Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Biol Chem ; 296: 100694, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33895132

RESUMO

Myosin heavy chain 7b (MYH7b) is an ancient member of the myosin heavy chain motor protein family that is expressed in striated muscles. In mammalian cardiac muscle, MYH7b RNA is expressed along with two other myosin heavy chains, ß-myosin heavy chain (ß-MyHC) and α-myosin heavy chain (α-MyHC). However, unlike ß-MyHC and α-MyHC, which are maintained in a careful balance at the protein level, the MYH7b locus does not produce a full-length protein in the heart due to a posttranscriptional exon-skipping mechanism that occurs in a tissue-specific manner. Whether this locus has a role in the heart beyond producing its intronic microRNA, miR-499, was unclear. Using cardiomyocytes derived from human induced pluripotent stem cells as a model system, we found that the noncoding exon-skipped RNA (lncMYH7b) affects the transcriptional landscape of human cardiomyocytes, independent of miR-499. Specifically, lncMYH7b regulates the ratio of ß-MyHC to α-MyHC, which is crucial for cardiac contractility. We also found that lncMYH7b regulates beat rate and sarcomere formation in cardiomyocytes. This regulation is likely achieved through control of a member of the TEA domain transcription factor family (TEAD3, which is known to regulate ß-MyHC). Therefore, we conclude that this ancient gene has been repurposed by alternative splicing to produce a regulatory long-noncoding RNA in the human heart that affects cardiac myosin composition.


Assuntos
Miosinas Cardíacas/metabolismo , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/metabolismo , RNA Longo não Codificante/genética , Miosinas Cardíacas/química , Humanos , Células-Tronco Pluripotentes Induzidas , MicroRNAs/genética , Simulação de Dinâmica Molecular , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/química , Conformação Proteica
2.
Mol Ther ; 25(3): 694-704, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28202391

RESUMO

MicroRNAs (miRNAs) are important regulators of biology and disease. Recent animal efficacy studies validate the therapeutic benefit of miRNA modulation and underscore the therapeutic value of miRNA-targeting oligonucleotides. However, whether disease conditions (stress) influence the pharmacological effects of an anti-miR is currently unknown. To study the effect of disease on target regulation after anti-miR treatment, we injected animals with anti-miR-208a, a synthetic oligonucleotide that inhibits the cardiomyocyte-specific miR-208a. Our data indicate that the presence of stress increases the number of regulated miR-208a targets, and that higher stress levels correlate with stronger target derepression. Additionally, the type of stress also influences which targets are regulated upon miR-208a inhibition. Studies in a large animal model indicate a similar stress-dependent anti-miR effect. Subsequent in vitro studies suggest that the influence of stress on anti-miR efficacy depends at least in part on increased cellular anti-miR uptake. These data indicate that the pharmacological effect of anti-miRs is stronger under disease conditions, and that both the type and severity of disease determine the therapeutic outcome. These facts will be important for assessing the therapeutic dose and predicting the therapeutic outcome when applying anti-miRs in a clinical setting.


Assuntos
Antagomirs/genética , MicroRNAs/genética , Miócitos Cardíacos/metabolismo , Estresse Fisiológico/genética , Animais , Células Cultivadas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Masculino , Interferência de RNA , Ratos , Suínos
3.
Proc Natl Acad Sci U S A ; 112(41): 12812-7, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26417068

RESUMO

The contribution of endothelial-derived miR-17∼92 to ischemia-induced arteriogenesis has not been investigated in an in vivo model. In the present study, we demonstrate a critical role for the endothelial-derived miR-17∼92 cluster in shaping physiological and ischemia-triggered arteriogenesis. Endothelial-specific deletion of miR-17∼92 results in an increase in collateral density limbs and hearts and in ischemic limbs compared with control mice, and consequently improves blood flow recovery. Individual cluster components positively or negatively regulate endothelial cell (EC) functions in vitro, and, remarkably, ECs lacking the cluster spontaneously form cords in a manner rescued by miR-17a, -18a, and -19a. Using both in vitro and in vivo analyses, we identified FZD4 and LRP6 as targets of miR-19a/b. Both of these targets were up-regulated in 17∼92 KO ECs compared with control ECs, and both were shown to be targeted by miR-19 using luciferase assays. We demonstrate that miR-19a negatively regulates FZD4, its coreceptor LRP6, and WNT signaling, and that antagonism of miR-19a/b in aged mice improves blood flow recovery after ischemia and reduces repression of these targets. Collectively, these data provide insights into miRNA regulation of arterialization and highlight the importance of vascular WNT signaling in maintaining arterial blood flow.


Assuntos
Receptores Frizzled/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , MicroRNAs/metabolismo , Família Multigênica/fisiologia , Neovascularização Fisiológica/fisiologia , Via de Sinalização Wnt/fisiologia , Animais , Receptores Frizzled/genética , Isquemia/genética , Isquemia/metabolismo , Isquemia/patologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética
4.
Cardiovasc Drugs Ther ; 31(4): 445-458, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28735360

RESUMO

PURPOSE: The need for novel approaches to cardiovascular drug development served as the impetus to convene an open meeting of experts from the pharmaceutical industry and academia to assess the challenges and develop solutions for drug discovery in cardiovascular disease. METHODS: The Novel Cardiovascular Therapeutics Summit first reviewed recent examples of ongoing or recently completed programs translating basic science observations to targeted drug development, highlighting successes (protein convertase sutilisin/kexin type 9 [PCSK9] and neprilysin inhibition) and targets still under evaluation (cholesteryl ester transfer protein [CETP] inhibition), with the hope of gleaning key lessons to successful drug development in the current era. Participants then reviewed the use of innovative approaches being explored to facilitate rapid and more cost-efficient evaluations of drug candidates in a short timeframe. RESULTS: We summarize observations gleaned from this summit and offer insight into future cardiovascular drug development. CONCLUSIONS: The rapid development in genetic and high-throughput drug evaluation technologies, coupled with new approaches to rapidly evaluate potential cardiovascular therapies with in vitro techniques, offer opportunities to identify new drug targets for cardiovascular disease, study new therapies with better efficiency and higher throughput in the preclinical setting, and more rapidly bring the most promising therapies to human testing. However, there must be a critical interface between industry and academia to guide the future of cardiovascular drug development. The shared interest among academic institutions and pharmaceutical companies in developing promising therapies to address unmet clinical needs for patients with cardiovascular disease underlies and guides innovation and discovery platforms that are significantly altering the landscape of cardiovascular drug development.


Assuntos
Fármacos Cardiovasculares/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Desenho de Fármacos , Animais , Fármacos Cardiovasculares/farmacologia , Doenças Cardiovasculares/fisiopatologia , Descoberta de Drogas/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Indústria Farmacêutica , Humanos
5.
Genes Dev ; 23(14): 1625-30, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19605684

RESUMO

Histone deacetylases (Hdacs) are transcriptional repressors with crucial roles in mammalian development. Here we provide evidence that Hdac8 specifically controls patterning of the skull by repressing a subset of transcription factors in cranial neural crest cells. Global deletion of Hdac8 in mice leads to perinatal lethality due to skull instability, and this is phenocopied by conditional deletion of Hdac8 in cranial neural crest cells. Hdac8 specifically represses the aberrant expression of homeobox transcription factors such as Otx2 and Lhx1. These findings reveal how the identity and patterning of vertebrate-specific portions of the skull are epigenetically controlled by a histone deacetylase.


Assuntos
Padronização Corporal/genética , Epigênese Genética , Histona Desacetilases/metabolismo , Crânio/embriologia , Animais , Deleção de Genes , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas com Homeodomínio LIM , Camundongos , Fatores de Transcrição Otx/metabolismo , Crânio/anormalidades , Fatores de Transcrição
6.
Nat Rev Genet ; 10(1): 32-42, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19065135

RESUMO

Histone deacetylases (HDACs) are part of a vast family of enzymes that have crucial roles in numerous biological processes, largely through their repressive influence on transcription. The expression of many HDAC isoforms in eukaryotic cells raises questions about their possible specificity or redundancy, and whether they control global or specific programmes of gene expression. Recent analyses of HDAC knockout mice have revealed highly specific functions of individual HDACs in development and disease. Mutant mice lacking individual HDACs are a powerful tool for defining the functions of HDACs in vivo and the molecular targets of HDAC inhibitors in disease.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Histona Desacetilases/fisiologia , Acetilação , Animais , Cardiomegalia/metabolismo , Cardiomegalia/terapia , Condrócitos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inibidores de Histona Desacetilases , Histona Desacetilases/genética , Histonas/metabolismo , Camundongos , Modelos Biológicos , Músculo Esquelético/metabolismo
7.
Proc Natl Acad Sci U S A ; 109(8): E481-9, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22223663

RESUMO

Dramatic changes in chromatin structure and histone modification occur during oocyte growth, as well as a global cessation of transcription. The role of histone modifications in these processes is poorly understood. We report the effect of conditionally deleting Hdac1 and Hdac2 on oocyte development. Deleting either gene has little or no effect on oocyte development, whereas deleting both genes results in follicle development arrest at the secondary follicle stage. This developmental arrest is accompanied by substantial perturbation of the transcriptome and a global reduction in transcription even though histone acetylation is markedly increased. There is no apparent change in histone repressive marks, but there is a pronounced decrease in histone H3K4 methylation, an activating mark. The decrease in H3K4 methylation is likely a result of increased expression of Kdm5b because RNAi-mediated targeting of Kdm5b in double-mutant oocytes results in an increase in H3K4 methylation. An increase in TRP53 acetylation also occurs in mutant oocytes and may contribute to the observed increased incidence of apoptosis. Taken together, these results suggest seminal roles of acetylation of histone and nonhistone proteins in oocyte development.


Assuntos
Apoptose/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Oócitos/enzimologia , Transcrição Gênica , Acetilação/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Marcação de Genes , Histona Desacetilase 1/genética , Histona Desacetilase 2/genética , Histonas/metabolismo , Ácidos Hidroxâmicos/farmacologia , Infertilidade Feminina/enzimologia , Infertilidade Feminina/patologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Lisina/metabolismo , Metilação/efeitos dos fármacos , Camundongos , Oócitos/efeitos dos fármacos , Oócitos/patologia , Oogênese/efeitos dos fármacos , Oogênese/genética , Tamanho do Órgão/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , Folículo Ovariano/patologia , Interferência de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/genética , Proteína Supressora de Tumor p53/metabolismo
8.
Circ Res ; 110(1): 71-81, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22052914

RESUMO

RATIONALE: Myocardial infarction (MI) is a leading cause of death worldwide. Because endogenous cardiac repair mechanisms are not sufficient for meaningful tissue regeneration, MI results in loss of cardiac tissue and detrimental remodeling events. MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression in a sequence dependent manner. Our previous data indicate that miRNAs are dysregulated in response to ischemic injury of the heart and actively contribute to cardiac remodeling after MI. OBJECTIVE: This study was designed to determine whether miRNAs are dysregulated on ischemic damage in porcine cardiac tissues and whether locked nucleic acid (LNA)-modified anti-miR chemistries can target cardiac expressed miRNAs to therapeutically inhibit miR-15 on ischemic injury. METHODS AND RESULTS: Our data indicate that the miR-15 family, which includes 6 closely related miRNAs, is regulated in the infarcted region of the heart in response to ischemia-reperfusion injury in mice and pigs. LNA-modified chemistries can effectively silence miR-15 family members in vitro and render cardiomyocytes resistant to hypoxia-induced cardiomyocyte cell death. Correspondingly, systemic delivery of miR-15 anti-miRs dose-dependently represses miR-15 in cardiac tissue of both mice and pigs, whereas therapeutic targeting of miR-15 in mice reduces infarct size and cardiac remodeling and enhances cardiac function in response to MI. CONCLUSIONS: Oligonucleotide-based therapies using LNA-modified chemistries for modulating cardiac miRNAs in the setting of heart disease are efficacious and validate miR-15 as a potential therapeutic target for the manipulation of cardiac remodeling and function in the setting of ischemic injury.


Assuntos
MicroRNAs/antagonistas & inibidores , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Sobrevivência Celular/efeitos dos fármacos , Feminino , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/efeitos dos fármacos , Modelos Animais , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Suínos
9.
Circulation ; 124(14): 1537-47, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21900086

RESUMO

BACKGROUND: Diastolic dysfunction in response to hypertrophy is a major clinical syndrome with few therapeutic options. MicroRNAs act as negative regulators of gene expression by inhibiting translation or promoting degradation of target mRNAs. Previously, we reported that genetic deletion of the cardiac-specific miR-208a prevents pathological cardiac remodeling and upregulation of Myh7 in response to pressure overload. Whether this miRNA might contribute to diastolic dysfunction or other forms of heart disease is currently unknown. METHODS AND RESULTS: Here, we show that systemic delivery of an antisense oligonucleotide induces potent and sustained silencing of miR-208a in the heart. Therapeutic inhibition of miR-208a by subcutaneous delivery of antimiR-208a during hypertension-induced heart failure in Dahl hypertensive rats dose-dependently prevents pathological myosin switching and cardiac remodeling while improving cardiac function, overall health, and survival. Transcriptional profiling indicates that antimiR-208a evokes prominent effects on cardiac gene expression; plasma analysis indicates significant changes in circulating levels of miRNAs on antimiR-208a treatment. CONCLUSIONS: These studies indicate the potential of oligonucleotide-based therapies for modulating cardiac miRNAs and validate miR-208 as a potent therapeutic target for the modulation of cardiac function and remodeling during heart disease progression.


Assuntos
Terapia Genética , Insuficiência Cardíaca Diastólica/tratamento farmacológico , Coração/fisiopatologia , Terapia de Alvo Molecular , Oligonucleotídeos Antissenso/uso terapêutico , Animais , Vias de Administração de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Eletrocardiografia , Perfilação da Expressão Gênica , Insuficiência Cardíaca Diastólica/diagnóstico por imagem , Insuficiência Cardíaca Diastólica/etiologia , Insuficiência Cardíaca Diastólica/genética , Hipertensão/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/antagonistas & inibidores , MicroRNAs/sangue , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/genética , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/farmacologia , Interferência de RNA , Ratos , Ratos Endogâmicos Dahl , Transcrição Gênica/efeitos dos fármacos , Ultrassonografia , Remodelação Ventricular/efeitos dos fármacos
10.
Proc Natl Acad Sci U S A ; 106(19): 7876-81, 2009 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-19380719

RESUMO

The molecular mechanism by which neural progenitor cells commit to a specified lineage of the central nervous system remains unknown. We show that HDAC1 and HDAC2 redundantly control neuronal development and are required for neuronal specification. Mice lacking HDAC1 or HDAC2 in neuronal precursors show no overt histoarchitectural phenotypes, whereas deletion of both HDAC1 and HDAC2 in developing neurons results in severe hippocampal abnormalities, absence of cerebellar foliation, disorganization of cortical neurons, and lethality by postnatal day 7. These abnormalities in brain formation can be attributed to a failure of neuronal precursors to differentiate into mature neurons and to excessive cell death. These results reveal redundant and essential roles for HDAC1 and HDAC2 in the progression of neuronal precursors to mature neurons in vivo.


Assuntos
Encéfalo/embriologia , Histona Desacetilases/fisiologia , Neurônios/fisiologia , Proteínas Repressoras/fisiologia , Alelos , Animais , Diferenciação Celular , Movimento Celular , Córtex Cerebral/metabolismo , Deleção de Genes , Hipocampo/embriologia , Histona Desacetilase 1 , Histona Desacetilase 2 , Inibidores de Histona Desacetilases , Camundongos , Modelos Biológicos , Neurônios/metabolismo , Fenótipo
11.
Proc Natl Acad Sci U S A ; 106(19): 7751-5, 2009 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-19416910

RESUMO

Histone deacetylase inhibitors (HDACi) represent a new group of drugs currently being tested in a wide variety of clinical applications. They are especially effective in preclinical models of cancer where they show antiproliferative action in many different types of cancer cells. Recently, the first HDACi was approved for the treatment of cutaneous T cell lymphomas. Most HDACi currently in clinical development act by unspecifically interfering with the enzymatic activity of all class I HDACs (HDAC1, 2, 3, and 8), and it is widely believed that the development of isoform-specific HDACi could lead to better therapeutic efficacy. The contribution of the individual class I HDACs to different disease states, however, has so far not been fully elucidated. Here, we use a genetic approach to dissect the involvement of the different class I HDACs in tumor cells. We show that deletion of a single HDAC is not sufficient to induce cell death, but that HDAC1 and 2 play redundant and essential roles in tumor cell survival. Their deletion leads to nuclear bridging, nuclear fragmentation, and mitotic catastrophe, mirroring the effects of HDACi on cancer cells. These findings suggest that pharmacological inhibition of HDAC1 and 2 may be sufficient for anticancer activity, providing an experimental framework for the development of isoform-specific HDAC inhibitors.


Assuntos
Histona Desacetilases/genética , Histona Desacetilases/fisiologia , Animais , Antineoplásicos/farmacologia , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inibidores de Histona Desacetilases , Humanos , Camundongos , Camundongos Nus , Modelos Genéticos , Transplante de Neoplasias , Neoplasias/metabolismo , Isoformas de Proteínas
12.
EBioMedicine ; 85: 104304, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36265417

RESUMO

BACKGROUND: MicroRNAs are non-coding RNAs that negatively regulate gene networks. Previously, we reported that systemically delivered miR-29 mimic MRG-201 reduced fibrosis in animal models, supporting the consideration of miR-29-based therapies for idiopathic pulmonary fibrosis (IPF). METHODS: We generated MRG-229, a next-generation miR-29 mimic based on MRG-201 with improved chemical stability due to additional sugar modifications and conjugation with the internalization moiety BiPPB (PDGFbetaR-specific bicyclic peptide)1. We investigated the anti-fibrotic efficacy of MRG-229 on TGF-ß1 treated human lung fibroblasts (NHLFs), human precision cut lung slices (hPCLS), and in vivo bleomycin studies; toxicology was assessed in two animal models, rats, and non-human primates. Finally, we examined miR-29b levels in a cohort of 46 and 213 patients with IPF diagnosis recruited from Yale and Nottingham Universities (Profile Cohort), respectively. FINDINGS: The peptide-conjugated MRG-229 mimic decreased expression of pro-fibrotic genes and reduced collagen production in each model. In bleomycin-treated mice, the peptide-conjugated MRG-229 mimic downregulated profibrotic gene programs at doses more than ten-fold lower than the original compound. In rats and non-human primates, the peptide-conjugated MRG-229 mimic was well tolerated at clinically relevant doses with no adverse findings observed. In human peripheral blood from IPF patients decreased miR-29 concentrations were associated with increased mortality in two cohorts potentially identified as a target population for treatment. INTERPRETATION: Collectively, our results provide support for the development of the peptide-conjugated MRG-229 mimic as a potential therapy in humans with IPF. FUNDING: This work was supported by NIH NHLBI grants UH3HL123886, R01HL127349, R01HL141852, U01HL145567.


Assuntos
Fibrose Pulmonar Idiopática , MicroRNAs , Humanos , Camundongos , Ratos , Animais , Pulmão/metabolismo , Fibrose Pulmonar Idiopática/diagnóstico , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/terapia , Bleomicina , MicroRNAs/genética , MicroRNAs/metabolismo , Fibroblastos/metabolismo
13.
J Biol Chem ; 285(19): 14663-70, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20190228

RESUMO

Adipocyte differentiation is a well defined process that is under the control of transcriptional activators and repressors. We show that histone deacetylase (HDAC) inhibitors efficiently block adipocyte differentiation in vitro. This effect is specific to adipogenesis, as another mesenchymal differentiation process, osteoblastogenesis, is enhanced upon HDAC inhibition. Through the systematic genetic deletion of HDAC genes in cultured mesenchymal precursor cells, we show that deletion of HDAC1 and HDAC2 leads to reduced lipid accumulation, revealing redundant and requisite roles of these class I HDACs in adipogenesis. These findings unveil a previously unrecognized role for HDACs in the control of adipogenesis.


Assuntos
Adipogenia/fisiologia , Embrião de Mamíferos/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células 3T3-L1 , Adipogenia/efeitos dos fármacos , Animais , Biomarcadores/metabolismo , Western Blotting , Butiratos/farmacologia , Diferenciação Celular , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Perfilação da Expressão Gênica , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/genética , Histona Desacetilase 2/antagonistas & inibidores , Histona Desacetilase 2/genética , Inibidores de Histona Desacetilases/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Osteogênese , PPAR gama/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Clin Invest ; 118(11): 3588-97, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18830415

RESUMO

Histone deacetylase (HDAC) inhibitors show remarkable therapeutic potential for a variety of disorders, including cancer, neurological disease, and cardiac hypertrophy. However, the specific HDAC isoforms that mediate their actions are unclear, as are the physiological and pathological functions of individual HDACs in vivo. To explore the role of Hdac3 in the heart, we generated mice with a conditional Hdac3 null allele. Although global deletion of Hdac3 resulted in lethality by E9.5, mice with a cardiac-specific deletion of Hdac3 survived until 3-4 months of age. At this time, they showed massive cardiac hypertrophy and upregulation of genes associated with fatty acid uptake, fatty acid oxidation, and electron transport/oxidative phosphorylation accompanied by fatty acid-induced myocardial lipid accumulation and elevated triglyceride levels. These abnormalities in cardiac metabolism can be attributed to excessive activity of the nuclear receptor PPARalpha. The phenotype associated with cardiac-specific Hdac3 gene deletion differs from that of all other Hdac gene mutations. These findings reveal a unique role for Hdac3 in maintenance of cardiac function and regulation of myocardial energy metabolism.


Assuntos
Metabolismo Energético/genética , Deleção de Genes , Histona Desacetilases/genética , Miocárdio/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Regulação Enzimológica da Expressão Gênica , Coração , Histona Desacetilases/metabolismo , Histona Desacetilases/fisiologia , Imuno-Histoquímica , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/ultraestrutura , PPAR alfa/metabolismo , Regulação para Cima
15.
J Cardiovasc Pharmacol ; 57(1): 1-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20729755

RESUMO

In the last 10 years it has become increasingly clear that a large class of small noncoding RNAs, known as microRNAs (miRNAs) are potent and crucial regulators of important cellular processes such as differentiation, growth, and survival. miRNAs regulate gene expression through binding to 3' UTRs of target messenger RNAs whereby inducing either messenger RNA degradation or inhibition of protein translation. Although we have only just begun to gain some insight into the biology surrounding miRNAs, their apparent relevance and potency during the onset and progression of disease has generated a lot of interest in assessing the feasibility of therapeutic regulation of miRNAs. As a result of the short RNA nature of miRNAs and lessons learned from small interfering RNA therapeutics and gene therapy, within a timespan of a few years, incredible progress has been made in advancing miRNA regulation into the clinic. We summarize the various therapeutic tools that are currently being investigated to manipulate miRNAs with a special focus on cardiovascular disease and speculate on the future developments of miRNA therapeutics.


Assuntos
Doenças Cardiovasculares/genética , Marcação de Genes , MicroRNAs/genética , Animais , Doenças Cardiovasculares/tratamento farmacológico , Terapia Genética/métodos , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/uso terapêutico , Pequeno RNA não Traduzido/uso terapêutico
16.
J Neurosci ; 29(25): 8288-97, 2009 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-19553468

RESUMO

The structural assembly of synapses can be accomplished in a rapid time frame, although most nascent synapses formed during early development are not fully functional and respond poorly to presynaptic action potentials. The mechanisms that are responsible for this delay in synapse maturation are unknown. Histone deacetylases (HDACs) regulate the activity state of chromatin and repress gene expression through the removal of acetyl groups from histones. Class I HDACs, which include HDAC1 and HDAC2, are expressed in the CNS, although their specific role in neuronal function has not been studied. To delineate the contribution of HDAC1 and HDAC2 in the brain, we have used pharmacological inhibitors of HDACs and mice with conditional alleles to HDAC1 and HDAC2. We found that a decrease in the activities of both HDAC1 and HDAC2 during early synaptic development causes a robust facilitation of excitatory synapse maturation and a modest increase in synapse numbers. In contrast, in mature neurons a decrease in HDAC2 levels alone was sufficient to attenuate basal excitatory neurotransmission without a significant change in the numbers of detectable nerve terminals. Therefore, we propose that HDAC1 and HDAC2 form a developmental switch that controls synapse maturation and function acting in a manner dependent on the maturational states of neuronal networks.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipocampo/crescimento & desenvolvimento , Histona Desacetilases/fisiologia , Neurônios/fisiologia , Proteínas Repressoras/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Animais , Animais Recém-Nascidos , Western Blotting , Células Cultivadas , Eletrofisiologia , Fluorescência , Hipocampo/metabolismo , Hipocampo/fisiologia , Histona Desacetilase 1 , Histona Desacetilase 2 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Técnicas de Patch-Clamp , Plasmídeos , RNA Mensageiro , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sinapses/metabolismo , Transmissão Sináptica/genética , Transfecção
17.
J Neurosci ; 28(29): 7344-9, 2008 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-18632938

RESUMO

The molecular mechanisms underlying the transition from recreational drug use to chronic addiction remain poorly understood. One molecule implicated in this process is DeltaFosB, a transcription factor that accumulates in striatum after repeated drug exposure and mediates sensitized behavioral responses to psychostimulants and other drugs of abuse. The downstream transcriptional mechanisms by which DeltaFosB regulates drug-induced behaviors are incompletely understood. We reported previously the chromatin remodeling mechanisms by which DeltaFosB activates the expression of certain genes; however, the mechanisms underlying DeltaFosB-mediated gene repression remain unknown. Here, we identify c-fos, an immediate early gene rapidly induced in striatum after acute psychostimulant exposure, as a novel downstream target that is repressed chronically by DeltaFosB. We show that accumulation of DeltaFosB in striatum after chronic amphetamine treatment desensitizes c-fos mRNA induction to a subsequent drug dose. DeltaFosB desensitizes c-fos expression by recruiting histone deacetylase 1 (HDAC1) to the c-fos gene promoter, which, in turn, deacetylates surrounding histones and attenuates gene activity. Accordingly, local knock-out of HDAC1 in striatum abolishes amphetamine-induced desensitization of the c-fos gene. In concert, chronic amphetamine increases histone H3 methylation on the c-fos promoter, a chromatin modification also known to repress gene activity, as well as expression levels of the H3 histone methyltransferase, KMT1A (lysine methyltransferase 1A, formerly SUV39H1). This study reveals a novel epigenetic pathway through which DeltaFosB mediates distinct transcriptional programs that may ultimately alter behavioral plasticity to chronic amphetamine exposure.


Assuntos
Anfetamina/administração & dosagem , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteínas Proto-Oncogênicas c-fos/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Esquema de Medicação , Epigênese Genética/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Histona Desacetilase 1 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Camundongos , Camundongos Transgênicos , Células PC12 , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Proteínas Proto-Oncogênicas c-fos/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
18.
EMBO Mol Med ; 8(6): 643-53, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27137489

RESUMO

Abnormal remodeling of atherosclerotic plaques can lead to rupture, acute myocardial infarction, and death. Enhancement of plaque extracellular matrix (ECM) may improve plaque morphology and stabilize lesions. Here, we demonstrate that chronic administration of LNA-miR-29 into an atherosclerotic mouse model improves indices of plaque morphology. This occurs due to upregulation of miR-29 target genes of the ECM (col1A and col3A) resulting in reduced lesion size, enhanced fibrous cap thickness, and reduced necrotic zones. Sustained LNA-miR-29 treatment did not affect circulating lipids, blood chemistry, or ECM of solid organs including liver, lung, kidney, spleen, or heart. Collectively, these data support the idea that antagonizing miR-29 may promote beneficial plaque remodeling as an independent approach to stabilize vulnerable atherosclerotic lesions.


Assuntos
Aterosclerose/patologia , MicroRNAs/antagonistas & inibidores , Animais , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Colágeno Tipo III/metabolismo , Modelos Animais de Doenças , Camundongos
19.
EMBO Mol Med ; 6(10): 1347-56, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25239947

RESUMO

Over the last decade, great enthusiasm has evolved for microRNA (miRNA) therapeutics. Part of the excitement stems from the fact that a miRNA often regulates numerous related mRNAs. As such, modulation of a single miRNA allows for parallel regulation of multiple genes involved in a particular disease. While many studies have shown therapeutic efficacy using miRNA inhibitors, efforts to restore or increase the function of a miRNA have been lagging behind. The miR-29 family has gained a lot of attention for its clear function in tissue fibrosis. This fibroblast-enriched miRNA family is downregulated in fibrotic diseases which induces a coordinate increase of many extracellular matrix genes. Here, we show that intravenous injection of synthetic RNA duplexes can increase miR-29 levels in vivo for several days. Moreover, therapeutic delivery of these miR-29 mimics during bleomycin-induced pulmonary fibrosis restores endogenous miR-29 function whereby decreasing collagen expression and blocking and reversing pulmonary fibrosis. Our data support the feasibility of using miRNA mimics to therapeutically increase miRNAs and indicate miR-29 to be a potent therapeutic miRNA for treating pulmonary fibrosis.


Assuntos
MicroRNAs/genética , Mimetismo Molecular/genética , Fibrose Pulmonar/genética , Animais , Bleomicina , Northern Blotting , Linhagem Celular , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Mimetismo Molecular/fisiologia , Células NIH 3T3 , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/fisiopatologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Eur J Heart Fail ; 15(6): 650-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23388090

RESUMO

AIMS: Recent studies have shown that microRNAs (miRNAs), besides being potent regulators of gene expression, can additionally serve as circulating biomarkers of disease. The aim of this study is to determine if plasma miRNAs can be used as indicators of disease progression or therapeutic efficacy in hypertension-induced heart disease. METHODS AND RESULTS: In order to define circulating miRNAs that change during hypertension-induced heart failure and that respond to therapeutic treatment, we performed miRNA arrays on plasma RNA from hypertensive rats that show signs of heart failure. Array analysis indicated that approximately one-third of the miRNAs on the array are detectable in plasma. Quantitative real-time polymerase chain reaction (PCR) analysis for a selected panel of miRNAs indicated that circulating levels of miR-16, miR-20b, miR-93, miR-106b, miR-223, and miR-423-5p were significantly increased in response to hypertension-induced heart failure, while this effect was blunted in response to treatment with antimiR-208a as well as an ACE inhibitor. Moreover, treatment with antimiR-208a resulted in a dramatic increase in one miRNA, miR-19b. A time course study indicated that several of these miRNA changes track with disease progression. CONCLUSIONS: Circulating levels of miRNAs are responsive to therapeutic interventions and change during the progression of hypertension-induced heart disease.


Assuntos
Biomarcadores/sangue , Progressão da Doença , Insuficiência Cardíaca/tratamento farmacológico , Hipertensão/tratamento farmacológico , MicroRNAs/sangue , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Captopril/uso terapêutico , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/diagnóstico , Hipertensão/induzido quimicamente , Hipertensão/diagnóstico , Masculino , MicroRNAs/genética , Peptídeo Natriurético Encefálico/sangue , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Endogâmicos Dahl , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Cloreto de Sódio/toxicidade , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA