Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Nature ; 608(7921): 181-191, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35732239

RESUMO

The heart, the first organ to develop in the embryo, undergoes complex morphogenesis that when defective results in congenital heart disease (CHD). With current therapies, more than 90% of patients with CHD survive into adulthood, but many suffer premature death from heart failure and non-cardiac causes1. Here, to gain insight into this disease progression, we performed single-nucleus RNA sequencing on 157,273 nuclei from control hearts and hearts from patients with CHD, including those with hypoplastic left heart syndrome (HLHS) and tetralogy of Fallot, two common forms of cyanotic CHD lesions, as well as dilated and hypertrophic cardiomyopathies. We observed CHD-specific cell states in cardiomyocytes, which showed evidence of insulin resistance and increased expression of genes associated with FOXO signalling and CRIM1. Cardiac fibroblasts in HLHS were enriched in a low-Hippo and high-YAP cell state characteristic of activated cardiac fibroblasts. Imaging mass cytometry uncovered a spatially resolved perivascular microenvironment consistent with an immunodeficient state in CHD. Peripheral immune cell profiling suggested deficient monocytic immunity in CHD, in agreement with the predilection in CHD to infection and cancer2. Our comprehensive phenotyping of CHD provides a roadmap towards future personalized treatments for CHD.


Assuntos
Cardiopatias Congênitas , Fenótipo , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/imunologia , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/imunologia , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/patologia , Progressão da Doença , Fibroblastos/metabolismo , Fibroblastos/patologia , Fatores de Transcrição Forkhead/metabolismo , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/imunologia , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/patologia , Humanos , Síndrome do Coração Esquerdo Hipoplásico/genética , Síndrome do Coração Esquerdo Hipoplásico/imunologia , Síndrome do Coração Esquerdo Hipoplásico/metabolismo , Síndrome do Coração Esquerdo Hipoplásico/patologia , Citometria por Imagem , Resistência à Insulina , Monócitos/imunologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , RNA-Seq , Transdução de Sinais/genética , Análise de Célula Única , Tetralogia de Fallot/genética , Tetralogia de Fallot/imunologia , Tetralogia de Fallot/metabolismo , Tetralogia de Fallot/patologia , Proteínas de Sinalização YAP/metabolismo
2.
Nature ; 547(7662): 227-231, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28581498

RESUMO

The regenerative capacity of the adult mammalian heart is limited, because of the reduced ability of cardiomyocytes to progress through mitosis. Endogenous cardiomyocytes have regenerative capacity at birth but this capacity is lost postnatally, with subsequent organ growth occurring through cardiomyocyte hypertrophy. The Hippo pathway, a conserved kinase cascade, inhibits cardiomyocyte proliferation in the developing heart to control heart size and prevents regeneration in the adult heart. The dystrophin-glycoprotein complex (DGC), a multicomponent transmembrane complex linking the actin cytoskeleton to extracellular matrix, is essential for cardiomyocyte homeostasis. DGC deficiency in humans results in muscular dystrophy, including the lethal Duchenne muscular dystrophy. Here we show that the DGC component dystroglycan 1 (Dag1) directly binds to the Hippo pathway effector Yap to inhibit cardiomyocyte proliferation in mice. The Yap-Dag1 interaction was enhanced by Hippo-induced Yap phosphorylation, revealing a connection between Hippo pathway function and the DGC. After injury, Hippo-deficient postnatal mouse hearts maintained organ size control by repairing the defect with correct dimensions, whereas postnatal hearts deficient in both Hippo and the DGC showed cardiomyocyte overproliferation at the injury site. In the hearts of mature Mdx mice (which have a point mutation in Dmd)-a model of Duchenne muscular dystrophy-Hippo deficiency protected against overload-induced heart failure.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Distrofina/metabolismo , Glicoproteínas/metabolismo , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Miócitos Cardíacos/citologia , Fosfoproteínas/metabolismo , Animais , Cardiomiopatias , Proteínas de Ciclo Celular , Proliferação de Células , Distroglicanas/metabolismo , Distrofina/deficiência , Distrofina/genética , Glicoproteínas/deficiência , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/prevenção & controle , Via de Sinalização Hippo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Complexos Multiproteicos/deficiência , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Miócitos Cardíacos/metabolismo , Tamanho do Órgão , Fosforilação , Pressão , Ligação Proteica , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Sinalização YAP
3.
Nature ; 550(7675): 260-264, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-28976966

RESUMO

Mammalian organs vary widely in regenerative capacity. Poorly regenerative organs, such as the heart are particularly vulnerable to organ failure. Once established, heart failure commonly results in mortality. The Hippo pathway, a kinase cascade that prevents adult cardiomyocyte proliferation and regeneration, is upregulated in human heart failure. Here we show that deletion of the Hippo pathway component Salvador (Salv) in mouse hearts with established ischaemic heart failure after myocardial infarction induces a reparative genetic program with increased scar border vascularity, reduced fibrosis, and recovery of pumping function compared with controls. Using translating ribosomal affinity purification, we isolate cardiomyocyte-specific translating messenger RNA. Hippo-deficient cardiomyocytes have increased expression of proliferative genes and stress response genes, such as the mitochondrial quality control gene, Park2. Genetic studies indicate that Park2 is essential for heart repair, suggesting a requirement for mitochondrial quality control in regenerating myocardium. Gene therapy with a virus encoding Salv short hairpin RNA improves heart function when delivered at the time of infarct or after ischaemic heart failure following myocardial infarction was established. Our findings indicate that the failing heart has a previously unrecognized reparative capacity involving more than cardiomyocyte renewal.


Assuntos
Proteínas de Ciclo Celular/deficiência , Insuficiência Cardíaca Sistólica/metabolismo , Insuficiência Cardíaca Sistólica/terapia , Infarto do Miocárdio/complicações , Proteínas Serina-Treonina Quinases/deficiência , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Terapia Genética , Insuficiência Cardíaca Sistólica/etiologia , Insuficiência Cardíaca Sistólica/patologia , Via de Sinalização Hippo , Humanos , Camundongos , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Controle de Qualidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Ubiquitina-Proteína Ligases/genética
4.
Nature ; 534(7605): 119-23, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27251288

RESUMO

Myocardial infarction results in compromised myocardial function and heart failure owing to insufficient cardiomyocyte self-renewal. Unlike many vertebrates, mammalian hearts have only a transient neonatal renewal capacity. Reactivating primitive reparative ability in the mature mammalian heart requires knowledge of the mechanisms that promote early heart repair. By testing an established Hippo-deficient heart regeneration mouse model for factors that promote renewal, here we show that the expression of Pitx2 is induced in injured, Hippo-deficient ventricles. Pitx2-deficient neonatal mouse hearts failed to repair after apex resection, whereas adult mouse cardiomyocytes with Pitx2 gain-of-function efficiently regenerated after myocardial infarction. Genomic analyses indicated that Pitx2 activated genes encoding electron transport chain components and reactive oxygen species scavengers. A subset of Pitx2 target genes was cooperatively regulated with the Hippo pathway effector Yap. Furthermore, Nrf2, a regulator of the antioxidant response, directly regulated the expression and subcellular localization of Pitx2. Pitx2 mutant myocardium had increased levels of reactive oxygen species, while antioxidant supplementation suppressed the Pitx2 loss-of-function phenotype. These findings reveal a genetic pathway activated by tissue damage that is essential for cardiac repair.


Assuntos
Antioxidantes/metabolismo , Traumatismos Cardíacos/metabolismo , Proteínas de Homeodomínio/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Regeneração/fisiologia , Fatores de Transcrição/metabolismo , Cicatrização/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Animais Recém-Nascidos , Antioxidantes/farmacologia , Proteínas de Ciclo Celular , Modelos Animais de Doenças , Transporte de Elétrons/efeitos dos fármacos , Transporte de Elétrons/genética , Feminino , Sequestradores de Radicais Livres/metabolismo , Traumatismos Cardíacos/genética , Traumatismos Cardíacos/patologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Via de Sinalização Hippo , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Espécies Reativas de Oxigênio/metabolismo , Regeneração/efeitos dos fármacos , Regeneração/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Cicatrização/efeitos dos fármacos , Cicatrização/genética , Proteínas de Sinalização YAP , Proteína Homeobox PITX2
5.
Development ; 145(18)2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30143541

RESUMO

Loss of the paired-like homeodomain transcription factor 2 (Pitx2) in cardiomyocytes predisposes mice to atrial fibrillation and compromises neonatal regenerative capacity. In addition, Pitx2 gain-of-function protects mature cardiomyocytes from ischemic injury and promotes heart repair. Here, we characterized the long-term myocardial phenotype following myocardial infarction (MI) in Pitx2 conditional-knockout (Pitx2 CKO) mice. We found adipose-like tissue in Pitx2 CKO hearts 60 days after MI induced surgically at postnatal day 2 but not at day 8. Molecular and cellular analyses showed the onset of adipogenic signaling in mutant hearts after MI. Lineage tracing experiments showed a non-cardiomyocyte origin of the de novo adipose-like tissue. Interestingly, we found that Pitx2 promotes mitochondrial function through its gene regulatory network, and that the knockdown of a key mitochondrial Pitx2 target gene, Cox7c, also leads to the accumulation of myocardial fat tissue. Single-nuclei RNA-seq revealed that Pitx2-deficient hearts were oxidatively stressed. Our findings reveal a role for Pitx2 in maintaining proper cardiac cellular composition during heart regeneration via the maintenance of proper mitochondrial structure and function.


Assuntos
Adipogenia/fisiologia , Proteínas de Homeodomínio/metabolismo , Mitocôndrias/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Regeneração/fisiologia , Fatores de Transcrição/metabolismo , Tecido Adiposo/patologia , Animais , Linhagem Celular , Complexo IV da Cadeia de Transporte de Elétrons/genética , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Infarto do Miocárdio/genética , Estresse Oxidativo/genética , Regeneração/genética , Fatores de Transcrição/genética , Proteína Homeobox PITX2
6.
Development ; 140(16): 3395-402, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23863481

RESUMO

Congenital heart disease (CHD) is a devastating anomaly that affects ∼1% of live births. Defects of the outflow tract (OFT) make up a large percentage of human CHD. We investigated Bmp signaling in mouse OFT development by conditionally deleting both Bmp4 and Bmp7 in the second heart field (SHF). SHF Bmp4/7 deficiency resulted in defective epithelial to mesenchymal transition (EMT) and reduced cardiac neural crest ingress, with resultant persistent truncus arteriosus. Using a candidate gene approach, we found that Vegfa was upregulated in the Bmp4/7 mutant hearts. To determine if Vegfa is a downstream Bmp effector during EMT, we examined whether Vegfa is transcriptionally regulated by the Bmp receptor-regulated Smad. Our findings indicate that Smad directly binds to Vegfa chromatin and represses Vegfa transcriptional activity. We also found that Vegfa is a direct target for the miR-17-92 cluster, which is also regulated by Bmp signaling in the SHF. Deletion of miR-17-92 reveals similar phenotypes to Bmp4/7 SHF deletion. To directly address the function of Vegfa repression in Bmp-mediated EMT, we performed ex vivo explant cultures from Bmp4/7 and miR-17-92 mutant hearts. EMT was defective in explants from the Bmp4/7 double conditional knockout (dCKO; Mef2c-Cre;Bmp4/7(f/f)) and miR-17-92 null. By antagonizing Vegfa activity in explants, EMT was rescued in Bmp4/7 dCKO and miR-17-92 null culture. Moreover, overexpression of miR-17-92 partially suppressed the EMT defect in Bmp4/7 mutant embryos. Our study reveals that Vegfa levels in the OFT are tightly controlled by Smad- and microRNA-dependent pathways to modulate OFT development.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Obstrução do Fluxo Ventricular Externo/patologia , Animais , Sequência de Bases , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/metabolismo , Células Cultivadas , Embrião de Mamíferos/metabolismo , Transição Epitelial-Mesenquimal , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Dados de Sequência Molecular , Mutação , Miocárdio/metabolismo , Miocárdio/patologia , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteína Smad1/genética , Proteína Smad1/metabolismo , Transcrição Gênica , Persistência do Tronco Arterial/metabolismo , Persistência do Tronco Arterial/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Obstrução do Fluxo Ventricular Externo/metabolismo
7.
Development ; 140(23): 4683-90, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24255096

RESUMO

Heart failure due to cardiomyocyte loss after ischemic heart disease is the leading cause of death in the United States in large part because heart muscle regenerates poorly. The endogenous mechanisms preventing mammalian cardiomyocyte regeneration are poorly understood. Hippo signaling, an ancient organ size control pathway, is a kinase cascade that inhibits developing cardiomyocyte proliferation but it has not been studied postnatally or in fully mature adult cardiomyocytes. Here, we investigated Hippo signaling in adult cardiomyocyte renewal and regeneration. We found that unstressed Hippo-deficient adult mouse cardiomyocytes re-enter the cell cycle and undergo cytokinesis. Moreover, Hippo deficiency enhances cardiomyocyte regeneration with functional recovery after adult myocardial infarction as well as after postnatal day eight (P8) cardiac apex resection and P8 myocardial infarction. In damaged hearts, Hippo mutant cardiomyocytes also have elevated proliferation. Our findings reveal that Hippo signaling is an endogenous repressor of adult cardiomyocyte renewal and regeneration. Targeting the Hippo pathway in human disease might be beneficial for the treatment of heart disease.


Assuntos
Coração/fisiologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Regeneração/fisiologia , Animais , Ciclo Celular , Linhagem Celular , Proliferação de Células , Via de Sinalização Hippo , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio , Miócitos Cardíacos/citologia , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais
8.
J Pharmacol Exp Ther ; 358(3): 548-57, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27384074

RESUMO

The present study was designed to assess both preventive and therapeutic effects of (S)-1-(2-Hydroxyethyl)-4-methyl-N-[4-(methylsulfonyl) phenyl]-5-[2-(trifluoromethyl) phenyl]-1H-pyrrole-3-carboxamide (CS-3150), a novel nonsteroidal mineralocorticoid receptor antagonist, on renal injury in deoxycorticosterone acetate (DOCA)/salt-induced hypertensive rats (DOCA rats). From 7 weeks of age, DOCA was subcutaneously administered once a week for 4 weeks to uninephrectomized rats fed a high-salt diet. In experiment 1, CS-3150 (0.3-3 mg/kg) was orally administered once a day for 4 weeks coincident with DOCA administration. In experiment 2, after establishment of renal injury by 4 weeks of DOCA/salt loading, CS-3150 (3 mg/kg) was orally administered once a day for 4 weeks with or without continuous DOCA administration. In experiment 1, DOCA/salt loading significantly increased systolic blood pressure (SBP), which was prevented by CS-3150 in a dose-dependent manner. Development of renal injury (proteinuria, renal hypertrophy, and histopathological changes in glomeruli and tubule) was also suppressed by CS-3150 with inhibition of mRNA expression of fibrosis, inflammation, and oxidative stress markers. In experiment 2, under continuous DOCA treatment, CS-3150 clearly ameliorated existing renal injury without lowering SBP, indicating that CS-3150 regressed renal injury independent of its antihypertensive action. Moreover, CS-3150 treatment in combination with withdrawal of DOCA showed further therapeutic effect on renal injury accompanied by reduction in SBP. These results demonstrate that CS-3150 not only prevents but also ameliorates hypertension and renal injury in DOCA rats. Therefore, CS-3150 could be a promising agent for the treatment of hypertension and renal disorders, and may have potential to promote regression of renal injury.


Assuntos
Acetato de Desoxicorticosterona/efeitos adversos , Hipertensão/induzido quimicamente , Hipertensão/prevenção & controle , Rim/efeitos dos fármacos , Pirróis/farmacologia , Receptores de Mineralocorticoides/metabolismo , Cloreto de Sódio na Dieta/efeitos adversos , Sulfonas/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hipertensão/metabolismo , Hipertensão/patologia , Rim/lesões , Rim/metabolismo , Rim/patologia , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Tamanho do Órgão/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos
9.
Life Sci Alliance ; 7(2)2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38012001

RESUMO

Modulation of the heart's immune microenvironment is crucial for recovery after ischemic events such as myocardial infarction (MI). Endothelial cells (ECs) can have immune regulatory functions; however, interactions between ECs and the immune environment in the heart after MI remain poorly understood. We identified an EC-specific IFN responsive and immune regulatory gene signature in adult and pediatric heart failure (HF) tissues. Single-cell transcriptomic analysis of murine hearts subjected to MI uncovered an EC population (IFN-ECs) with immunologic gene signatures similar to those in human HF. IFN-ECs were enriched in regenerative-stage mouse hearts and expressed genes encoding immune responsive transcription factors (Irf7, Batf2, and Stat1). Single-cell chromatin accessibility studies revealed an enrichment of these TF motifs at IFN-EC signature genes. Expression of immune regulatory ligand genes by IFN-ECs suggests bidirectional signaling between IFN-ECs and macrophages in regenerative-stage hearts. Our data suggest that ECs may adopt immune regulatory signatures after cardiac injury to accompany the reparative response. The presence of these signatures in human HF and murine MI models suggests a potential role for EC-mediated immune regulation in responding to stress induced by acute injury in MI and chronic adverse remodeling in HF.


Assuntos
Insuficiência Cardíaca , Infarto do Miocárdio , Camundongos , Humanos , Animais , Criança , Células Endoteliais/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Coração , Transdução de Sinais/genética
10.
Nat Cardiovasc Res ; 3(3): 283-300, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38510108

RESUMO

After myocardial infarction (MI), mammalian hearts do not regenerate, and the microenvironment is disrupted. Hippo signaling loss of function with activation of transcriptional co-factor YAP induces heart renewal and rebuilds the post-MI microenvironment. In this study, we investigated adult renewal-competent mouse hearts expressing an active version of YAP, called YAP5SA, in cardiomyocytes (CMs). Spatial transcriptomics and single-cell RNA sequencing revealed a conserved, renewal-competent CM cell state called adult (a)CM2 with high YAP activity. aCM2 co-localized with cardiac fibroblasts (CFs) expressing complement pathway component C3 and macrophages (MPs) expressing C3ar1 receptor to form a cellular triad in YAP5SA hearts and renewal-competent neonatal hearts. Although aCM2 was detected in adult mouse and human hearts, the cellular triad failed to co-localize in these non-renewing hearts. C3 and C3ar1 loss-of-function experiments indicated that C3a signaling between MPs and CFs was required to assemble the pro-renewal aCM2, C3+ CF and C3ar1+ MP cellular triad.

11.
Circ Res ; 108(8): 940-9, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21350214

RESUMO

RATIONALE: The basic helix-loop-helix (bHLH) transcription factors Hand1 and Hand2 are essential for embryonic development. Given their requirement for cardiogenesis, it is imperative to determine their impact on cardiovascular function. OBJECTIVE: To deduce the role of Hand2 within the epicardium. METHOD AND RESULTS: We engineered a Hand1 allele expressing Cre recombinase. Cardiac Hand1 expression is largely limited to cells of the primary heart field, overlapping little with Hand2 expression. Hand1 is expressed within the septum transversum, and the Hand1 lineage marks the proepicardial organ and epicardium. To examine Hand factor functional overlap, we conditionally deleted Hand2 from Hand1-expressing cells. Hand2 mutants display defective epicardialization and fail to form coronary arteries, coincident with altered extracellular matrix deposition and Pdgfr expression. CONCLUSIONS: These data demonstrate a hierarchal relationship whereby transient Hand1 septum transversum expression defines epicardial precursors that are subsequently dependent on Hand2 function.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Vasos Coronários/embriologia , Vasos Coronários/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Pericárdio/embriologia , Pericárdio/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem da Célula/genética , Células Cultivadas , Vasos Coronários/citologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pericárdio/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/patologia
12.
Gastroenterology ; 141(2): 576-87, 587.e1-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21669203

RESUMO

BACKGROUND & AIMS: Hand2 is a basic helix-loop-helix transcription factor required for terminal differentiation of enteric neurons. We studied Hand2 haploinsufficient mice, to determine whether reduced expression of Hand2 allows sufficient enteric neurogenesis for survival, but not for development of a normal enteric nervous system (ENS). METHODS: Enteric transcripts that encode Hand2 and the neuron-specific embryonic lethal abnormal vision proteins HuB, HuC, and HuD were quantified. Immunocytochemistry was used to identify and quantify neurons. Apoptosis was analyzed with the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling procedure. Intracellular microelectrodes were used to record inhibitory junction potentials. Gastrointestinal transit and colonic motility were measured in vivo. RESULTS: Levels of of enteric Hand2 transcripts were associated with genotypes of mice, in the following order: Hand2(+/+) > Hand2(LoxP/+) > Hand2(+/-) > Hand2(LoxP/-). Parallel reductions were found in expression of HuD and in regional and phenotypic manners. Numbers of neurons, numbers of neuronal nitric oxide synthase(+) and calretinin(+), but not substance P(+) or vasoactive intestinal peptide(+) neurons, decreased. No effects were observed in stomach or cecum. Apoptosis was not detected, consistent with the concept that Hand2 inhibits neuronal differentiation, rather than regulates survival. The amplitude of inhibitory junction potentials in colonic circular muscle was similar in Hand2 wild-type and haploinsufficient mice, although in haploinsufficient mice, the purinergic component was reduced and a nitrergic component appeared. The abnormal ENS of haploinsufficient mice slowed gastrointestinal motility but protected mice against colitis. CONCLUSIONS: Reduced expression of factors required for development of the ENS can cause defects in the ENS that are subtle enough to escape detection yet cause significant abnormalities in bowel function.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sistema Nervoso Entérico/citologia , Motilidade Gastrointestinal/fisiologia , Neurônios/citologia , Neurônios/metabolismo , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Calbindina 2 , Contagem de Células , Colite/induzido quimicamente , Colite/prevenção & controle , Colo/inervação , Colo/metabolismo , Colo/fisiologia , Proteínas ELAV/metabolismo , Proteína Semelhante a ELAV 2 , Proteína Semelhante a ELAV 3 , Proteína Semelhante a ELAV 4 , Sistema Nervoso Entérico/crescimento & desenvolvimento , Motilidade Gastrointestinal/genética , Genótipo , Junções Intercelulares/fisiologia , Camundongos , Músculo Liso/inervação , Músculo Liso/fisiologia , Neuroglia/citologia , Óxido Nítrico Sintase/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo , Substância P/metabolismo , Transmissão Sináptica/fisiologia , Ácido Trinitrobenzenossulfônico , Peptídeo Intestinal Vasoativo/metabolismo
13.
Development ; 136(21): 3575-84, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19793887

RESUMO

Induction of the sympathetic nervous system (SNS) from its neural crest (NC) precursors is dependent on BMP signaling from the dorsal aorta. To determine the roles of BMP signaling and the pathways involved in SNS development, we conditionally knocked out components of the BMP pathways. To determine if BMP signaling is a cell-autonomous requirement of SNS development, the Alk3 (BMP receptor IA) was deleted in the NC lineage. The loss of Alk3 does not prevent NC cell migration, but the cells die immediately after reaching the dorsal aorta. The paired homeodomain factor Phox2b, known to be essential for survival of SNS precursors, is downregulated, suggesting that Phox2b is a target of BMP signaling. To determine if Alk3 signals through the canonical BMP pathway, Smad4 was deleted in the NC lineage. Loss of Smad4 does not affect neurogenesis and ganglia formation; however, proliferation and noradrenergic differentiation are reduced. Analysis of transcription factors regulating SNS development shows that the basic helix-loop-helix factor Ascl1 is downregulated by loss of Smad4 and that Ascl1 regulates SNS proliferation but not noradrenergic differentiation. To determine if the BMP-activated Tak1 (Map3k7) pathway plays a role in SNS development, Tak1 was deleted in the NC lineage. We show that Tak1 is not involved in SNS development. Taken together, our results suggest multiple roles for BMP signaling during SNS development. The Smad4-independent pathway acts through the activation of Phox2b to regulate survival of SNS precursors, whereas the Smad4-dependent pathway controls noradrenergic differentiation and regulates proliferation by maintaining Ascl1 expression.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Neurogênese , Transdução de Sinais , Proteína Smad4/metabolismo , Sistema Nervoso Simpático/embriologia , Sobrevivência Celular , Embrião de Mamíferos/metabolismo , Norepinefrina/metabolismo , Sistema Nervoso Simpático/metabolismo
14.
Dev Biol ; 340(2): 459-67, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20144605

RESUMO

The neural crest (NC) lineage gives rise to a wide array of cell types ranging from neurons and glia of the peripheral nervous system to skeletal elements of the head. The mechanisms regulating NC differentiation into such a large number of cell types remain largely unknown. MicroRNAs (miRNAs) play key roles in regulating developmental events suggesting they may also play a role during NC differentiation. To determine what roles miRNAs play in differentiation of NC-derived tissues, we deleted the miRNA processing gene Dicer in NC cells using the Wnt1-Cre deleter line. We show that deletion of Dicer soon after NC cells have formed does not affect their migration and colonization of their targets in the embryo. However, the post-migratory NC is dependent on Dicer for survival. In the head, loss of Dicer leads to a loss of NC-derived craniofacial bones while in the trunk, cells of the enteric, sensory and sympathetic nervous systems are lost during development. We found that loss of Dicer does not prevent the initial differentiation of NC but as development progresses, NC derivatives are lost due to apoptotic cell death. When Dicer is deleted, both Caspase-dependent and -independent apoptotic pathways are activated in the sensory ganglia but only the Caspase-dependent apoptotic program was activated in the sympathetic nervous system showing that the specific endogenous apoptotic programs are turned on by loss of Dicer. Our results show that Dicer and miRNAs, are required for survival of NC-derived tissues by preventing apoptosis during differentiation.


Assuntos
RNA Helicases DEAD-box/metabolismo , Endorribonucleases/metabolismo , Crista Neural/citologia , Crista Neural/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Sobrevivência Celular , Cruzamentos Genéticos , RNA Helicases DEAD-box/genética , Embrião de Mamíferos/metabolismo , Endorribonucleases/genética , Feminino , Corantes Fluorescentes/metabolismo , Histocitoquímica , Imuno-Histoquímica , Indóis/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Ribonuclease III , beta-Galactosidase/metabolismo
15.
Genesis ; 48(8): 479-84, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20506548

RESUMO

Hand1 regulates development of numerous tissues within the embryo, extraembryonic mesoderm, and trophectoderm. Systemic loss of Hand1 results in early embryonic lethality but the cause has remained unknown. To determine if Hand1 expression in extraembryonic mesoderm is essential for embryonic survival, Hand1 was conditionally deleted using the HoxB6-Cre mouse line that expresses Cre in extraembryonic and lateral mesoderm. Deletion of Hand1 using HoxB6-Cre resulted in embryonic lethality identical to systemic knockout. To determine if lethality is due to Hand1 function in extraembryonic mesoderm or lateral mesoderm, we generated a Tlx2-Cre mouse line expressing Cre in lateral mesoderm but not extraembryonic tissues. Deletion of Hand1 using the Tlx2-Cre line results in embryonic survival with embryos exhibiting herniated gut and thin enteric smooth muscle. Our results show that Hand1 regulates development of lateral mesoderm derivatives and its loss in extraembryonic mesoderm is the primary cause of lethality in Hand1-null embryos.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Embrião de Mamíferos/embriologia , Proteínas de Homeodomínio/metabolismo , Mesoderma/embriologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Embrião de Mamíferos/metabolismo , Feminino , Trato Gastrointestinal/anormalidades , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Integrases/genética , Integrases/metabolismo , Óperon Lac/genética , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , beta-Galactosidase/metabolismo
16.
Dev Biol ; 330(1): 131-41, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19341725

RESUMO

The basic helix-loop-helix (bHLH) transcription factor Hand2 has been implicated in the development of multiple organs, including craniofacial organs. Mice carrying Hand2 hypomorphic alleles (Hand2(LoxP/-)) display a cleft palate phenotype. A specific deletion of the Hand2 branchial arch-specific enhancer also leads to a hypoplastic mandible and cleft palate formation in mice. However, the underlying mechanism of Hand2 regulation of palate development remains unknown. Here we show that Hand2 is expressed in both the epithelium and mesenchyme of the developing palate. While mesenchymal specific inactivation of Hand2 has no impact on palate development, epithelial specific deletion of Hand2 creates a cleft palate phenotype. Hand2 appears to exert distinct roles in the anterior and posterior palate. In the anterior palate of Hand2(LoxP/-) mice, premature death of periderm cells and a down-regulation of Shh are observed in the medial edge epithelium (MEE), accompanied by a decreased level of cell proliferation in the palatal mesenchyme. In the posterior palate, a lower dose of Hand2 causes aberrant periderm cell death on the surface of the epithelium, triggering abnormal fusion between the palatal shelf and mandible and preventing palatal shelf elevation. We further demonstrate that BMP activities are essential for the expression of Hand2 in the palate. We conclude that Hand2 is an intrinsic regulator in the epithelium and is required for palate development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Epitélio/metabolismo , Palato/embriologia , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proliferação de Células , Epitélio/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Palato/citologia , Palato/metabolismo , Transdução de Sinais
17.
Circ Res ; 103(12): 1422-9, 2008 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-19008477

RESUMO

The cardiac neural crest (cNC) lineage plays key roles in heart development by directly contributing to heart structures and regulating development of other heart lineages. The basic helix-loop-helix factor Hand2 regulates development of cardiovascular structures and NC-derived tissues including those that contribute to face and peripheral nervous system. Although Hand2 is expressed in cNC, its role has not been examined because of an early embryonic lethality when Hand2 is deleted in the NC lineage. We find that the lethality is attributable to loss of norepinephrine synthesis that can be overcome by activating adrenergic receptors. In rescued embryos, loss of Hand2 in the NC lineage leads to the misalignment of the outflow tract and aortic arch arteries. Defects include pulmonary stenosis, interrupted aortic artery, retroesophageal right subclavian artery, and ventricular septum defect, which resemble congenital heart defects attributed to defects in the NC. Hand2 functions in part by regulating signaling from the cNC to other cardiac lineages but not by regulating migration or survival of the cNC. Loss of Hand2 in NC also uncovered a novel role for the cNC in regulating proliferation and differentiation of the second heart field-derived myocardium that persists late into development. These results show that the cNC functions as a major signaling center for heart development and Hand2 plays a pivotal role in regulating both cell-autonomous and -nonautonomous functions of the cNC.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Coração/embriologia , Crista Neural/embriologia , Crista Neural/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Feminino , Coração/crescimento & desenvolvimento , Cardiopatias Congênitas/etiologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Gravidez
19.
Hypertens Res ; 43(11): 1204-1213, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32616846

RESUMO

Esaxerenone is a novel selective mineralocorticoid receptor (MR) blocker that was recently approved in Japan to treat hypertension. In phase II and III studies, esaxerenone plus a renin-angiotensin system inhibitor markedly reduced the urinary albumin-to-creatinine ratio (UACR) in hypertensive patients with diabetic nephropathy. To evaluate a direct renoprotective effect by MR blockade independent of an antihypertensive effect in the context of diabetic nephropathy, esaxerenone (3 mg/kg), olmesartan (an angiotensin II receptor blocker; 1 mg/kg), or both were orally administered to KK-Ay mice, a type 2 diabetes model, once daily for 56 days. Urinary albumin (Ualb), UACR, and markers, such as podocalyxin, monocyte chemoattractant protein-1 (MCP-1), and 8-hydroxy-2'-deoxyguanosine (8-OHdG), were measured, along with systolic blood pressure (SBP), fasting blood glucose, and serum K+ levels. Prior to the initiation of drug administration, KK-Ay mice showed higher blood glucose, insulin, Ualb excretion, and UACR levels than C57BL/6 J mice, a nondiabetic control, indicating the development of diabetic renal injury. Combined treatment with esaxerenone and olmesartan significantly reduced the change in UACR from baseline compared with the change associated with vehicle at week 8 (-1.750 vs. 0.339 g/gCre; P < 0.002) and significantly inhibited the change in Ualb from baseline compared with the change associated with vehicle at week 8 (P < 0.002). The combination treatment also reduced urinary excretion of podocalyxin and MCP-1, but did not influence 8-OHdG excretion, SBP, blood glucose, or serum K+ levels. Overall, esaxerenone plus olmesartan treatment ameliorated diabetic nephropathy in KK-Ay mice without affecting SBP, suggesting that the renoprotective effects of esaxerenone could be exerted independently of its antihypertensive effect.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Nefropatias Diabéticas/tratamento farmacológico , Imidazóis/uso terapêutico , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Pirróis/uso terapêutico , Sulfonas/uso terapêutico , Tetrazóis/uso terapêutico , Albuminúria/tratamento farmacológico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Diabetes Mellitus Tipo 2/complicações , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Imidazóis/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Pirróis/farmacologia , Sulfonas/farmacologia , Tetrazóis/farmacologia
20.
Nat Cell Biol ; 22(11): 1346-1356, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33046882

RESUMO

Cardiomyocyte loss after injury results in adverse remodelling and fibrosis, inevitably leading to heart failure. The ERBB2-Neuregulin and Hippo-YAP signalling pathways are key mediators of heart regeneration, yet the crosstalk between them is unclear. We demonstrate that transient overexpression of activated ERBB2 in cardiomyocytes (OE CMs) promotes cardiac regeneration in a heart failure model. OE CMs present an epithelial-mesenchymal transition (EMT)-like regenerative response manifested by cytoskeletal remodelling, junction dissolution, migration and extracellular matrix turnover. We identified YAP as a critical mediator of ERBB2 signalling. In OE CMs, YAP interacts with nuclear-envelope and cytoskeletal components, reflecting an altered mechanical state elicited by ERBB2. We identified two YAP-activating phosphorylations on S352 and S274 in OE CMs, which peak during metaphase, that are ERK dependent and Hippo independent. Viral overexpression of YAP phospho-mutants dampened the proliferative competence of OE CMs. Together, we reveal a potent ERBB2-mediated YAP mechanotransduction signalling, involving EMT-like characteristics, resulting in robust heart regeneration.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Transição Epitelial-Mesenquimal , Insuficiência Cardíaca/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptor ErbB-2/metabolismo , Regeneração , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular/genética , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrose , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Mecanotransdução Celular , Camundongos Transgênicos , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Fosforilação , Receptor ErbB-2/genética , Proteínas de Sinalização YAP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA