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










Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 134(5): 529-546, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38348657

RESUMO

BACKGROUND: Mature endothelial cells (ECs) are heterogeneous, with subtypes defined by tissue origin and position within the vascular bed (ie, artery, capillary, vein, and lymphatic). How this heterogeneity is established during the development of the vascular system, especially arteriovenous specification of ECs, remains incompletely characterized. METHODS: We used droplet-based single-cell RNA sequencing and multiplexed error-robust fluorescence in situ hybridization to define EC and EC progenitor subtypes from E9.5, E12.5, and E15.5 mouse embryos. We used trajectory inference to analyze the specification of arterial ECs (aECs) and venous ECs (vECs) from EC progenitors. Network analysis identified candidate transcriptional regulators of arteriovenous differentiation, which we tested by CRISPR (clustered regularly interspaced short palindromic repeats) loss of function in human-induced pluripotent stem cells undergoing directed differentiation to aECs or vECs (human-induced pluripotent stem cell-aECs or human-induced pluripotent stem cell-vECs). RESULTS: From the single-cell transcriptomes of 7682 E9.5 to E15.5 ECs, we identified 19 EC subtypes, including Etv2+Bnip3+ EC progenitors. Spatial transcriptomic analysis of 15 448 ECs provided orthogonal validation of these EC subtypes and established their spatial distribution. Most embryonic ECs were grouped by their vascular-bed types, while ECs from the brain, heart, liver, and lung were grouped by their tissue origins. Arterial (Eln, Dkk2, Vegfc, and Egfl8), venous (Fam174b and Clec14a), and capillary (Kcne3) marker genes were identified. Compared with aECs, embryonic vECs and capillary ECs shared fewer markers than their adult counterparts. Early capillary ECs with venous characteristics functioned as a branch point for differentiation of aEC and vEC lineages. CONCLUSIONS: Our results provide a spatiotemporal map of embryonic EC heterogeneity at single-cell resolution and demonstrate that the diversity of ECs in the embryo arises from both tissue origin and vascular-bed position. Developing aECs and vECs share common venous-featured capillary precursors and are regulated by distinct transcriptional regulatory networks.


Assuntos
Células Endoteliais , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Adulto , Humanos , Animais , Camundongos , Hibridização in Situ Fluorescente , Artérias , Encéfalo , Veias
2.
Nat Med ; 29(2): 412-421, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36797483

RESUMO

Dominant missense pathogenic variants in cardiac myosin heavy chain cause hypertrophic cardiomyopathy (HCM), a currently incurable disorder that increases risk for stroke, heart failure and sudden cardiac death. In this study, we assessed two different genetic therapies-an adenine base editor (ABE8e) and a potent Cas9 nuclease delivered by AAV9-to prevent disease in mice carrying the heterozygous HCM pathogenic variant myosin R403Q. One dose of dual-AAV9 vectors, each carrying one half of RNA-guided ABE8e, corrected the pathogenic variant in ≥70% of ventricular cardiomyocytes and maintained durable, normal cardiac structure and function. An additional dose provided more editing in the atria but also increased bystander editing. AAV9 delivery of RNA-guided Cas9 nuclease effectively inactivated the pathogenic allele, albeit with dose-dependent toxicities, necessitating a narrow therapeutic window to maintain health. These preclinical studies demonstrate considerable potential for single-dose genetic therapies to correct or silence pathogenic variants and prevent the development of HCM.


Assuntos
Cardiomiopatia Hipertrófica , Edição de Genes , Animais , Camundongos , Mutação de Sentido Incorreto , Miócitos Cardíacos , RNA
3.
Curr Protoc ; 2(10): e579, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36286606

RESUMO

This protocol describes a robust pipeline for simultaneously analyzing multiple samples by single-nucleus (sn)RNA-seq. cDNA obtained from each single sample are labeled with the same lipid-coupled oligonucleotide barcode (10X Genomics). Nuclei from as many as 12 individual samples can be pooled together and simultaneously processed for cDNA library construction and subsequent DNA sequencing. While previous protocols using lipid-coupled oligonucleotide barcodes were optimized for analysis of samples consisting of viable cells, this protocol is optimized for analyses of quick-frozen cell samples. The protocol ensures efficient recovery of nuclei both by incorporating high sucrose buffered solutions and by including a tracking dye (trypan blue) during nuclei isolation. The protocol also describes a procedure for removing single nuclei 'artifacts' by removing cell debris prior to single nuclear fractionation. This protocol informs the use of computational tools for filtering poorly labeled nuclei and assigning sample identity using barcode unique molecular identifier (UMI) read counts percentages. The computational pipeline is applicable to either cultured or primary, fresh or frozen cells, regardless of their cell types and species. Overall, this protocol reduces batch effects and experimental costs while enhancing sample comparison. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Labeling cells with lipid oligo barcodes and generating multiplexed single-nucleus RNA-seq libraries Basic Protocol 2: Bioinformatic deconvolution of the multiplexed snRNAseq libraries.


