Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Pediatr ; 23(1): 82, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36800953

RESUMO

BACKGROUND: Constantly elevated intra-abdominal pressure (IAH) can lead to abdominal compartment syndrome (ACS), which is associated with organ dysfunction and even multiorgan failure. Our 2010 survey revealed an inconsistent acceptance of definitions and guidelines among pediatric intensivists regarding the diagnosis and treatment of IAH and ACS in Germany. This is the first survey to assess the impact of the updated guidelines on neonatal/pediatric intensive care units (NICU/PICU) in German-speaking countries after WSACS published those in 2013. METHODS: We conducted a follow-up survey and sent 473 questionnaires to all 328 German-speaking pediatric hospitals. We compared our findings regarding awareness, diagnostics and therapy of IAH and ACS with the results of our 2010 survey. RESULTS: The response rate was 48% (n = 156). The majority of respondents was from Germany (86%) and working in PICUs with mostly neonatal patients (53%). The number of participants who stated that IAH and ACS play a role in their clinical practice rose from 44% in 2010 to 56% in 2016. Similar to the 2010 investigations, only a few neonatal/pediatric intensivists knew the correct WSACS definition of an IAH (4% vs 6%). Different from the previous study, the number of participants who correctly defined an ACS increased from 18 to 58% (p < 0,001). The number of respondents measuring intra-abdominal pressure (IAP) increased from 20 to 43% (p < 0,001). Decompressive laparotomies (DLs) were performed more frequently than in 2010 (36% vs. 19%, p < 0,001), and the reported survival rate was higher when a DL was used (85% ± 17% vs. 40 ± 34%). CONCLUSIONS: Our follow-up survey of neonatal/pediatric intensivists showed an improvement in the awareness and knowledge of valid definitions of ACS. Moreover, there has been an increase in the number of physicians measuring IAP in patients. However, a significant number has still never diagnosed IAH/ACS, and more than half of the respondents have never measured IAP. This reinforces the suspicion that IAH and ACS are only slowly coming into the focus of neonatal/pediatric intensivists in German-speaking pediatric hospitals. The goal should be to raise awareness of IAH and ACS through education and training and to establish diagnostic algorithms, especially for pediatric patients. The increased survival rate after conducting a prompt DL consolidates the impression that the probability of survival can be increased by timely surgical decompression in the case of full-blown ACS.


Assuntos
Síndromes Compartimentais , Hipertensão Intra-Abdominal , Recém-Nascido , Humanos , Criança , Hipertensão Intra-Abdominal/diagnóstico , Hipertensão Intra-Abdominal/etiologia , Hipertensão Intra-Abdominal/terapia , Unidades de Terapia Intensiva Neonatal , Seguimentos , Inquéritos e Questionários , Unidades de Terapia Intensiva Pediátrica , Síndromes Compartimentais/diagnóstico , Síndromes Compartimentais/terapia , Unidades de Terapia Intensiva
2.
Hum Genet ; 139(11): 1443-1454, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32514796

RESUMO

Dilated cardiomyopathy (DCM) belongs to the most frequent forms of cardiomyopathy mainly characterized by cardiac dilatation and reduced systolic function. Although most cases of DCM are classified as sporadic, 20-30% of cases show a heritable pattern. Familial forms of DCM are genetically heterogeneous, and mutations in several genes have been identified that most commonly play a role in cytoskeleton and sarcomere-associated processes. Still, a large number of familial cases remain unsolved. Here, we report five individuals from three independent families who presented with severe dilated cardiomyopathy during the neonatal period. Using whole-exome sequencing (WES), we identified causative, compound heterozygous missense variants in RPL3L (ribosomal protein L3-like) in all the affected individuals. The identified variants co-segregated with the disease in each of the three families and were absent or very rare in the human population, in line with an autosomal recessive inheritance pattern. They are located within the conserved RPL3 domain of the protein and were classified as deleterious by several in silico prediction software applications. RPL3L is one of the four non-canonical riboprotein genes and it encodes the 60S ribosomal protein L3-like protein that is highly expressed only in cardiac and skeletal muscle. Three-dimensional homology modeling and in silico analysis of the affected residues in RPL3L indicate that the identified changes specifically alter the interaction of RPL3L with the RNA components of the 60S ribosomal subunit and thus destabilize its binding to the 60S subunit. In conclusion, we report that bi-allelic pathogenic variants in RPL3L are causative of an early-onset, severe neonatal form of dilated cardiomyopathy, and we show for the first time that cytoplasmic ribosomal proteins are involved in the pathogenesis of non-syndromic cardiomyopathies.


