing cytokines for use in antitumoral treatment |
WO1994012649A2 (en) | 1992-12-03 | 1994-06-09 | Genzyme Corporation | Gene therapy for cystic fibrosis |
WO1994028938A1 (en) | 1993-06-07 | 1994-12-22 | The Regents Of The University Of Michigan | Adenovirus vectors for gene therapy sponsorship |
WO1995000655A1 (en) | 1993-06-24 | 1995-01-05 | Mc Master University | Adenovirus vectors for gene therapy |
WO1995011984A2 (en) | 1993-10-25 | 1995-05-04 | Canji, Inc. | Recombinant adenoviral vector and methods of use |
WO2001025255A2 (en) | 1999-10-07 | 2001-04-12 | Baylor College Of Medicine | Novel zinc finger protein |
WO2003066828A2 (en) | 2002-02-07 | 2003-08-14 | The Scripps Research Institute | Zinc finger libraries |
US9487787B2 (en) | 2007-02-09 | 2016-11-08 | The Board Of Trustees Of The Leland Stanford Junior University | Method for regulating protein function in cells using synthetic small molecules |
WO2013163628A2 (en) | 2012-04-27 | 2013-10-31 | Duke University | Genetic correction of mutated genes |
US11001863B2 (en) | 2012-05-25 | 2021-05-11 | The Regents Of The University Of California | Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription |
WO2014093661A2 (en) | 2012-12-12 | 2014-06-19 | The Broad Institute, Inc. | Crispr-cas systems and methods for altering expression of gene products |
WO2014134412A1 (en) | 2013-03-01 | 2014-09-04 | Regents Of The University Of Minnesota | Talen-based gene correction |
WO2015013583A2 (en) | 2013-07-26 | 2015-01-29 | President And Fellows Of Harvard College | Genome engineering |
US20180002682A1 (en) * | 2015-01-12 | 2018-01-04 | The Regents Of The University Of California | Heterodimeric cas9 and methods of use thereof |
WO2016148994A1 (en) * | 2015-03-13 | 2016-09-22 | The Jackson Laboratory | A three-component crispr/cas complex system and uses thereof |
WO2018237323A1 (en) | 2017-06-23 | 2018-12-27 | The Board Of Trustees Of The Leland Stanford Junior University | Pde5a destabilizing domains |
WO2019060297A1 (en) * | 2017-09-19 | 2019-03-28 | Unity Biotechnology, Inc. | Mitochondrial rejuvenation as a treatment for adverse age-related conditions and neurodegeneration |
WO2019233990A1 (en) | 2018-06-04 | 2019-12-12 | University Of Copenhagen | Mutant cpf1 endonucleases |
US20200054682A1 (en) * | 2018-08-14 | 2020-02-20 | Imel Biotherapeutics, Inc. | Methods and compositions for treating mitochondrial disease or disorders and heteroplasmy |
Non-Patent Citations (31)
Title |
---|
"Mitochondrial Function", vol. 547, 1 January 2014, ELSEVIER, ISBN: 978-0-12-801415-8, article BACMAN SANDRA R. ET AL: "The Use of Mitochondria-Targeted Endonucleases to Manipulate mtDNA", pages: 373 - 397, XP055908878, DOI: 10.1016/B978-0-12-801415-8.00018-7 * |
"UniProt", Database accession no. AOA4R7BM34 |
ALI ET AL., HUM GENE THER, vol. 9, 1998, pages 81 - 86 |
ALI ET AL., HUM MOL GENET, vol. 5, 1996, pages 591 - 594 |
BANASZYNSKI ET AL., CELL, vol. 126, no. 5, 2006, pages 995 - 1104 |
BAYONA-BAFALUY MARIA PILAR ET AL: "RAPID DIRECTIONAL SHIFT OF MITOCHONDRIAL DNA HETEROPLASMY IN ANIMAL TISSUES BY A MITOCHONDRIALLY TARGETED RESTRICTION ENDONUCLEASE", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, vol. 102, no. 40, 1 October 2005 (2005-10-01), pages 14392 - 14397, XP001249333, ISSN: 0027-8424, DOI: 10.1073/PNAS.0502896102 * |
BENDTSEN ET AL., J. MOL. BIOL., vol. 340, 2004, pages 783 - 795 |
BENNETT ET AL., INVEST OPTHALMOL VIS SCI, vol. 38, 1997, pages 2857 - 2863 |
BORRAS ET AL., GENE THER, vol. 6, 1999, pages 515 - 524 |
CLAROS, COMPUT APL BIOSCI., vol. 11, no. 4, 1995, pages 441 - 7 |
FLANNERY ET AL., PNAS, vol. 94, 1997, pages 10319 - 6921 |
FUKASAWA ET AL., MOL CELL PROTEOMICS, vol. 14, no. 4, 2015, pages 1113 - 1126 |
HARDMAN ET AL.: "Goodman and Gilman's The Pharmacological Basis of Therapeutics", 2001, MCGRAW-HILL |
HERTZOG SANTOS, FREE RADIC BIOL MED., vol. 170, 2021, pages 69 - 69 |
IWAMOTO ET AL., CHEM BIOL., vol. 17, no. 9, 2010, pages 981 - 8 |
JOMARY ET AL., GENE THER, vol. 4, 1997, pages 683 - 690 |