Assuntos
Sacarose , Azul Tripano , DNA Complementar , Análise de Sequência de RNA/métodos , Oligonucleotídeos , Lipídeos/genética
4.
Science ; 377(6606): eabo1984, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35926050

RESUMO

Pathogenic variants in genes that cause dilated cardiomyopathy (DCM) and arrhythmogenic cardiomyopathy (ACM) convey high risks for the development of heart failure through unknown mechanisms. Using single-nucleus RNA sequencing, we characterized the transcriptome of 880,000 nuclei from 18 control and 61 failing, nonischemic human hearts with pathogenic variants in DCM and ACM genes or idiopathic disease. We performed genotype-stratified analyses of the ventricular cell lineages and transcriptional states. The resultant DCM and ACM ventricular cell atlas demonstrated distinct right and left ventricular responses, highlighting genotype-associated pathways, intercellular interactions, and differential gene expression at single-cell resolution. Together, these data illuminate both shared and distinct cellular and molecular architectures of human heart failure and suggest candidate therapeutic targets.


Assuntos
Displasia Arritmogênica Ventricular Direita , Cardiomiopatia Dilatada , Insuficiência Cardíaca , Análise de Célula Única , Transcriptoma , Displasia Arritmogênica Ventricular Direita/genética , Atlas como Assunto , Cardiomiopatia Dilatada/genética , Núcleo Celular/genética , Insuficiência Cardíaca/genética , Ventrículos do Coração , Humanos , RNA-Seq
5.
Proc Natl Acad Sci U S A ; 119(28): e2204174119, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35787042

RESUMO

Myocardial fibrosis is a key pathologic feature of hypertrophic cardiomyopathy (HCM). However, the fibrotic pathways activated by HCM-causing sarcomere protein gene mutations are poorly defined. Because lysophosphatidic acid is a mediator of fibrosis in multiple organs and diseases, we tested the role of the lysophosphatidic acid pathway in HCM. Lysphosphatidic acid receptor 1 (LPAR1), a cell surface receptor, is required for lysophosphatidic acid mediation of fibrosis. We bred HCM mice carrying a pathogenic myosin heavy-chain variant (403+/-) with Lpar1-ablated mice to create mice carrying both genetic changes (403+/- LPAR1 -/-) and assessed development of cardiac hypertrophy and fibrosis. Compared with 403+/- LPAR1WT, 403+/- LPAR1 -/- mice developed significantly less hypertrophy and fibrosis. Single-nucleus RNA sequencing of left ventricular tissue demonstrated that Lpar1 was predominantly expressed by lymphatic endothelial cells (LECs) and cardiac fibroblasts. Lpar1 ablation reduced the population of LECs, confirmed by immunofluorescence staining of the LEC markers Lyve1 and Ccl21a and, by in situ hybridization, for Reln and Ccl21a. Lpar1 ablation also altered the distribution of fibroblast cell states. FB1 and FB2 fibroblasts decreased while FB0 and FB3 fibroblasts increased. Our findings indicate that Lpar1 is expressed predominantly by LECs and fibroblasts in the heart and is required for development of hypertrophy and fibrosis in an HCM mouse model. LPAR1 antagonism, including agents in clinical trials for other fibrotic diseases, may be beneficial for HCM.


Assuntos
Cardiomiopatia Hipertrófica , Receptores de Ácidos Lisofosfatídicos/genética , Animais , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Proteínas de Transporte , Modelos Animais de Doenças , Células Endoteliais/patologia , Fibrose , Hipertrofia/patologia , Camundongos
6.
Proc Natl Acad Sci U S A ; 119(21): e2203928119, 2022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35584116