Assuntos
Cardiomiopatia Dilatada/genética , Mutação de Sentido Incorreto/genética , Proteínas Ribossômicas/genética , Ribossomos/genética , Alelos , Exoma/genética , Feminino , Coração/fisiopatologia , Humanos , Lactente , Recém-Nascido , Masculino , Músculo Esquelético/fisiopatologia , Linhagem , Fenótipo , RNA/genética , Proteína Ribossômica L3
3.
Dev Biol ; 413(2): 153-159, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26988120

RESUMO

A subset of macrophages that reside in adult tissues originate from the fetal yolk sac, while others derive from circulating monocytes. These ontologically different macrophage subsets have distinct roles in tissue injury responses, with the embryonic population overall having beneficial activity in cardiac repair. Here we show that fetal yolk macrophages are recruited to a niche within and just below the epicardium, the mesothelial covering of the heart. The epicardium was required for establishment of yolk sac macrophages in this region of the fetal heart, and this function of epicardium depended on its expression of the transcription factor WT1. Thus, tissue-specific cues and transcriptional programs recruit or retain embryonic macrophages in their final abodes, where they help to shape organ homeostasis and injury responses.


Assuntos
Macrófagos/citologia , Miocárdio/citologia , Pericárdio/citologia , Saco Vitelino/citologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Diferenciação Celular , Linhagem da Célula , Epitélio , Glicoproteínas/metabolismo , Coração/embriologia , Macrófagos/metabolismo , Proteínas de Membrana Transportadoras , Camundongos , Comunicação Parácrina
4.
Am J Respir Cell Mol Biol ; 54(2): 222-30, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26121126

RESUMO

The lung is enveloped by a layer of specialized epithelium, the pulmonary mesothelium. In other organs, mesothelial cells undergo epithelial-mesenchymal transition and contribute to organ stromal cells. The contribution of pulmonary mesothelial cells (PMCs) to the developing lung has been evaluated with differing conclusions. PMCs have also been indirectly implicated in lung fibrosis in the progressive, fatal lung disease idiopathic pulmonary fibrosis. We used fetal or postnatal genetic pulse labeling of PMCs to assess their fate in murine development, normal lung homeostasis, and models of pulmonary fibrosis. We found that most fetal PMC-derived mesenchymal cells (PMCDCs) expressed markers of pericytes and fibroblasts, only a small minority expressed smooth muscle markers, and none expressed endothelial cell markers. Postnatal PMCs did not contribute to lung mesenchyme during normal lung homeostasis or in models of lung fibrosis. However, fetal PMCDCs were abundant and actively proliferating within fibrotic regions in lung fibrosis models, suggesting that they actively participate in the fibrotic process. These data clarify the role of fetal and postnatal PMCDCs in lung development and disease.


Assuntos
Linhagem da Célula , Fibroblastos/patologia , Pulmão/patologia , Mesoderma/patologia , Fibrose Pulmonar/patologia , Animais , Biomarcadores/metabolismo , Bleomicina , Proliferação de Células , Rastreamento de Células , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Fibroblastos/metabolismo , Pulmão/metabolismo , Mesoderma/metabolismo , Camundongos Transgênicos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fenótipo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
5.
Circ Res ; 116(1): 35-45, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25249570