LI ET AL., INVEST OPTHALMOL VIS SCI, vol. 35, 1994, pages 2543 - 2549 |
LIDAVIDSON, PNAS, vol. 92, 1995, pages 7700 - 7704 |
LIU ET AL., INT. J. PARASITOL., vol. 44, no. 10, 2014, pages 729 - 735 |
MARI IWAMOTO ET AL: "A General Chemical Method to Regulate Protein Stability in the Mammalian Central Nervous System", CHEMISTRY & BIOLOGY, vol. 17, no. 9, 1 September 2010 (2010-09-01), GB, pages 981 - 988, XP055552908, ISSN: 1074-5521, DOI: 10.1016/j.chembiol.2010.07.009 * |
MORAES CARLOS T ET AL: "Current strategies towards therapeutic manipulation of mtDNA heteroplasmy", FRONTIERS IN BIOSCIENCE, vol. 22, no. 6, 1 January 2017 (2017-01-01), US, pages 991 - 1010, XP055871512, ISSN: 1093-9946, DOI: 10.2741/4529 * |
PENG HUI ET AL: "Non-antibiotic Small-Molecule Regulation of DHFR-Based Destabilizing Domains In Vivo", MOLECULAR THERAPY- METHODS & CLINICAL DEVELOPMENT, vol. 15, 1 December 2019 (2019-12-01), GB, pages 27 - 39, XP055829622, ISSN: 2329-0501, DOI: 10.1016/j.omtm.2019.08.002 * |
PENG HUI ET AL: "Utility of the DHFR-based destabilizing domain across mouse models of retinal degeneration and aging", ISCIENCE, vol. 25, no. 5, 6 April 2022 (2022-04-06), US, XP093126529, ISSN: 2589-0042, DOI: 10.1016/j.isci.2022.104206 * |
PICKRELL ALICIA M. ET AL: "Striatal Dysfunctions Associated with Mitochondrial DNA Damage in Dopaminergic Neurons in a Mouse Model of Parkinson's Disease", THE JOURNAL OF NEUROSCIENCE, vol. 31, no. 48, 30 November 2011 (2011-11-30), US, pages 17649 - 17658, XP093126382, ISSN: 0270-6474, DOI: 10.1523/JNEUROSCI.4871-11.2011 * |
ROLLING ET AL., HUM GENE THER, vol. 10, 1999, pages 641 - 648 |
SAKAMOTO ET AL., H GENE THER, vol. 5, 1999, pages 1088 - 1097 |
SCHON E A ET AL: "Therapeutic prospects for mitochondrial disease", TRENDS IN MOLECULAR MEDICINE, ELSEVIER CURRENT TRENDS, GB, vol. 16, no. 6, 1 June 2010 (2010-06-01), pages 268 - 276, XP027085406, ISSN: 1471-4914, [retrieved on 20100601] * |
TAKAHASHI ET AL., J VIROL, vol. 73, 1999, pages 7812 - 7816 |
TEUFEL ET AL., NAT BIOTECHNOL., 2022 |
WANG SHOULIANG ET AL: "The mitophagy pathway and its implications in human diseases", SIGNAL TRANSDUCTION AND TARGETED THERAPY, vol. 8, no. 1, 30 November 2011 (2011-11-30), XP093126385, ISSN: 2059-3635, DOI: 10.1038/s41392-023-01503-7 * |
WEINERKOTKOSKIE: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS, AND WILKINS |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11702671B2 (en) | Importation of mitochondrial protein by an enhanced allotopic approach | |
US20200282025A1 (en) | Methods and compositions for rna-guided treatment of hiv infection | |
US12049650B2 (en) | Destabilising domains for conditionally stabilising a protein | |
KR20200083550A (en) | How to rescue a stop codon through gene redirection by ACE-tRNA | |
WO2016149484A2 (en) | Compositions and methods for specific reactivation of hiv latent reservoir | |
US20170166875A1 (en) | Methods relating to nucleic acid sequence editing | |
KR20180092989A (en) | Transposon systems, kits containing them, and uses thereof | |
US20130084327A1 (en) | Polynucleotide encoding a trim-cyp polypeptide, compositions thereof, and methods of using same | |
EP4225916A1 (en) | Functional nucleic acid molecules | |
CN116685329A (en) | Nucleic acid constructs and their use for the treatment of spinal muscular atrophy | |
CA2859920A1 (en) | Variants of yeast ndi1 gene, and uses thereof in the treatment of disease associated with mitochondrial dysfunction | |
CN114381465B (en) | Optimized CYP4V2 gene and application thereof | |
AU2022200678B2 (en) | Nucleic acid molecules containing spacers and methods of use thereof | |
WO2024105633A1 (en) | Compositions for mitophagy induction and uses thereof | |
US9296994B2 (en) | Archease as RNA ligaes complex member | |
Zhang et al. | Cellular protein TTRAP interacts with HIV-1 integrase to facilitate viral integration | |
KR20230037586A (en) | Methods for treating Alzheimer's disease targeting the MAPT gene |
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: 23813069 Country of ref document: EP Kind code of ref document: A1 |