RESUMO

Microtia is a congenital malformation that encompasses mild hypoplasia to complete loss of the external ear, or pinna. Although the contribution of genetic variation and environmental factors to microtia remains elusive, Amerindigenous populations have the highest reported incidence. Here, using both transmission disequilibrium tests and association studies in microtia trios (parents and affected child) and microtia cohorts enrolled in Latin America, we map an ∼10-kb microtia locus (odds ratio = 4.7; P = 6.78e-18) to the intergenic region between Roundabout 1 (ROBO1) and Roundabout 2 (ROBO2) (chr3: 78546526 to 78555137). While alleles at the microtia locus significantly increase the risk of microtia, their penetrance is low (<1%). We demonstrate that the microtia locus contains a polymorphic complex repeat element that is expanded in affected individuals. The locus is located near a chromatin loop region that regulates ROBO1 and ROBO2 expression in induced pluripotent stem cell­derived neural crest cells. Furthermore, we use single nuclear RNA sequencing to demonstrate ROBO1 and ROBO2 expression in both fibroblasts and chondrocytes of the mature human pinna. Because the microtia allele is enriched in Amerindigenous populations and is shared by some East Asian subjects with craniofacial malformations, we propose that both populations share a mutation that arose in a common ancestor prior to the ancient migration of Eurasian populations into the Americas and that the high incidence of microtia among Amerindigenous populations reflects the population bottleneck that occurred during the migration out of Eurasia.


Assuntos
Indígena Americano ou Nativo do Alasca , Microtia Congênita , Microtia Congênita/genética , Orelha Externa , Efeito Fundador , Humanos , Mutação , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Indígena Americano ou Nativo do Alasca/genética
7.
Curr Protoc ; 1(5): e132, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34043278

RESUMO

Both single-cell RNA sequencing (scRNAseq) and single-nucleus RNA sequencing (snRNAseq) can be used to characterize the transcriptional profile of individual cells, and based on these transcriptional profiles, help define cell type distribution in mixed cell populations. However, scRNAseq analyses are confounded if some of the cells are large (>50 µm) or if some of cells adhere more tightly to some adjacent cells than to others. Further, single cell isolation for scRNAseq requires fresh tissue, which may not be available for human or animal model tissues. Additionally, the current enzymatic and mechanical methods for single-cell dissociation can lead to stress-induced transcriptional artifacts. Nuclei for snRNAseq, on the other hand, can be isolated from any cell, regardless of size, and from either fresh or frozen tissues, and compared to whole cells, they are more resistant to mechanical pressures and appear not to exhibit as many cell isolation-based transcriptional artifacts. Here, we describe a time- and cost-effective procedure to isolate nuclei from mammalian cells and tissues. The protocol incorporates steps to mechanically disrupt samples to release nuclei. Compared to conventional nuclei isolation protocols, the approach described here increases overall efficiency, eliminates risk of contaminant exposure, and reduces volumes of expensive reagents. A series of RNA quality control checks are also incorporated to ensure success and reduce costs of subsequent snRNAseq experiments. Nuclei isolated by this procedure can be separated on the 10× Genomics Chromium system for either snRNAseq and/or Single-Nucleus ATAC-Seq (snATAC-Seq), and is also compatible with other single cell platforms. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Sample preparation and quality control check via RNA Isolation and Analysis Basic Protocol 2: Nuclei Isolation.


Assuntos
Núcleo Celular , Núcleo Solitário , Animais , Separação Celular , Modelos Animais de Doenças , Humanos , Análise de Sequência de RNA
8.
Elife ; 92020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33054971

RESUMO

Damaging GATA6 variants cause cardiac outflow tract defects, sometimes with pancreatic and diaphragmic malformations. To define molecular mechanisms for these diverse developmental defects, we studied transcriptional and epigenetic responses to GATA6 loss of function (LoF) and missense variants during cardiomyocyte differentiation of isogenic human induced pluripotent stem cells. We show that GATA6 is a pioneer factor in cardiac development, regulating SMYD1 that activates HAND2, and KDR that with HAND2 orchestrates outflow tract formation. LoF variants perturbed cardiac genes and also endoderm lineage genes that direct PDX1 expression and pancreatic development. Remarkably, an exon 4 GATA6 missense variant, highly associated with extra-cardiac malformations, caused ectopic pioneer activities, profoundly diminishing GATA4, FOXA1/2, and PDX1 expression and increasing normal retinoic acid signaling that promotes diaphragm development. These aberrant epigenetic and transcriptional signatures illuminate the molecular mechanisms for cardiovascular malformations, pancreas and diaphragm dysgenesis that arise in patients with distinct GATA6 variants.