RESUMO

RATIONALE: Yes-associated protein (YAP), the nuclear effector of Hippo signaling, regulates cellular growth and survival in multiple organs, including the heart, by interacting with TEA (transcriptional enhancer activator)-domain sequence-specific DNA-binding proteins. Recent studies showed that YAP stimulates cardiomyocyte proliferation and survival. However, the direct transcriptional targets through which YAP exerts its effects are poorly defined. OBJECTIVE: To identify direct YAP targets that mediate its mitogenic and antiapoptotic effects in the heart. METHODS AND RESULTS: We identified direct YAP targets by combining differential gene expression analysis in YAP gain- and loss-of-function with genome-wide identification of YAP-bound loci using chromatin immunoprecipitation and high throughput sequencing. This screen identified Pik3cb, encoding p110ß, a catalytic subunit of phosphoinositol-3-kinase, as a candidate YAP effector that promotes cardiomyocyte proliferation and survival. YAP and TEA-domain occupied a conserved enhancer within the first intron of Pik3cb, and this enhancer drove YAP-dependent reporter gene expression. Yap gain- and loss-of-function studies indicated that YAP is necessary and sufficient to activate the phosphoinositol-3-kinase-Akt pathway. Like Yap, Pik3cb gain-of-function stimulated cardiomyocyte proliferation, and Pik3cb knockdown dampened YAP mitogenic activity. Reciprocally, impaired heart function in Yap loss-of-function was significantly rescued by adeno-associated virus-mediated Pik3cb expression. CONCLUSIONS: Pik3cb is a crucial direct target of YAP, through which the YAP activates phosphoinositol-3-kinase-AKT pathway and regulates cardiomyocyte proliferation and survival.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Proliferação de Células/fisiologia , Classe Ib de Fosfatidilinositol 3-Quinase/biossíntese , Miócitos Cardíacos/fisiologia , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Proto-Oncogênicas c-akt/biossíntese , Animais , Animais Recém-Nascidos , Proteínas Reguladoras de Apoptose/genética , Sequência de Bases , Sobrevivência Celular/fisiologia , Células Cultivadas , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Via de Sinalização Hippo , Camundongos , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Transdução de Sinais/fisiologia , Proteínas de Sinalização YAP
7.
J Biol Chem ; 289(27): 18681-92, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24831012

RESUMO

Cardiac malformations due to aberrant development of the atrioventricular (AV) valves are among the most common forms of congenital heart diseases. Normally, heart valve mesenchyme is formed from an endothelial to mesenchymal transition (EMT) of endothelial cells of the endocardial cushions. Yes-associated protein 1 (YAP1) has been reported to regulate EMT in vitro, in addition to its known role as a major regulator of organ size and cell proliferation in vertebrates, leading us to hypothesize that YAP1 is required for heart valve development. We tested this hypothesis by conditional inactivation of YAP1 in endothelial cells and their derivatives. This resulted in markedly hypocellular endocardial cushions due to impaired formation of heart valve mesenchyme by EMT and to reduced endocardial cell proliferation. In endothelial cells, TGFß induces nuclear localization of Smad2/3/4 complex, which activates expression of Snail, Twist1, and Slug, key transcription factors required for EMT. YAP1 interacts with this complex, and loss of YAP1 disrupts TGFß-induced up-regulation of Snail, Twist1, and Slug. Together, our results identify a role of YAP1 in regulating EMT through modulation of TGFß-Smad signaling and through proliferative activity during cardiac cushion development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transdiferenciação Celular , Coxins Endocárdicos/citologia , Coxins Endocárdicos/embriologia , Células Endoteliais/citologia , Mesoderma/citologia , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Linhagem da Célula , Endocárdio/citologia , Endocárdio/embriologia , Endocárdio/metabolismo , Feminino , Deleção de Genes , Masculino , Camundongos , Mutação , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Transdução de Sinais , Proteínas Smad/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Proteínas de Sinalização YAP
8.
Circ Res ; 115(3): 354-63, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24833660

