Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 186(2): 287-304.e26, 2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36610399

RESUMO

Whether and how certain transposable elements with viral origins, such as endogenous retroviruses (ERVs) dormant in our genomes, can become awakened and contribute to the aging process is largely unknown. In human senescent cells, we found that HERVK (HML-2), the most recently integrated human ERVs, are unlocked to transcribe viral genes and produce retrovirus-like particles (RVLPs). These HERVK RVLPs constitute a transmissible message to elicit senescence phenotypes in young cells, which can be blocked by neutralizing antibodies. The activation of ERVs was also observed in organs of aged primates and mice as well as in human tissues and serum from the elderly. Their repression alleviates cellular senescence and tissue degeneration and, to some extent, organismal aging. These findings indicate that the resurrection of ERVs is a hallmark and driving force of cellular senescence and tissue aging.


Assuntos
Envelhecimento , Retrovirus Endógenos , Idoso , Animais , Humanos , Camundongos , Envelhecimento/genética , Envelhecimento/patologia , Senescência Celular , Retrovirus Endógenos/genética , Primatas
2.
Cell ; 180(5): 984-1001.e22, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32109414

RESUMO

Aging causes a functional decline in tissues throughout the body that may be delayed by caloric restriction (CR). However, the cellular profiles and signatures of aging, as well as those ameliorated by CR, remain unclear. Here, we built comprehensive single-cell and single-nucleus transcriptomic atlases across various rat tissues undergoing aging and CR. CR attenuated aging-related changes in cell type composition, gene expression, and core transcriptional regulatory networks. Immune cells were increased during aging, and CR favorably reversed the aging-disturbed immune ecosystem. Computational prediction revealed that the abnormal cell-cell communication patterns observed during aging, including the excessive proinflammatory ligand-receptor interplay, were reversed by CR. Our work provides multi-tissue single-cell transcriptional landscapes associated with aging and CR in a mammal, enhances our understanding of the robustness of CR as a geroprotective intervention, and uncovers how metabolic intervention can act upon the immune system to modify the process of aging.


Assuntos
Envelhecimento/genética , Restrição Calórica , Sistema Imunitário/metabolismo , Transcriptoma/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Reprogramação Celular/genética , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Humanos , Ratos , Análise de Célula Única
3.
Cell ; 180(3): 585-600.e19, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-32004457

RESUMO

Molecular mechanisms of ovarian aging and female age-related fertility decline remain unclear. We surveyed the single-cell transcriptomic landscape of ovaries from young and aged non-human primates (NHPs) and identified seven ovarian cell types with distinct gene-expression signatures, including oocyte and six types of ovarian somatic cells. In-depth dissection of gene-expression dynamics of oocytes revealed four subtypes at sequential and stepwise developmental stages. Further analysis of cell-type-specific aging-associated transcriptional changes uncovered the disturbance of antioxidant signaling specific to early-stage oocytes and granulosa cells, indicative of oxidative damage as a crucial factor in ovarian functional decline with age. Additionally, inactivated antioxidative pathways, increased reactive oxygen species, and apoptosis were observed in granulosa cells from aged women. This study provides a comprehensive understanding of the cell-type-specific mechanisms underlying primate ovarian aging at single-cell resolution, revealing new diagnostic biomarkers and potential therapeutic targets for age-related human ovarian disorders.


Assuntos
Envelhecimento/genética , Ovário/fisiologia , Análise de Célula Única/métodos , Transcriptoma , Idoso , Animais , Antioxidantes/metabolismo , Apoptose/fisiologia , Atlas como Assunto , Biomarcadores , Linhagem Celular Tumoral , Feminino , Células da Granulosa/metabolismo , Humanos , Macaca fascicularis , Oócitos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia
4.
Circulation ; 149(20): 1578-1597, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38258575