Assuntos
Diafragma/crescimento & desenvolvimento , Fator de Transcrição GATA6/genética , Coração/crescimento & desenvolvimento , Células-Tronco Pluripotentes Induzidas/metabolismo , Pâncreas/crescimento & desenvolvimento , Diferenciação Celular/genética , Epigênese Genética/genética , Perfilação da Expressão Gênica , Humanos , Mutação de Sentido Incorreto/genética , Miócitos Cardíacos/metabolismo
9.
Nature ; 588(7838): 466-472, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32971526

RESUMO

Cardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and therapeutic strategies require a deeper understanding of the molecular processes involved in the healthy heart. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavour. Here, using state-of-the-art analyses of large-scale single-cell and single-nucleus transcriptomes, we characterize six anatomical adult heart regions. Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes and fibroblasts, and reveal distinct atrial and ventricular subsets of cells with diverse developmental origins and specialized properties. We define the complexity of the cardiac vasculature and its changes along the arterio-venous axis. In the immune compartment, we identify cardiac-resident macrophages with inflammatory and protective transcriptional signatures. Furthermore, analyses of cell-to-cell interactions highlight different networks of macrophages, fibroblasts and cardiomyocytes between atria and ventricles that are distinct from those of skeletal muscle. Our human cardiac cell atlas improves our understanding of the human heart and provides a valuable reference for future studies.


Assuntos
Miocárdio/citologia , Análise de Célula Única , Transcriptoma , Adipócitos/classificação , Adipócitos/metabolismo , Adulto , Enzima de Conversão de Angiotensina 2/análise , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Células Epiteliais/classificação , Células Epiteliais/metabolismo , Epitélio , Feminino , Fibroblastos/classificação , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Átrios do Coração/anatomia & histologia , Átrios do Coração/citologia , Átrios do Coração/inervação , Ventrículos do Coração/anatomia & histologia , Ventrículos do Coração/citologia , Ventrículos do Coração/inervação , Homeostase/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Miócitos Cardíacos/classificação , Miócitos Cardíacos/metabolismo , Neurônios/classificação , Neurônios/metabolismo , Pericitos/classificação , Pericitos/metabolismo , Receptores de Coronavírus/análise , Receptores de Coronavírus/genética , Receptores de Coronavírus/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Células Estromais/classificação , Células Estromais/metabolismo
10.
Curr Protoc Mol Biol ; 122(1): e55, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29851244

RESUMO

Understanding the transcriptional heterogeneity that occurs on the level of a single cell is critical to understanding the gene-regulatory mechanisms underlying development and disease. Population-level whole-transcriptome profiling approaches average gene expression across thousands to millions of cells and are unable to delineate the transcriptional signature of individual cells. Considerable biological heterogeneity between individual cells arises from differences in cell lineage, environment, or response to stimulus. The development of single-cell RNA sequencing (RNA-seq) enabled a high-resolution and unbiased analysis of cell transcriptomes. This unit describes a procedure utilizing an automated microfluidic platform, the Fluidigm C1 system, to simultaneously isolate dozens of single cells in a size- and shape-dependent manner. The microfluidic platform processes cells in individual nanoliter-scale reactions to convert their contents into double-stranded cDNA. This cDNA is used to make dual-indexed libraries using the Illumina Nextera XT library preparation kit for eventual RNA-seq analysis. © 2018 by John Wiley & Sons, Inc.


Assuntos
Expressão Gênica , Microfluídica/métodos , RNA/metabolismo , Análise de Célula Única/métodos , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Reação em Cadeia da Polimerase , RNA/genética , Transcrição Reversa/genética
11.
Circulation ; 138(4): 377-393, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29588317