RESUMO

RATIONALE: Yes-associated protein (YAP), the terminal effector of the Hippo signaling pathway, is crucial for regulating embryonic cardiomyocyte proliferation. OBJECTIVE: We hypothesized that YAP activation after myocardial infarction (MI) would preserve cardiac function and improve survival. METHODS AND RESULTS: We used a cardiac-specific, inducible expression system to activate YAP in adult mouse heart. Activation of YAP in adult heart promoted cardiomyocyte proliferation and did not deleteriously affect heart function. Furthermore, YAP activation after MI preserved heart function and reduced infarct size. Using adeno-associated virus subtype 9 (AAV9) as a delivery vector, we expressed human YAP (hYAP) in the adult murine myocardium immediately after MI. We found that AAV9:hYAP significantly improved cardiac function and mouse survival. AAV9:hYAP did not exert its salutary effects by reducing cardiomyocyte apoptosis. Rather, AAV9:hYAP stimulated adult cardiomyocyte proliferation. Gene expression profiling indicated that AAV9:hYAP stimulated expression of cell cycle genes and promoted a less mature cardiac gene expression signature. CONCLUSIONS: Cardiac-specific YAP activation after MI mitigated myocardial injury, improved cardiac function, and enhanced survival. These findings suggest that therapeutic activation of YAP or its downstream targets, potentially through AAV-mediated gene therapy, may be a strategy to improve outcome after MI.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Animais , Apoptose/genética , Apoptose/fisiologia , Cardiomegalia , Proliferação de Células , Sobrevivência Celular/fisiologia , Dependovirus/genética , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Contração Miocárdica/fisiologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/mortalidade , Miócitos Cardíacos/citologia , Cadeias Pesadas de Miosina/genética , Regeneração/genética , Regeneração/fisiologia , Taxa de Sobrevida , Fatores de Transcrição , Transcriptoma , Proteínas de Sinalização YAP
9.
Dev Cell ; 27(1): 5-18, 2013 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-24135228

RESUMO

Adrenal glands and gonads share a common primordium (AGP), but the molecular events driving differentiation are poorly understood. Here we demonstrate that the Wilms tumor suppressor WT1 is a key factor defining AGP identity by inhibiting the steroidogenic differentiation process. Indeed, ectopic expression of WT1 precludes differentiation into adrenocortical steroidogenic cells by locking them into a progenitor state. Chromatin immunoprecipitation experiments identify Tcf21 and Gli1 as direct targets of WT1. Moreover, cell lineage tracing analyses identify a long-living progenitor population within the adrenal gland, characterized by the expression of WT1, GATA4, GLI1, and TCF21, that can generate steroidogenic cells in vivo. Strikingly, gonadectomy dramatically activates these WT1(+) cells and leads to their differentiation into gonadal steroidogenic tissue. Thus, our data describe a mechanism of response to organ loss by recreating hormone-producing cells at a heterotopic site.


Assuntos
Glândulas Suprarrenais/citologia , Células-Tronco Embrionárias/metabolismo , Gônadas/citologia , Proteínas WT1/metabolismo , Glândulas Suprarrenais/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Castração , Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Hormônios Esteroides Gonadais/deficiência , Gônadas/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas WT1/genética , Proteína GLI1 em Dedos de Zinco
11.
Nat Biotechnol ; 31(10): 898-907, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24013197

RESUMO

In a cell-free approach to regenerative therapeutics, transient application of paracrine factors in vivo could be used to alter the behavior and fate of progenitor cells to achieve sustained clinical benefits. Here we show that intramyocardial injection of synthetic modified RNA (modRNA) encoding human vascular endothelial growth factor-A (VEGF-A) results in the expansion and directed differentiation of endogenous heart progenitors in a mouse myocardial infarction model. VEGF-A modRNA markedly improved heart function and enhanced long-term survival of recipients. This improvement was in part due to mobilization of epicardial progenitor cells and redirection of their differentiation toward cardiovascular cell types. Direct in vivo comparison with DNA vectors and temporal control with VEGF inhibitors revealed the greatly increased efficacy of pulse-like delivery of VEGF-A. Our results suggest that modRNA is a versatile approach for expressing paracrine factors as cell fate switches to control progenitor cell fate and thereby enhance long-term organ repair.


Assuntos
Linhagem da Célula , Infarto do Miocárdio/terapia , Miocárdio/patologia , RNA Mensageiro/metabolismo , Regeneração , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Apoptose , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/patologia , Técnicas de Transferência de Genes , Humanos , Cinética , Luciferases/metabolismo , Camundongos , Modelos Biológicos , Músculo Esquelético/metabolismo , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , RNA Mensageiro/genética , Transplante de Células-Tronco , Análise de Sobrevida , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
Circ Res ; 110(12): 1628-45, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22679138

RESUMO

Epithelial to mesenchymal transition (EMT) converts epithelial cells to mobile and developmentally plastic mesenchymal cells. All cells in the heart arise from one or more EMTs. Endocardial and epicardial EMTs produce most of the noncardiomyocyte lineages of the mature heart. Endocardial EMT generates valve progenitor cells and is necessary for formation of the cardiac valves and for complete cardiac septation. Epicardial EMT is required for myocardial growth and coronary vessel formation, and it generates cardiac fibroblasts, vascular smooth muscle cells, a subset of coronary endothelial cells, and possibly a subset of cardiomyocytes. Emerging studies suggest that these developmental mechanisms are redeployed in adult heart valve disease, in cardiac fibrosis, and in myocardial responses to ischemic injury. Redirection and amplification of disease-related EMTs offer potential new therapeutic strategies and approaches for treatment of heart disease. Here, we review the role and molecular regulation of endocardial and epicardial EMT in fetal heart development, and we summarize key literature implicating reactivation of endocardial and epicardial EMT in adult heart disease.


Assuntos
Endocárdio/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Cardiopatias/fisiopatologia , Coração/crescimento & desenvolvimento , Pericárdio/fisiologia , Animais , Endocárdio/citologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Coração/embriologia , Cardiopatias/patologia , Humanos , Pericárdio/citologia
13.
Proc Natl Acad Sci U S A ; 109(7): 2394-9, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308401

RESUMO

Heart growth is tightly controlled so that the heart reaches a predetermined size. Fetal heart growth occurs through cardiomyocyte proliferation, whereas postnatal heart growth involves primarily physiological cardiomyocyte hypertrophy. The Hippo kinase cascade is an important regulator of organ growth. A major target of this kinase cascade is YAP1, a transcriptional coactivator that is inactivated by Hippo kinase activity. Here, we used both genetic gain and loss of Yap1 function to investigate its role in regulating proliferative and physiologic hypertrophic heart growth. Fetal Yap1 inactivation caused marked, lethal myocardial hypoplasia and decreased cardiomyocyte proliferation, whereas fetal activation of YAP1 stimulated cardiomyocyte proliferation. Enhanced proliferation was particularly dramatic in trabecular cardiomyocytes that normally exit from the cell cycle. Remarkably, YAP1 activation was sufficient to stimulate proliferation of postnatal cardiomyocytes, both in culture and in the intact heart. A dominant negative peptide that blocked YAP1 binding to TEAD transcription factors inhibited YAP1 proliferative activity, indicating that this activity requires YAP1-TEAD interaction. Although Yap1 was a critical regulator of cardiomyocyte proliferation, it did not influence physiological hypertrophic growth of cardiomyocytes, because postnatal Yap1 gain or loss of function did not significantly alter cardiomyocyte size. These studies demonstrate that Yap1 is a crucial regulator of cardiomyocyte proliferation, cardiac morphogenesis, and myocardial trabeculation. Activation of Yap1 in postnatal cardiomyocytes may be a useful strategy to stimulate cardiomyocyte expansion in therapeutic myocardial regeneration.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Cardiomegalia/metabolismo , Coração/crescimento & desenvolvimento , Miocárdio/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Animais , Genes cdc , Ratos , Serina-Treonina Quinase 3 , Proteínas de Sinalização YAP
14.
Genes Dev ; 26(1): 37-42, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22215809

RESUMO

Polycomb-repressive complex 2 (PRC2) promotes tissue-specific differentiation by depositing trimethylated histone H3 Lys 27 (H3K27me3) epigenetic marks to silence ectopic gene expression programs. Here, we show that EZH2, the catalytic subunit of PRC2, is required for cardiac morphogenesis. Both in vitro and in fetal hearts, EZH2 interacted with cardiac transcription factor GATA4 and directly methylated it at Lys 299. PRC2 methylation of GATA4 attenuated its transcriptional activity by reducing its interaction with and acetylation by p300. Our results reveal a new mechanism of PRC2-mediated transcriptional repression in which PRC2 methylates a transcription factor to inhibit its transcriptional activity.


Assuntos
Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Repressoras/metabolismo , Animais , Proteína p300 Associada a E1A/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste , Histona-Lisina N-Metiltransferase/metabolismo , Metilação , Camundongos , Complexo Repressor Polycomb 2 , Proteínas do Grupo Polycomb , Ligação Proteica
15.
Circ Res ; 110(3): 406-15, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22158708

RESUMO

RATIONALE: Epigenetic marks are crucial for organogenesis, but their role in heart development is poorly understood. Polycomb repressive complex 2 (PRC2) trimethylates histone H3 at lysine 27, which establishes H3K27me3 repressive epigenetic marks that promote tissue-specific differentiation by silencing ectopic gene programs. OBJECTIVE: We studied the function of PRC2 in murine heart development using a tissue-restricted conditional inactivation strategy. METHODS AND RESULTS: Inactivation of the PRC2 subunit Ezh2 by Nkx2-5(Cre) (Ezh2(NK)) caused lethal congenital heart malformations, namely, compact myocardial hypoplasia, hypertrabeculation, and ventricular septal defect. Candidate and genome-wide RNA expression profiling and chromatin immunoprecipitation analyses of Ezh2(NK) heart identified genes directly repressed by EZH2. Among these were the potent cell cycle inhibitors Ink4a/b (inhibitors of cyclin-dependent kinase 4 A and B), the upregulation of which was associated with decreased cardiomyocyte proliferation in Ezh2(NK). EZH2-repressed genes were enriched for transcriptional regulators of noncardiomyocyte expression programs such as Pax6, Isl1, and Six1. EZH2 was also required for proper spatiotemporal regulation of cardiac gene expression, because Hcn4, Mlc2a, and Bmp10 were inappropriately upregulated in ventricular RNA. PRC2 was also required later in heart development, as indicated by cardiomyocyte-restricted TNT-Cre inactivation of the PRC2 subunit Eed. However, Ezh2 inactivation by TNT-Cre did not cause an overt phenotype, likely because of functional redundancy with Ezh1. Thus, early Ezh2 inactivation by Nk2-5(Cre) caused later disruption of cardiomyocyte gene expression and heart development. CONCLUSIONS: Our study reveals a previously undescribed role of EZH2 in regulating heart formation and shows that perturbation of the epigenetic landscape early in cardiogenesis has sustained disruptive effects at later developmental stages.


Assuntos
Epigênese Genética/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Coração/embriologia , Coração/fisiologia , Proteínas Repressoras/fisiologia , Animais , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste , Estudo de Associação Genômica Ampla , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/fisiologia , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Modelos Animais , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Complexo Repressor Polycomb 2 , Proteínas do Grupo Polycomb , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Proteínas Repressoras/genética , Fatores de Transcrição/fisiologia
16.
J Mol Cell Cardiol ; 52(1): 43-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21907210

RESUMO

Myocardial infarction (MI) is one of the leading causes of morbidity and mortality world-wide. Whether endogenous repair and regenerative ability could be augmented by drug administration is an important issue for generation of novel therapeutic approach. Recently it was reported that in mice pretreated with thymosin beta 4 (TB4) and subsequently subjected to experimental MI, a subset of epicardial cells differentiated into cardiomyocytes. In clinical settings, epicardial priming with TB4 prior to MI is impractical. Here we tested if TB4 treatment after MI could reprogram epicardium into cardiomyocytes and augment the epicardium's injury response. Using epicardium genetic lineage trace line Wt1(CreERT2/+) and double reporter line Rosa26(mTmG/+), we found post-MI TB4 treatment significantly increased the thickness of epicardium and coronary capillary density. However, epicardium-derived cells did not adopt cardiomyocyte fate, nor did they migrate into myocardium to become coronary endothelial cells. Our result thus indicates that TB4 treatment after MI does not alter epicardial cell fate to include the cardiomyocyte lineage, providing both cautions and insights for the full exploration of the potential benefits of TB4 in the clinical settings. This article is part of a Special Issue entitled 'Possible Editorial'.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Pericárdio/citologia , Pericárdio/efeitos dos fármacos , Timosina/farmacologia , Timosina/uso terapêutico , Animais , Diferenciação Celular/efeitos dos fármacos , Camundongos , Infarto do Miocárdio/metabolismo
17.
J Mol Med (Berl) ; 89(10): 971-83, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21735044

RESUMO

The number of circulating endothelial progenitor cells (EPCs) inversely correlates with cardiovascular risk and clinical outcome, and thus has been proposed as a valuable biomarker for risk assessment, disease progression, and response to therapy. However, current strategies for isolation of these rare cells are limited to complex, laborious approaches. The goal of this study was the design and validation of a disposable microfluidic platform capable of selectively capturing and enumerating EPCs directly from human whole blood in healthy and diseased subjects, eliminating sample preprocessing. We then applied the "EPC capture chip" clinically and determined EPC numbers in blood from patients with pulmonary arterial hypertension (PAH). Blood was collected in tubes and injected into polymeric microfluidic chips containing microcolumns pre-coated with anti-CD34 antibody. Captured cells were immunofluorescently stained for the expression of stem and endothelial antigens, identified and counted. The EPC capture chip was validated with conventional flow cytometry counts (r = 0.83). The inter- and intra-day reliability of the microfluidic devices was confirmed at different time points in triplicates over 1-5 months. In a cohort of 43 patients with three forms of PAH (idiopathic/heritable, drug-induced, and connective tissue disease), EPC numbers are ≈50% lower in PAH subjects vs. matched controls and inversely related to two potential disease modifiers: body mass index and postmenopausal status. The EPC capture chip (5 × 30 × 0.05 mm(3)) requires only 200 µL of human blood and has the strong potential to serve as a rapid bedside test for the screening and monitoring of patients with PAH and other proliferative cardiovascular, pulmonary, malignant, and neurodegenerative diseases.


Assuntos
Células Endoteliais/patologia , Hipertensão Pulmonar/diagnóstico , Hipertensão Pulmonar/patologia , Técnicas Analíticas Microfluídicas/instrumentação , Técnicas Analíticas Microfluídicas/métodos , Células-Tronco/patologia , Adulto , Idoso , Envelhecimento/patologia , Índice de Massa Corporal , Estudos de Casos e Controles , Contagem de Células , Separação Celular , Desenho de Equipamento , Hipertensão Pulmonar Primária Familiar , Feminino , Humanos , Hipertensão Pulmonar/sangue , Masculino , Pessoa de Meia-Idade , Pós-Menopausa/sangue , Reprodutibilidade dos Testes
18.
Dev Biol ; 356(2): 421-31, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21663736

RESUMO

An epithelial sheet, the epicardium, lines the surface of the heart. In the developing embryo, the epicardium expresses the transcriptional regulator Wilm's Tumor Gene 1 (Wt1). Through incompletely understood mechanisms, Wt1 inactivation derails normal heart development. We investigated mechanisms by which Wt1 regulates heart development and epicardial epithelial to mesenchymal transition (EMT). We used genetic lineage tracing approaches to track and isolate epicardium and epicardium derivatives in hearts lacking Wt1 (Wt1(KO)). Wt1(KO) hearts had diminished proliferation of compact myocardium and impaired coronary plexus formation. Wt1(KO) epicardium failed to undergo EMT. Wt1(KO) epicardium expressed reduced Lef1 and Ctnnb1 (ß-catenin), key components of the canonical Wnt/ß-catenin signaling pathway. Wt1(KO) epicardium expressed decreased levels of canonical Wnt downstream targets Axin2, Cyclin D1, and Cyclin D2 and exhibited decreased activity of the Batgal Wnt/ß-catenin reporter transgene, suggestive of diminished canonical Wnt signaling. Hearts with epicardium-restricted Ctnnb1 loss of function resembled Wt1(KO) hearts and also failed to undergo epicardial EMT. However, Ctnnb1 inactivation did not alter WT1 expression, positioning Wt1 upstream of canonical Wnt/ß-catenin signaling. Wnt5a, a prototypic non-canonical Wnt with enriched epicardial expression, and Raldh2, a key regulator of retinoic acid signaling confined to the epicardium, were also markedly downregulated in Wt1(KO) epicardium. Hearts lacking Wnt5a or Raldh2 shared phenotypic features with Wt1(KO). Although Wt1 has been proposed to regulate EMT by repressing E-cadherin, we detected no change in E-cadherin in Wt1(KO) epicardium. Collectively, our study shows that Wt1 regulates epicardial EMT and heart development through canonical Wnt, non-canonical Wnt, and retinoic acid signaling pathways.


Assuntos
Transição Epitelial-Mesenquimal , Pericárdio/embriologia , Transdução de Sinais/fisiologia , Tretinoína/fisiologia , Proteínas WT1/fisiologia , beta Catenina/fisiologia , Aldeído Oxirredutases/genética , Animais , Camundongos , Fatores de Transcrição da Família Snail , Fatores de Transcrição/fisiologia
20.
Dev Biol ; 338(2): 251-61, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20025864

RESUMO

The annulus fibrosis electrically insulates the atria and ventricles, allowing the timed sequential beating of these structures that is necessary for efficient heart function. Abnormal development of the annulus fibrosis leads to persistence of accessory electrical pathways from atria to ventricles, providing the anatomical substrate for re-entrant cardiac arrhythmias such as Wolff-Parkinson-White syndrome. To better understand the development of the annulus fibrosis and the etiology of these cardiac arrhythmias, we used Cre-LoxP technology to assess the contribution of epicardium derived cells (EPDCs) to the annulus fibrosis. We found that EPDCs migrated into the region of the forming annulus fibrosis, marked by the protein periostin. These EPDCs also stained positive for procollagen I, suggesting that the EPDCs themselves synthesize proteins of the annulus fibrosis. To further test the hypothesis that EPDCs contribute to cells that synthesize the annulus fibrosis, we purified genetically marked EPDCs from the atrioventricular region and measured gene expression by quantitative PCR. These EPDCs were highly enriched for mRNAs encoding periostin, procollagen I, fibronectin I, vimentin, discoidin domain receptor 2, and tenascin C, markers of fibroblasts and components of the annulus fibrosis. In addition, these EPDCs were highly enriched for Snail, Smad1, Slug, and Twist1, markers for epithelial-to-mesenchymal transition (EMT), and a metalloprotease, Mmp2, that contributes to cellular migration. Our work provides for the first time definitive evidence that epicardium contributes to formation of the mammalian annulus fibrosis through EMT. Abnormalities of this differentiation process may underlie development of some forms of re-entrant atrioventricular tachycardia.


Assuntos
Arritmias Cardíacas/etiologia , Sistema de Condução Cardíaco/citologia , Miocárdio/citologia , Pericárdio/citologia , Animais , Eletrofisiologia Cardíaca , Movimento Celular , Fibrose , Perfilação da Expressão Gênica , Átrios do Coração/citologia , Ventrículos do Coração/citologia , Camundongos , RNA Mensageiro/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...