RESUMO

BACKGROUND: Calcification of the aortic valve leads to increased leaflet stiffness and consequently results in the development of calcific aortic valve disease (CAVD). However, the underlying molecular and cellular mechanisms of calcification remain unclear. Here, we identified a novel aortic valve calcification-associated PIWI-interacting RNA (piRNA; AVCAPIR) that increases valvular calcification and promotes CAVD progression. METHODS: Using piRNA sequencing, we identified piRNAs contributing to the pathogenesis of CAVD that we termed AVCAPIRs. High-cholesterol diet-fed ApoE-/- mice with AVCAPIR knockout were used to examine the role of AVCAPIR in aortic valve calcification (AVC). Gain- and loss-of-function assays were conducted to determine the role of AVCAPIR in the induced osteogenic differentiation of human valvular interstitial cells. To dissect the mechanisms underlying AVCAPIR-elicited procalcific effects, we performed various analyses, including an RNA pulldown assay followed by liquid chromatography-tandem mass spectrometry, methylated RNA immunoprecipitation sequencing, and RNA sequencing. RNA pulldown and RNA immunoprecipitation assays were used to study piRNA interactions with proteins. RESULTS: We found that AVCAPIR was significantly upregulated during AVC and exhibited potential diagnostic value for CAVD. AVCAPIR deletion markedly ameliorated AVC in high-cholesterol diet-fed ApoE-/- mice, as shown by reduced thickness and calcium deposition in the aortic valve leaflets, improved echocardiographic parameters (decreased peak transvalvular jet velocity and mean transvalvular pressure gradient, as well as increased aortic valve area), and diminished levels of osteogenic markers (Runx2 and Osterix) in aortic valves. These results were confirmed in osteogenic medium-induced human valvular interstitial cells. Using unbiased protein-RNA screening and molecular validation, we found that AVCAPIR directly interacts with FTO (fat mass and obesity-associated protein), subsequently blocking its N6-methyladenosine demethylase activity. Further transcriptomic and N6-methyladenosine modification epitranscriptomic screening followed by molecular validation confirmed that AVCAPIR hindered FTO-mediated demethylation of CD36 mRNA transcripts, thus enhancing CD36 mRNA stability through the N6-methyladenosine reader IGF2BP1 (insulin-like growth factor 2 mRNA binding protein 1). In turn, the AVCAPIR-dependent increase in CD36 stabilizes its binding partner PCSK9 (proprotein convertase subtilisin/kexin type 9), a procalcific gene, at the protein level, which accelerates the progression of AVC. CONCLUSIONS: We identified a novel piRNA that induced AVC through an RNA epigenetic mechanism and provide novel insights into piRNA-directed theranostics in CAVD.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica , Calcinose , RNA Interferente Pequeno , Animais , Calcinose/metabolismo , Calcinose/genética , Calcinose/patologia , Valva Aórtica/metabolismo , Valva Aórtica/patologia , Valva Aórtica/anormalidades , Humanos , Camundongos , Estenose da Valva Aórtica/metabolismo , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/patologia , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/genética , Masculino , Osteogênese , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais de Doenças , Valvopatia Aórtica/metabolismo , Valvopatia Aórtica/genética , Valvopatia Aórtica/patologia , RNA de Interação com Piwi
5.
Nucleic Acids Res ; 50(D1): D1085-D1090, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34591960

RESUMO

Regeneration plays an instrumental role in biological development and damage repair by constructing and replacing cells, tissues, and organs. Since regenerative capacity declines with age, promoting regeneration is heralded as a potential strategy for delaying aging. On this premise, mechanisms that regulate regeneration have been extensively studied across species and in different tissues. However, an open and comprehensive database collecting and standardizing the abundant data generated in regeneration research, such as high-throughput sequencing data, remains to be developed. In this work, we constructed Regeneration Roadmap to systematically and comprehensively collect such information over 2.38 million data entries across 11 species and 36 tissues, including regeneration-related genes, bulk and single-cell transcriptomics, epigenomics, and pharmacogenomics data. In this database, users can explore regulatory and expression changes of regeneration-associated genes in different species and tissues. Regeneration Roadmap provides the research community with a long-awaited and valuable data resource featuring convenient computing and visualizing tools, which is publicly available at https://ngdc.cncb.ac.cn/regeneration/index.


Assuntos
Bases de Dados Factuais , Bases de Dados Genéticas , Regeneração/genética , Transcriptoma/genética , Envelhecimento/genética , Animais , Epigenômica , Humanos
6.
Nucleic Acids Res ; 49(8): 4203-4219, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33706382

RESUMO

Sirtuin 3 (SIRT3) is an NAD+-dependent deacetylase linked to a broad range of physiological and pathological processes, including aging and aging-related diseases. However, the role of SIRT3 in regulating human stem cell homeostasis remains unclear. Here we found that SIRT3 expression was downregulated in senescent human mesenchymal stem cells (hMSCs). CRISPR/Cas9-mediated depletion of SIRT3 led to compromised nuclear integrity, loss of heterochromatin and accelerated senescence in hMSCs. Further analysis indicated that SIRT3 interacted with nuclear envelope proteins and heterochromatin-associated proteins. SIRT3 deficiency resulted in the detachment of genomic lamina-associated domains (LADs) from the nuclear lamina, increased chromatin accessibility and aberrant repetitive sequence transcription. The re-introduction of SIRT3 rescued the disorganized heterochromatin and the senescence phenotypes. Taken together, our study reveals a novel role for SIRT3 in stabilizing heterochromatin and counteracting hMSC senescence, providing new potential therapeutic targets to ameliorate aging-related diseases.


Assuntos
Envelhecimento/metabolismo , Heterocromatina/metabolismo , Sirtuína 3/fisiologia , Envelhecimento/genética , Animais , Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Células Cultivadas , Senescência Celular/genética , Senescência Celular/fisiologia , Técnicas de Inativação de Genes , Células HEK293 , Heterocromatina/genética , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Nus , Camundongos SCID , Membrana Nuclear/metabolismo , Domínios Proteicos , Sirtuína 3/química , Sirtuína 3/genética
7.
PLoS Biol ; 17(4): e3000201, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30933975

RESUMO

Cellular senescence is a driver of various aging-associated disorders, including osteoarthritis. Here, we identified a critical role for Yes-associated protein (YAP), a major effector of Hippo signaling, in maintaining a younger state of human mesenchymal stem cells (hMSCs) and ameliorating osteoarthritis in mice. Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR associated protein 9 nuclease (Cas9)-mediated knockout (KO) of YAP in hMSCs resulted in premature cellular senescence. Mechanistically, YAP cooperated with TEA domain transcriptional factor (TEAD) to activate the expression of forkhead box D1 (FOXD1), a geroprotective protein. YAP deficiency led to the down-regulation of FOXD1. In turn, overexpression of YAP or FOXD1 rejuvenated aged hMSCs. Moreover, intra-articular administration of lentiviral vector encoding YAP or FOXD1 attenuated the development of osteoarthritis in mice. Collectively, our findings reveal YAP-FOXD1, a novel aging-associated regulatory axis, as a potential target for gene therapy to alleviate osteoarthritis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Osteoartrite/metabolismo , Osteoartrite/patologia , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proliferação de Células/genética , Senescência Celular/fisiologia , Fatores de Transcrição Forkhead/genética , Xenoenxertos , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Osteoartrite/genética , Transdução de Sinais , Fatores de Transcrição/genética , Ativação Transcricional , Regulação para Cima , Proteínas de Sinalização YAP
8.
Nucleic Acids Res ; 48(19): 11083-11096, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33035345

RESUMO

N6-Methyladenosine (m6A) messenger RNA methylation is a well-known epitranscriptional regulatory mechanism affecting central biological processes, but its function in human cellular senescence remains uninvestigated. Here, we found that levels of both m6A RNA methylation and the methyltransferase METTL3 were reduced in prematurely senescent human mesenchymal stem cell (hMSC) models of progeroid syndromes. Transcriptional profiling of m6A modifications further identified MIS12, for which m6A modifications were reduced in both prematurely senescent hMSCs and METTL3-deficient hMSCs. Knockout of METTL3 accelerated hMSC senescence whereas overexpression of METTL3 rescued the senescent phenotypes. Mechanistically, loss of m6A modifications accelerated the turnover and decreased the expression of MIS12 mRNA while knockout of MIS12 accelerated cellular senescence. Furthermore, m6A reader IGF2BP2 was identified as a key player in recognizing and stabilizing m6A-modified MIS12 mRNA. Taken together, we discovered that METTL3 alleviates hMSC senescence through m6A modification-dependent stabilization of the MIS12 transcript, representing a novel epitranscriptional mechanism in premature stem cell senescence.