RESUMO

BACKGROUND: No pharmacological therapy exists for calcific aortic valve disease (CAVD), which confers a dismal prognosis without invasive valve replacement. The search for therapeutics and early diagnostics is challenging because CAVD presents in multiple pathological stages. Moreover, it occurs in the context of a complex, multi-layered tissue architecture; a rich and abundant extracellular matrix phenotype; and a unique, highly plastic, and multipotent resident cell population. METHODS: A total of 25 human stenotic aortic valves obtained from valve replacement surgeries were analyzed by multiple modalities, including transcriptomics and global unlabeled and label-based tandem-mass-tagged proteomics. Segmentation of valves into disease stage-specific samples was guided by near-infrared molecular imaging, and anatomic layer-specificity was facilitated by laser capture microdissection. Side-specific cell cultures were subjected to multiple calcifying stimuli, and their calcification potential and basal/stimulated proteomes were evaluated. Molecular (protein-protein) interaction networks were built, and their central proteins and disease associations were identified. RESULTS: Global transcriptional and protein expression signatures differed between the nondiseased, fibrotic, and calcific stages of CAVD. Anatomic aortic valve microlayers exhibited unique proteome profiles that were maintained throughout disease progression and identified glial fibrillary acidic protein as a specific marker of valvular interstitial cells from the spongiosa layer. CAVD disease progression was marked by an emergence of smooth muscle cell activation, inflammation, and calcification-related pathways. Proteins overrepresented in the disease-prone fibrosa are functionally annotated to fibrosis and calcification pathways, and we found that in vitro, fibrosa-derived valvular interstitial cells demonstrated greater calcification potential than those from the ventricularis. These studies confirmed that the microlayer-specific proteome was preserved in cultured valvular interstitial cells, and that valvular interstitial cells exposed to alkaline phosphatase-dependent and alkaline phosphatase-independent calcifying stimuli had distinct proteome profiles, both of which overlapped with that of the whole tissue. Analysis of protein-protein interaction networks found a significant closeness to multiple inflammatory and fibrotic diseases. CONCLUSIONS: A spatially and temporally resolved multi-omics, and network and systems biology strategy identifies the first molecular regulatory networks in CAVD, a cardiac condition without a pharmacological cure, and describes a novel means of systematic disease ontology that is broadly applicable to comprehensive omics studies of cardiovascular diseases.


Assuntos
Estenose da Valva Aórtica/genética , Valva Aórtica/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Mapas de Interação de Proteínas , Proteômica/métodos , Espectrometria de Massas em Tandem , Transcriptoma , Valva Aórtica/patologia , Estenose da Valva Aórtica/metabolismo , Estenose da Valva Aórtica/patologia , Estudos de Casos e Controles , Células Cultivadas , Fibrose , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Índice de Gravidade de Doença , Transdução de Sinais/genética
12.
Nature ; 552(7683): 110-115, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29160304

RESUMO

Fibrosis is a common pathology in cardiovascular disease. In the heart, fibrosis causes mechanical and electrical dysfunction and in the kidney, it predicts the onset of renal failure. Transforming growth factor ß1 (TGFß1) is the principal pro-fibrotic factor, but its inhibition is associated with side effects due to its pleiotropic roles. We hypothesized that downstream effectors of TGFß1 in fibroblasts could be attractive therapeutic targets and lack upstream toxicity. Here we show, using integrated imaging-genomics analyses of primary human fibroblasts, that upregulation of interleukin-11 (IL-11) is the dominant transcriptional response to TGFß1 exposure and required for its pro-fibrotic effect. IL-11 and its receptor (IL11RA) are expressed specifically in fibroblasts, in which they drive non-canonical, ERK-dependent autocrine signalling that is required for fibrogenic protein synthesis. In mice, fibroblast-specific Il11 transgene expression or Il-11 injection causes heart and kidney fibrosis and organ failure, whereas genetic deletion of Il11ra1 protects against disease. Therefore, inhibition of IL-11 prevents fibroblast activation across organs and species in response to a range of important pro-fibrotic stimuli. These results reveal a central role of IL-11 in fibrosis and we propose that inhibition of IL-11 is a potential therapeutic strategy to treat fibrotic diseases.


Assuntos
Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patologia , Fibrose/metabolismo , Fibrose/patologia , Interleucina-11/metabolismo , Animais , Comunicação Autócrina , Células Cultivadas , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/induzido quimicamente , Coração , Humanos , Interleucina-11/antagonistas & inibidores , Interleucina-11/genética , Subunidade alfa de Receptor de Interleucina-11/deficiência , Subunidade alfa de Receptor de Interleucina-11/genética , Rim/patologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Miocárdio/metabolismo , Miocárdio/patologia , Escores de Disfunção Orgânica , Biossíntese de Proteínas , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Transgenes/genética
13.
Proc Natl Acad Sci U S A ; 114(43): E9096-E9104, 2017 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-29073106

RESUMO

Comparative analyses of transcriptional profiles from humans and mice with cardiovascular pathologies revealed consistently elevated expression of MICU2, a regulatory subunit of the mitochondrial calcium uniporter complex. To determine if MICU2 expression was cardioprotective, we produced and characterized Micu2-/- mice. Mutant mice had left atrial enlargement and Micu2-/- cardiomyocytes had delayed sarcomere relaxation and cytosolic calcium reuptake kinetics, indicating diastolic dysfunction. RNA sequencing (RNA-seq) of Micu2-/- ventricular tissues revealed markedly reduced transcripts encoding the apelin receptor (Micu2-/- vs. wild type, P = 7.8 × 10-40), which suppresses angiotensin II receptor signaling via allosteric transinhibition. We found that Micu2-/- and wild-type mice had comparable basal blood pressures and elevated responses to angiotensin II infusion, but that Micu2-/- mice exhibited systolic dysfunction and 30% lethality from abdominal aortic rupture. Aneurysms and rupture did not occur with norepinephrine-induced hypertension. Aortic tissue from Micu2-/- mice had increased expression of extracellular matrix remodeling genes, while single-cell RNA-seq analyses showed increased expression of genes related to reactive oxygen species, inflammation, and proliferation in fibroblast and smooth muscle cells. We concluded that Micu2-/- mice recapitulate features of diastolic heart disease and define previously unappreciated roles for Micu2 in regulating angiotensin II-mediated hypertensive responses that are critical in protecting the abdominal aorta from injury.


Assuntos
Canais de Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Cardiomiopatia Hipertrófica Familiar/genética , Angiotensina Amida/genética , Angiotensina II/farmacologia , Animais , Aorta Abdominal/patologia , Canais de Cálcio/genética , Proteínas de Ligação ao Cálcio/genética , Cardiomiopatia Hipertrófica Familiar/patologia , Eletrocardiografia , Regulação da Expressão Gênica , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mitocôndrias Hepáticas/fisiologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia
14.
Dev Cell ; 39(4): 480-490, 2016 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-27840107

RESUMO

Activation of complex molecular programs in specific cell lineages governs mammalian heart development, from a primordial linear tube to a four-chamber organ. To characterize lineage-specific, spatiotemporal developmental programs, we performed single-cell RNA sequencing of >1,200 murine cells isolated at seven time points spanning embryonic day 9.5 (primordial heart tube) to postnatal day 21 (mature heart). Using unbiased transcriptional data, we classified cardiomyocytes, endothelial cells, and fibroblast-enriched cells, thus identifying markers for temporal and chamber-specific developmental programs. By harnessing these datasets, we defined developmental ages of human and mouse pluripotent stem-cell-derived cardiomyocytes and characterized lineage-specific maturation defects in hearts of mice with heterozygous mutations in Nkx2.5 that cause human heart malformations. This spatiotemporal transcriptome analysis of heart development reveals lineage-specific gene programs underlying normal cardiac development and congenital heart disease.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Análise de Célula Única/métodos , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Haploinsuficiência/genética , Átrios do Coração/citologia , Ventrículos do Coração/citologia , Proteína Homeobox Nkx-2.5/metabolismo , Humanos , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Análise de Sequência de RNA , Fatores de Tempo , Transcriptoma/genética
15.
PLoS One ; 11(8): e0159710, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27505173

RESUMO

The epicardium plays an important role in coronary vessel formation and Tgfbr3-/- mice exhibit failed coronary vessel development associated with decreased epicardial cell invasion. Immortalized Tgfbr3-/- epicardial cells display the same defects. Tgfbr3+/+ and Tgfbr3-/- cells incubated for 72 hours with VEH or ligands known to promote invasion via TGFßR3 (TGFß1, TGFß2, BMP2), for 72 hours were harvested for RNA-seq analysis. We selected for genes >2-fold differentially expressed between Tgfbr3+/+ and Tgfbr3-/- cells when incubated with VEH (604), TGFß1 (515), TGFß2 (553), or BMP2 (632). Gene Ontology (GO) analysis of these genes identified dysregulated biological processes consistent with the defects observed in Tgfbr3-/- cells, including those associated with extracellular matrix interaction. GO and Gene Regulatory Network (GRN) analysis identified distinct expression profiles between TGFß1-TGFß2 and VEH-BMP2 incubated cells, consistent with the differential response of epicardial cells to these ligands in vitro. Despite the differences observed between Tgfbr3+/+ and Tgfbr3-/- cells after TGFß and BMP ligand addition, GRNs constructed from these gene lists identified NF-ĸB as a key nodal point for all ligands examined. Tgfbr3-/- cells exhibited decreased expression of genes known to be activated by NF-ĸB signaling. NF-ĸB activity was stimulated in Tgfbr3+/+ epicardial cells after TGFß2 or BMP2 incubation, while Tgfbr3-/- cells failed to activate NF-ĸB in response to these ligands. Tgfbr3+/+ epicardial cells incubated with an inhibitor of NF-ĸB signaling no longer invaded into a collagen gel in response to TGFß2 or BMP2. These data suggest that NF-ĸB signaling is dysregulated in Tgfbr3-/- epicardial cells and that NF-ĸB signaling is required for epicardial cell invasion in vitro. Our approach successfully identified a signaling pathway important in epicardial cell behavior downstream of TGFßR3. Overall, the genes and signaling pathways identified through our analysis yield the first comprehensive list of candidate genes whose expression is dependent on TGFßR3 signaling.


Assuntos
Pericárdio/citologia , Pericárdio/embriologia , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Transcriptoma , Animais , Linhagem Celular , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteoglicanas/deficiência , Receptores de Fatores de Crescimento Transformadores beta/deficiência , Análise de Sequência de RNA
16.
Biomaterials ; 35(9): 2809-15, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24433835

RESUMO

Epithelial-to-mesenchymal transition (EMT) of endocardial cells is a critical initial step in the formation of heart valves. The collagen gel in vitro model has provided significant information on the role of growth factors regulating EMT but has not permitted investigation of mechanical factors. Therefore we sought to develop a system to probe the effects of mechanical inputs on endocardial EMT by incorporating hyaluronic acid (HA), the primary component of endocardial cushions in developing heart valves, into the gel assay. This was achieved using a combination collagen and crosslinkable methacrylated HA hydrogel (Coll-MeHA). Avian atrioventricular canal explants on Coll-MeHA gels showed increased numbers of transformed cells. Analysis of the mechanical properties of Coll-MeHA gels shows that stiffness does not directly affect EMT. Hydrogel deformation from the beating myocardium of explants directly led to higher levels of regional gel deformation and larger average strain magnitudes associated with invaded cells on Coll-MeHA gels. Inhibition of this contraction reduced EMT on all gel types, although to a lesser extent on Coll-MeHA gels. Using the system we have developed, which permits the manipulation of mechanical factors, we have demonstrated that active mechanical forces play a role in the regulation of endocardial EMT.


Assuntos
Endocárdio/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Ácido Hialurônico/farmacologia , Mecanotransdução Celular/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Galinhas , Colágeno Tipo I/farmacologia , Reagentes de Ligações Cruzadas/farmacologia , Metacrilatos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tecidos Suporte/química
17.
J Mol Cell Cardiol ; 59: 196-204, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23557753

RESUMO

Valvular Interstitial Cells (VICs) are a common substrate for congenital and adult heart disease yet the signaling mechanisms governing their formation during early valvulogenesis are incompletely understood. We developed an unbiased strategy to identify genes important in endocardial epithelial-to-mesenchymal transformation (EMT) using a spatial transcriptional profile. Endocardial cells overlaying the cushions of the atrioventricular canal (AVC) and outflow tract (OFT) undergo an EMT to yield VICs. RNA sequencing (RNA-seq) analysis of gene expression between AVC, OFT, and ventricles (VEN) isolated from chick and mouse embryos at comparable stages of development (chick HH18; mouse E11.0) was performed. EMT occurs in the AVC and OFT cushions, but not VEN at this time. 198 genes in the chick (n=1) and 105 genes in the mouse (n=2) were enriched 2-fold in the cushions. Gene regulatory networks (GRN) generated from cushion-enriched gene lists confirmed TGFß as a nodal point and identified NF-κB as a potential node. To reveal previously unrecognized regulators of EMT four candidate genes, Hapln1, Id1, Foxp2, and Meis2, and a candidate pathway, NF-κB, were selected. In vivo spatial expression of each gene was confirmed by in situ hybridization and a functional role for each in endocardial EMT was determined by siRNA knockdown in a collagen gel assay. Our spatial-transcriptional profiling strategy yielded gene lists which reflected the known biology of the system. Further analysis accurately identified and validated previously unrecognized novel candidate genes and the NF-κB pathway as regulators of endocardial cell EMT in vitro.


Assuntos
Coxins Endocárdicos/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Miocárdio/metabolismo , Animais , Galinhas , Transição Epitelial-Mesenquimal/genética , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Camundongos , Proteoglicanas/genética , Proteoglicanas/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Análise de Sequência de RNA
18.
Cell Signal ; 24(1): 247-56, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21945156

RESUMO

An early event in heart valve formation is the epithelial-mesenchymal transformation (EMT) of a subpopulation of endothelial cells in specific regions of the heart tube, the endocardial cushions. The Type III TGFß receptor (TGFßR3) is required for TGFß2- or BMP-2-stimulated EMT in atrioventricular endocardial cushion (AVC) explants in vitro but the mediators downstream of TGFßR3 are not well described. Using AVC and ventricular explants as an in vitro assay, we found an absolute requirement for specific TGFßR3 cytoplasmic residues, GAIP-interacting protein, C terminus (GIPC), and specific Activin Receptor-Like Kinases (ALK)s for TGFßR3-mediated EMT when stimulated by TGFß2 or BMP-2. The introduction of TGFßR3 into nontransforming ventricular endocardial cells, followed by the addition of either TGFß2 or BMP-2, results in EMT. TGFßR3 lacking the entire cytoplasmic domain, or only the 3C-terminal amino acids that are required to bind GIPC, fails to support EMT in response to TGFß2 or BMP-2. Overexpression of GIPC in AVC endocardial cells enhanced EMT while siRNA-mediated silencing of GIPC in ventricular cells overexpressing TGFßR3 significantly inhibited EMT. Targeting of specific ALKs by siRNA revealed that TGFßR3-mediated EMT requires ALK2 and ALK3, in addition to ALK5, but not ALK4 or ALK6. Taken together, these data identify GIPC, ALK2, ALK3, and ALK5 as signaling components required for TGFßR3-mediated endothelial cell EMT.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Coxins Endocárdicos/fisiologia , Transição Epitelial-Mesenquimal , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Ativinas/metabolismo , Sequência de Aminoácidos , Animais , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 2/fisiologia , Embrião de Galinha , Coxins Endocárdicos/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Proteínas de Fluorescência Verde/biossíntese , Dados de Sequência Molecular , Domínios e Motivos de Interação entre Proteínas , Proteoglicanas/química , Receptores de Fatores de Crescimento Transformadores beta/química , Proteínas Recombinantes/biossíntese , Técnicas de Cultura de Tecidos , Fator de Crescimento Transformador beta2/farmacologia , Fator de Crescimento Transformador beta2/fisiologia
19.
Birth Defects Res A Clin Mol Teratol ; 91(6): 511-25, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21538818

RESUMO

Specific cell and tissue interactions drive the formation and function of the vertebrate cardiovascular system. Although much attention has been focused on the muscular components of the developing heart, the endocardium plays a key role in the formation of a functioning heart. Endocardial cells exhibit heterogeneity that allows them to participate in events such as the formation of the valves, septation of the outflow tract, and trabeculation. Here we review, the contributions of the endocardium to cardiovascular development and outline useful approaches developed in the chick and mouse that have revealed endocardial cell heterogeneity, the signaling molecules that direct endocardial cell behavior, and how these insights have contributed to our understanding of cardiovascular development and disease.


Assuntos
Modelos Animais de Doenças , Endocárdio/embriologia , Cardiopatias Congênitas/metabolismo , Animais , Embrião de Galinha , Endocárdio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Cardiopatias Congênitas/genética , Camundongos , Transdução de Sinais
20.
Dev Dyn ; 239(12): 3226-34, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20960543

RESUMO

Disease or malformation of heart valves is one of the leading causes of morbidity and mortality in both children and adults. These congenital anomalies can remain undetected until cardiac function is compromised, making it important to understand the underlying nature of these disorders. Here we show that ephrin-A1, a ligand for class A Eph receptor tyrosine kinases, regulates cardiac valve formation. Exogenous ephrin-A1-Fc or overexpression of ephrin-A1 in the heart inhibits epithelial-to-mesenchymal transformation (EMT) in chick atrioventricular cushion explants. In contrast, overexpression of wild-type EphA3 receptor promotes EMT via a kinase-dependent mechanism. To analyze ephrin-A1 in vivo, we generated an ephrin-A1 knockout mouse through gene targeting. Ephrin-A1 null animals are viable but exhibit impaired cardiac function. Loss of ephrin-A1 results in thickened aortic and mitral valves in newborn and adult animals. Analysis of early embryonic hearts revealed increased cellularity in outflow tract endocardial cushions and elevated mesenchymal marker expression, suggesting that excessive numbers of cells undergo EMT. Taken together, these data indicate that ephrin-A1 regulates cardiac valve development, making ephrin-A1-deficient mice a novel model for congenital heart defects.


Assuntos
Efrina-A1/metabolismo , Valvas Cardíacas/embriologia , Coração/embriologia , Morfogênese/fisiologia , Animais , Ecocardiografia , Efrina-A1/genética , Feminino , Masculino , Camundongos , Camundongos Knockout , Morfogênese/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...