Assuntos
Adenosina/análogos & derivados , Metiltransferases/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Progéria/genética , RNA Mensageiro/metabolismo , Síndrome de Werner/genética , Adenosina/genética , Células Cultivadas , Senescência Celular , Humanos , Células-Tronco Mesenquimais , Metilação , Proteínas de Ligação a RNA/metabolismo
9.
Nucleic Acids Res ; 48(11): 6001-6018, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32427330

RESUMO

Zinc finger protein with KRAB and SCAN domains 3 (ZKSCAN3) has long been known as a master transcriptional repressor of autophagy. Here, we identify a novel role for ZKSCAN3 in alleviating senescence that is independent of its autophagy-related activity. Downregulation of ZKSCAN3 is observed in aged human mesenchymal stem cells (hMSCs) and depletion of ZKSCAN3 accelerates senescence of these cells. Mechanistically, ZKSCAN3 maintains heterochromatin stability via interaction with heterochromatin-associated proteins and nuclear lamina proteins. Further study shows that ZKSCAN3 deficiency results in the detachment of genomic lamina-associated domains (LADs) from the nuclear lamina, loss of heterochromatin, a more accessible chromatin status and consequently, aberrant transcription of repetitive sequences. Overexpression of ZKSCAN3 not only rescues premature senescence phenotypes in ZKSCAN3-deficient hMSCs but also rejuvenates physiologically and pathologically senescent hMSCs. Together, these data reveal for the first time that ZKSCAN3 functions as an epigenetic modulator to maintain heterochromatin organization and thereby attenuate cellular senescence. Our findings establish a new functional link among ZKSCAN3, epigenetic regulation, and stem cell aging.


Assuntos
Senescência Celular , Epigênese Genética , Heterocromatina/metabolismo , Fatores de Transcrição/metabolismo , Animais , Senescência Celular/genética , Regulação para Baixo , Heterocromatina/genética , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Fatores de Transcrição/deficiência
10.
Eur Heart J ; 42(30): 2935-2951, 2021 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-34179958

RESUMO

AIMS: The morbidity and mortality rates of calcific aortic valve disease (CAVD) remain high while treatment options are limited. Here, we evaluated the role and therapeutic value of dual-specificity phosphatase 26 (DUSP26) in CAVD. METHODS AND RESULTS: Microarray profiling of human calcific aortic valves and normal controls demonstrated that DUSP26 was significantly up-regulated in calcific aortic valves. ApoE-/- mice fed a normal diet or a high cholesterol diet (HCD) were infected with adeno-associated virus serotype 2 carrying DUSP26 short-hairpin RNA to examine the effects of DUSP26 silencing on aortic valve calcification. DUSP26 silencing ameliorated aortic valve calcification in HCD-treated ApoE-/- mice, as evidenced by reduced thickness and calcium deposition in the aortic valve leaflets, improved echocardiographic parameters (decreased peak transvalvular jet velocity and mean transvalvular pressure gradient, as well as increased aortic valve area), and decreased levels of osteogenic markers (Runx2, osterix, and osteocalcin) in the aortic valves. These results were confirmed in osteogenic medium-induced human valvular interstitial cells. Immunoprecipitation, liquid chromatography-tandem mass spectrometry, and functional assays revealed that dipeptidyl peptidase-4 (DPP4) interacted with DUSP26 to mediate the procalcific effects of DUSP26. High N6-methyladenosine levels up-regulated DUSP26 in CAVD; in turn, DUSP26 activated DPP4 by antagonizing mouse double minute 2-mediated ubiquitination and degradation of DPP4, thereby promoting CAVD progression. CONCLUSION: DUSP26 promotes aortic valve calcification by inhibiting DPP4 degradation. Our findings identify a previously unrecognized mechanism of DPP4 up-regulation in CAVD, suggesting that DUSP26 silencing or inhibition is a viable therapeutic strategy to impede CAVD progression.


Assuntos
Estenose da Valva Aórtica , Valva Aórtica/patologia , Calcinose , Fosfatases de Especificidade Dupla , Fosfatases da Proteína Quinase Ativada por Mitógeno , Animais , Valva Aórtica/metabolismo , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/metabolismo , Calcinose/genética , Calcinose/metabolismo , Células Cultivadas , Dipeptidil Peptidase 4 , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Humanos , Camundongos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-mdm2 , Ubiquitinação
11.
Circ Res ; 123(7): 773-786, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30355081

RESUMO

Aging is associated with a progressive decline in cardiovascular structure and function. Accumulating evidence links cardiovascular aging to epigenetic alterations encompassing a complex interplay of DNA methylation, histone posttranslational modifications, and dynamic nucleosome occupancy governed by numerous epigenetic factors. Advances in genomics technology have led to a profound understanding of chromatin reorganization in both cardiovascular aging and diseases. This review summarizes recent discoveries in epigenetic mechanisms involved in cardiovascular aging and diseases and discusses potential therapeutic strategies to retard cardiovascular aging and conquer related diseases through the rejuvenation of epigenetic signatures to a young state.


Assuntos
Envelhecimento/genética , Doenças Cardiovasculares/genética , Sistema Cardiovascular , Metilação de DNA , Epigênese Genética , Fatores Etários , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patologia , Sistema Cardiovascular/fisiopatologia , Montagem e Desmontagem da Cromatina , Regulação da Expressão Gênica , Predisposição Genética para Doença , Nível de Saúde , Envelhecimento Saudável , Humanos , Expectativa de Vida , Fenótipo , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Fatores de Risco , Transdução de Sinais
13.
Circ Res ; 116(11): 1835-49, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25999423

RESUMO

Mitochondria are highly dynamic, except in adult cardiomyocytes. Yet, the fission and fusion-promoting proteins that mediate mitochondrial dynamism are highly expressed in, and essential to the normal functioning of, hearts. Here, we review accumulating evidence supporting important roles for mitochondrial fission and fusion in cardiac mitochondrial quality control, focusing on the PTEN-induced putative kinase 1-Parkin mitophagy pathway. Based in part on recent findings from in vivo mouse models in which mitofusin-mediated mitochondrial fusion or dynamin-related protein 1-mediated mitochondrial fission was conditionally interrupted in cardiac myocytes, we propose several new concepts that may provide insight into the cardiac mitochondrial dynamism-mitophagy interactome.


Assuntos
Mitocôndrias Cardíacas/metabolismo , Dinâmica Mitocondrial , Mitofagia , Transdução de Sinais , Animais , Dinaminas/metabolismo , Humanos , Modelos Biológicos , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
14.
Circ Res ; 117(4): 346-51, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26038571

RESUMO

RATIONALE: The role of Parkin in hearts is unclear. Germ-line Parkin knockout mice have normal hearts, but Parkin is protective in cardiac ischemia. Parkin-mediated mitophagy is reportedly either irrelevant, or a major factor, in the lethal cardiomyopathy evoked by cardiac myocyte-specific interruption of dynamin-related protein 1 (Drp1)-mediated mitochondrial fission. OBJECTIVE: To understand the role of Parkin-mediated mitophagy in normal and mitochondrial fission-defective adult mouse hearts. METHODS AND RESULTS: Parkin mRNA and protein were present at low levels in normal mouse hearts, but were upregulated after cardiac myocyte-directed Drp1 gene deletion in adult mice. Alone, forced cardiac myocyte Parkin overexpression activated mitophagy without adverse effects. Likewise, cardiac myocyte-specific Parkin deletion evoked no adult cardiac phenotype, revealing no essential function for, and tolerance of, Parkin-mediated mitophagy in normal hearts. Concomitant conditional Parkin deletion with Drp1 ablation in adult mouse hearts prevented Parkin upregulation in mitochondria of fission-defective hearts, also increasing 6-week survival, improving ventricular ejection performance, mitigating adverse cardiac remodeling, and decreasing cardiac myocyte necrosis and replacement fibrosis. Underlying the Parkin knockout rescue was suppression of Drp1-induced hyper-mitophagy, assessed as ubiquitination of mitochondrial proteins and mitochondrial association of autophagosomal p62/sequestosome 1 (SQSTM1) and processed microtubule-associated protein 1 light chain 3 (LC3-II). Consequently, mitochondrial content of Drp1-deficient hearts was preserved. Parkin deletion did not alter characteristic mitochondrial enlargement of Drp1-deficient cardiac myocytes. CONCLUSIONS: Parkin is rare in normal hearts and dispensable for constitutive mitophagic quality control. Ablating Drp1 in adult mouse cardiac myocytes not only interrupts mitochondrial fission, but also markedly upregulates Parkin, thus provoking mitophagic mitochondrial depletion that contributes to the lethal cardiomyopathy.


Assuntos
Cardiomiopatias/metabolismo , Mitocôndrias Cardíacas/metabolismo , Dinâmica Mitocondrial , Mitofagia , Miocárdio/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Cardiomiopatias/genética , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Dinaminas/genética , Dinaminas/metabolismo , Fibrose , Regulação da Expressão Gênica , Predisposição Genética para Doença , Camundongos Knockout , Mitocôndrias Cardíacas/ultraestrutura , Miocárdio/ultraestrutura , Necrose , Fenótipo , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Tempo , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética , Função Ventricular Esquerda , Remodelação Ventricular
15.
Circ Res ; 114(2): 257-65, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24192653

RESUMO

RATIONALE: Dysfunctional Parkin-mediated mitophagic culling of senescent or damaged mitochondria is a major pathological process underlying Parkinson disease and a potential genetic mechanism of cardiomyopathy. Despite epidemiological associations between Parkinson disease and heart failure, the role of Parkin and mitophagic quality control in maintaining normal cardiac homeostasis is poorly understood. OBJECTIVE: We used germline mutants and cardiac-specific RNA interference to interrogate Parkin regulation of cardiomyocyte mitochondria and examine functional crosstalk between mitophagy and mitochondrial dynamics in Drosophila heart tubes. METHODS AND RESULTS: Transcriptional profiling of Parkin knockout mouse hearts revealed compensatory upregulation of multiple related E3 ubiquitin ligases. Because Drosophila lack most of these redundant genes, we examined heart tubes of parkin knockout flies and observed accumulation of enlarged hollow donut mitochondria with dilated cardiomyopathy, which could be rescued by cardiomyocyte-specific Parkin expression. Identical abnormalities were induced by cardiomyocyte-specific Parkin suppression using 2 different inhibitory RNAs. Parkin-deficient cardiomyocyte mitochondria exhibited dysmorphology, depolarization, and reactive oxygen species generation without calcium cycling abnormalities, pointing to a primary mitochondrial defect. Suppressing cardiomyocyte mitochondrial fusion in Parkin-deficient fly heart tubes completely prevented the cardiomyopathy and corrected mitochondrial dysfunction without normalizing mitochondrial dysmorphology, demonstrating a central role for mitochondrial fusion in the cardiomyopathy provoked by impaired mitophagy. CONCLUSIONS: Parkin deficiency and resulting mitophagic disruption produces cardiomyopathy in part by contamination of the cardiomyocyte mitochondrial pool through fusion between improperly retained dysfunctional/senescent and normal mitochondria. Limiting mitochondrial contagion by inhibiting organelle fusion shows promise for minimizing organ dysfunction produced by defective mitophagic signaling.


Assuntos
Cardiomiopatia Dilatada/enzimologia , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias Cardíacas/enzimologia , Dinâmica Mitocondrial , Mitofagia , Miócitos Cardíacos/enzimologia , Ubiquitina-Proteína Ligases/deficiência , Animais , Sinalização do Cálcio , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/prevenção & controle , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Perfilação da Expressão Gênica , Genótipo , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Mitocôndrias Cardíacas/patologia , Mutação , Miócitos Cardíacos/patologia , Fenótipo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fatores de Tempo , Ubiquitina-Proteína Ligases/genética
16.
Circ Res ; 115(3): 348-53, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24874428

RESUMO

RATIONALE: Mitochondrial reactive oxygen species (ROS) are implicated in aging, chronic degenerative neurological syndromes, and myopathies. On the basis of free radical hypothesis, dietary, pharmacological, and genetic ROS suppression has been tested to minimize tissue damage, with remarkable therapeutic efficacy. The effects of mitochondrial-specific ROS suppression in primary mitophagic dysfunction are unknown. OBJECTIVE: An in vivo dose-ranging analysis of ROS suppression in an experimental cardiomyopathy provoked by defective mitochondrial clearance. METHODS AND RESULTS: Mice lacking mitofusin 2 (Mfn2) in hearts have impaired parkin-mediated mitophagy leading to accumulation of damaged ROS-producing organelles and progressive heart failure. As expected, cardiomyocyte-directed expression of mitochondrial-targeted catalase at modest levels normalized mitochondrial ROS production and prevented mitochondrial depolarization, respiratory impairment, and structural degeneration in Mfn2 null hearts. In contrast, catalase expression at higher levels that supersuppressed mitochondrial ROS failed to improve either mitochondrial fitness or cardiomyopathy, revealing that ROS toxicity is not the primary mechanism for cardiac degeneration. Lack of benefit from supersuppressing ROS was associated with failure to invoke secondary autophagic pathways of mitochondrial quality control, revealing a role for ROS signaling in mitochondrial clearance. Mitochondrial permeability transition pore function was normal, and genetic inhibition of mitochondrial permeability transition pore function did not alter mitochondrial or cardiac degeneration, in Mfn2 null hearts. CONCLUSIONS: Local mitochondrial ROS (1) contribute to mitochondrial degeneration and (2) activate mitochondrial quality control mechanisms. A therapeutic window for mitochondrial ROS suppression should minimize the former while retaining the latter, which we achieved by expressing lower levels of catalase.


Assuntos
Autofagia/fisiologia , Cardiomiopatias/metabolismo , GTP Fosfo-Hidrolases/genética , Mitocôndrias Cardíacas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Animais , Cardiomiopatias/patologia , Peptidil-Prolil Isomerase F , Ciclofilinas/genética , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos Knockout , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Membranas Mitocondriais/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/genética , Oxidantes/metabolismo
17.
J Mol Cell Cardiol ; 78: 123-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25252175

RESUMO

Cardiomyocyte mitochondria have an intimate physical and functional relationship with sarcoplasmic reticulum (SR). Under normal conditions mitochondrial ATP is essential to power SR calcium cycling that drives phasic contraction/relaxation, and changes in SR calcium release are sensed by mitochondria and used to modulate oxidative phosphorylation according to metabolic need. When perturbed, mitochondrial-SR calcium crosstalk can evoke programmed cell death. Physical proximity and functional interplay between mitochondria and SR are maintained in part through tethering of these two organelles by the membrane protein mitofusin 2 (Mfn2). Here we review and discuss findings from our two laboratories that derive from genetic manipulation of Mfn2 and closely related Mfn1 in mouse hearts and other experimental systems. By comparing the findings of our two independent research efforts we arrive at several conclusions that appear to be strongly supported, and describe a few areas of incomplete understanding that will require further study. In so doing we hope to clarify some misconceptions regarding the many varied roles of Mfn2 as both physical trans-organelle tether and mitochondrial fusion protein. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease."


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Retículo Sarcoplasmático/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio , GTP Fosfo-Hidrolases/genética , Deleção de Genes , Humanos , Mitocôndrias/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Proteínas Mitocondriais/genética , Miócitos Cardíacos/metabolismo
18.
J Mol Cell Cardiol ; 80: 71-80, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25555803

RESUMO

Mitochondrial dynamism (fusion and fission) is responsible for remodeling interconnected mitochondrial networks in some cell types. Adult cardiac myocytes lack mitochondrial networks, and their mitochondria are inherently "fragmented". Mitochondrial fusion/fission is so infrequent in cardiomyocytes as to not be observable under normal conditions, suggesting that mitochondrial dynamism may be dispensable in this cell type. However, we previously observed that cardiomyocyte-specific genetic suppression of mitochondrial fusion factors optic atrophy 1 (Opa1) and mitofusin/MARF evokes cardiomyopathy in Drosophila hearts. We posited that fusion-mediated remodeling of mitochondria may be critical for cardiac homeostasis, although never directly observed. Alternately, we considered that inner membrane Opa1 and outer membrane mitofusin/MARF might have other as-yet poorly described roles that affect mitochondrial and cardiac function. Here we compared heart tube function in three models of mitochondrial fragmentation in Drosophila cardiomyocytes: Drp1 expression, Opa1 RNAi, and mitofusin MARF RNA1. Mitochondrial fragmentation evoked by enhanced Drp1-mediated fission did not adversely impact heart tube function. In contrast, RNAi-mediated suppression of either Opa1 or mitofusin/MARF induced cardiac dysfunction associated with mitochondrial depolarization and ROS production. Inhibiting ROS by overexpressing superoxide dismutase (SOD) or suppressing ROMO1 prevented mitochondrial and heart tube dysfunction provoked by Opa1 RNAi, but not by mitofusin/MARF RNAi. In contrast, enhancing the ability of endoplasmic/sarcoplasmic reticulum to handle stress by expressing Xbp1 rescued the cardiomyopathy of mitofusin/MARF insufficiency without improving that caused by Opa1 deficiency. We conclude that decreased mitochondrial size is not inherently detrimental to cardiomyocytes. Rather, preservation of mitochondrial function by Opa1 located on the inner mitochondrial membrane, and prevention of ER stress by mitofusin/MARF located on the outer mitochondrial membrane, are central functions of these "mitochondrial fusion proteins".


Assuntos
Cardiomiopatias/metabolismo , Estresse do Retículo Endoplasmático , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Retículo Sarcoplasmático/metabolismo , Animais , Animais Geneticamente Modificados , Cardiomiopatias/genética , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Proteínas do Citoesqueleto/genética , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Estresse do Retículo Endoplasmático/genética , Feminino , Proteínas de Ligação ao GTP/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Mitocôndrias/genética , Dinâmica Mitocondrial/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Especificidade de Órgãos/genética , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase-1
20.
Adv Sci (Weinh) ; 11(18): e2307233, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38487926

RESUMO

The gut microbiome has emerged as a potential target for the treatment of cardiovascular disease. Ischemia/reperfusion (I/R) after myocardial infarction is a serious complication and whether certain gut bacteria can serve as a treatment option remains unclear. Lactobacillus reuteri (L. reuteri) is a well-studied probiotic that can colonize mammals including humans with known cholesterol-lowering properties and anti-inflammatory effects. Here, the prophylactic cardioprotective effects of L. reuteri or its metabolite γ-aminobutyric acid (GABA) against acute ischemic cardiac injury caused by I/R surgery are demonstrated. The prophylactic gavage of L. reuteri or GABA confers cardioprotection mainly by suppressing cardiac inflammation upon I/R. Mechanistically, GABA gavage results in a decreased number of proinflammatory macrophages in I/R hearts and GABA gavage no longer confers any cardioprotection in I/R hearts upon the clearance of macrophages. In vitro studies with LPS-stimulated bone marrow-derived macrophages (BMDM) further reveal that GABA inhibits the polarization of macrophages toward the proinflammatory M1 phenotype by inhibiting lysosomal leakage and NLRP3 inflammasome activation. Together, this study demonstrates that the prophylactic oral administration of L. reuteri or its metabolite GABA attenuates macrophage-mediated cardiac inflammation and therefore alleviates cardiac dysfunction after I/R, thus providing a new prophylactic strategy to mitigate acute ischemic cardiac injury.


Assuntos
Modelos Animais de Doenças , Limosilactobacillus reuteri , Camundongos Endogâmicos C57BL , Probióticos , Ácido gama-Aminobutírico , Animais , Limosilactobacillus reuteri/metabolismo , Camundongos , Ácido gama-Aminobutírico/metabolismo , Probióticos/administração & dosagem , Probióticos/uso terapêutico , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Macrófagos/metabolismo , Microbioma Gastrointestinal , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA