Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 595(7867): 438-443, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34163071

RESUMEN

In diseased organs, stress-activated signalling cascades alter chromatin, thereby triggering maladaptive cell state transitions. Fibroblast activation is a common stress response in tissues that worsens lung, liver, kidney and heart disease, yet its mechanistic basis remains unclear1,2. Pharmacological inhibition of bromodomain and extra-terminal domain (BET) proteins alleviates cardiac dysfunction3-7, providing a tool to interrogate and modulate cardiac cell states as a potential therapeutic approach. Here we use single-cell epigenomic analyses of hearts dynamically exposed to BET inhibitors to reveal a reversible transcriptional switch that underlies the activation of fibroblasts. Resident cardiac fibroblasts demonstrated robust toggling between the quiescent and activated state in a manner directly correlating with BET inhibitor exposure and cardiac function. Single-cell chromatin accessibility revealed previously undescribed DNA elements, the accessibility of which dynamically correlated with cardiac performance. Among the most dynamic elements was an enhancer that regulated the transcription factor MEOX1, which was specifically expressed in activated fibroblasts, occupied putative regulatory elements of a broad fibrotic gene program and was required for TGFß-induced fibroblast activation. Selective CRISPR inhibition of the single most dynamic cis-element within the enhancer blocked TGFß-induced Meox1 activation. We identify MEOX1 as a central regulator of fibroblast activation associated with cardiac dysfunction and demonstrate its upregulation after activation of human lung, liver and kidney fibroblasts. The plasticity and specificity of BET-dependent regulation of MEOX1 in tissue fibroblasts provide previously unknown trans- and cis-targets for treating fibrotic disease.


Asunto(s)
Elementos de Facilitación Genéticos , Fibroblastos/citología , Cardiopatías/genética , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Animales , Cromatina/metabolismo , Epigenómica , Regulación de la Expresión Génica , Humanos , Ratones , Proteínas/antagonistas & inhibidores , Análisis de la Célula Individual , Transcriptoma , Factor de Crecimiento Transformador beta/metabolismo
2.
Development ; 150(9)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36994838

RESUMEN

Transcriptional networks governing cardiac precursor cell (CPC) specification are incompletely understood owing, in part, to limitations in distinguishing CPCs from non-cardiac mesoderm in early gastrulation. We leveraged detection of early cardiac lineage transgenes within a granular single-cell transcriptomic time course of mouse embryos to identify emerging CPCs and describe their transcriptional profiles. Mesp1, a transiently expressed mesodermal transcription factor, is canonically described as an early regulator of cardiac specification. However, we observed perdurance of CPC transgene-expressing cells in Mesp1 mutants, albeit mislocalized, prompting us to investigate the scope of the role of Mesp1 in CPC emergence and differentiation. Mesp1 mutant CPCs failed to robustly activate markers of cardiomyocyte maturity and crucial cardiac transcription factors, yet they exhibited transcriptional profiles resembling cardiac mesoderm progressing towards cardiomyocyte fates. Single-cell chromatin accessibility analysis defined a Mesp1-dependent developmental breakpoint in cardiac lineage progression at a shift from mesendoderm transcriptional networks to those necessary for cardiac patterning and morphogenesis. These results reveal Mesp1-independent aspects of early CPC specification and underscore a Mesp1-dependent regulatory landscape required for progression through cardiogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Epigenómica , Miocitos Cardíacos , Animales , Ratones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Mesodermo/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo
3.
Nature ; 572(7767): 120-124, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31341279

RESUMEN

Organogenesis involves integration of diverse cell types; dysregulation of cell-type-specific gene networks results in birth defects, which affect 5% of live births. Congenital heart defects are the most common malformations, and result from disruption of discrete subsets of cardiac progenitor cells1, but the transcriptional changes in individual progenitors that lead to organ-level defects remain unknown. Here we used single-cell RNA sequencing to interrogate early cardiac progenitor cells as they become specified during normal and abnormal cardiogenesis, revealing how dysregulation of specific cellular subpopulations has catastrophic consequences. A network-based computational method for single-cell RNA-sequencing analysis that predicts lineage-specifying transcription factors2,3 identified Hand2 as a specifier of outflow tract cells but not right ventricular cells, despite the failure of right ventricular formation in Hand2-null mice4. Temporal single-cell-transcriptome analysis of Hand2-null embryos revealed failure of outflow tract myocardium specification, whereas right ventricular myocardium was specified but failed to properly differentiate and migrate. Loss of Hand2 also led to dysregulation of retinoic acid signalling and disruption of anterior-posterior patterning of cardiac progenitors. This work reveals transcriptional determinants that specify fate and differentiation in individual cardiac progenitor cells, and exposes mechanisms of disrupted cardiac development at single-cell resolution, providing a framework for investigating congenital heart defects.


Asunto(s)
Cardiopatías Congénitas/embriología , Cardiopatías Congénitas/patología , Corazón/embriología , Análisis de la Célula Individual , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Movimiento Celular , Análisis por Conglomerados , Femenino , Cardiopatías Congénitas/genética , Masculino , Ratones , Análisis de Secuencia de ARN , Tretinoina/metabolismo
4.
Nature ; 541(7636): 176-181, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28002412

RESUMEN

Respiratory dysfunction is a notorious cause of perinatal mortality in infants and sleep apnoea in adults, but the mechanisms of respiratory control are not clearly understood. Mechanical signals transduced by airway-innervating sensory neurons control respiration; however, the physiological significance and molecular mechanisms of these signals remain obscured. Here we show that global and sensory neuron-specific ablation of the mechanically activated ion channel Piezo2 causes respiratory distress and death in newborn mice. Optogenetic activation of Piezo2+ vagal sensory neurons causes apnoea in adult mice. Moreover, induced ablation of Piezo2 in sensory neurons of adult mice causes decreased neuronal responses to lung inflation, an impaired Hering-Breuer mechanoreflex, and increased tidal volume under normal conditions. These phenotypes are reproduced in mice lacking Piezo2 in the nodose ganglion. Our data suggest that Piezo2 is an airway stretch sensor and that Piezo2-mediated mechanotransduction within various airway-innervating sensory neurons is critical for establishing efficient respiration at birth and maintaining normal breathing in adults.


Asunto(s)
Apnea/fisiopatología , Canales Iónicos/metabolismo , Pulmón/fisiología , Pulmón/fisiopatología , Mecanotransducción Celular/fisiología , Reflejo/fisiología , Animales , Animales Recién Nacidos , Apnea/genética , Muerte , Femenino , Canales Iónicos/deficiencia , Canales Iónicos/genética , Masculino , Mecanotransducción Celular/genética , Ratones , Ganglio Nudoso/metabolismo , Reflejo/genética , Respiración , Células Receptoras Sensoriales/metabolismo , Volumen de Ventilación Pulmonar
5.
Proc Natl Acad Sci U S A ; 115(50): 12817-12822, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30482854

RESUMEN

PIEZO1 is a cation channel that is activated by mechanical forces such as fluid shear stress or membrane stretch. PIEZO1 loss-of-function mutations in patients are associated with congenital lymphedema with pleural effusion. However, the mechanistic link between PIEZO1 function and the development or function of the lymphatic system is currently unknown. Here, we analyzed two mouse lines lacking PIEZO1 in endothelial cells (via Tie2Cre or Lyve1Cre) and found that they exhibited pleural effusion and died postnatally. Strikingly, the number of lymphatic valves was dramatically reduced in these mice. Lymphatic valves are essential for ensuring proper circulation of lymph. Mechanical forces have been implicated in the development of lymphatic vasculature and valve formation, but the identity of mechanosensors involved is unknown. Expression of FOXC2 and NFATc1, transcription factors known to be required for lymphatic valve development, appeared normal in Tie2Cre;Piezo1cKO mice. However, the process of protrusion in the valve leaflets, which is associated with collective cell migration, actin polymerization, and remodeling of cell-cell junctions, was impaired in Tie2Cre;Piezo1cKO mice. Consistent with these genetic findings, activation of PIEZO1 by Yoda1 in cultured lymphatic endothelial cells induced active remodeling of actomyosin and VE-cadherin+ cell-cell adhesion sites. Our analysis provides evidence that mechanically activated ion channel PIEZO1 is a key regulator of lymphatic valve formation.


Asunto(s)
Canales Iónicos/metabolismo , Linfangiogénesis/fisiología , Sistema Linfático/metabolismo , Sistema Linfático/fisiología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/fisiología , Actomiosina/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Factores de Transcripción Forkhead/metabolismo , Uniones Intercelulares/metabolismo , Uniones Intercelulares/fisiología , Transporte Iónico/fisiología , Ratones , Factores de Transcripción NFATC/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
6.
Nature ; 516(7529): 121-5, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25471886

RESUMEN

The sense of touch provides critical information about our physical environment by transforming mechanical energy into electrical signals. It is postulated that mechanically activated cation channels initiate touch sensation, but the identity of these molecules in mammals has been elusive. Piezo2 is a rapidly adapting, mechanically activated ion channel expressed in a subset of sensory neurons of the dorsal root ganglion and in cutaneous mechanoreceptors known as Merkel-cell-neurite complexes. It has been demonstrated that Merkel cells have a role in vertebrate mechanosensation using Piezo2, particularly in shaping the type of current sent by the innervating sensory neuron; however, major aspects of touch sensation remain intact without Merkel cell activity. Here we show that mice lacking Piezo2 in both adult sensory neurons and Merkel cells exhibit a profound loss of touch sensation. We precisely localize Piezo2 to the peripheral endings of a broad range of low-threshold mechanoreceptors that innervate both hairy and glabrous skin. Most rapidly adapting, mechanically activated currents in dorsal root ganglion neuronal cultures are absent in Piezo2 conditional knockout mice, and ex vivo skin nerve preparation studies show that the mechanosensitivity of low-threshold mechanoreceptors strongly depends on Piezo2. This cellular phenotype correlates with an unprecedented behavioural phenotype: an almost complete deficit in light-touch sensation in multiple behavioural assays, without affecting other somatosensory functions. Our results highlight that a single ion channel that displays rapidly adapting, mechanically activated currents in vitro is responsible for the mechanosensitivity of most low-threshold mechanoreceptor subtypes involved in innocuous touch sensation. Notably, we find that touch and pain sensation are separable, suggesting that as-yet-unknown mechanically activated ion channel(s) must account for noxious (painful) mechanosensation.


Asunto(s)
Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Piel/inervación , Tacto/fisiología , Animales , Canales Iónicos/genética , Mecanorreceptores/metabolismo , Mecanotransducción Celular/genética , Células de Merkel/fisiología , Ratones , Ratones Noqueados , Células Receptoras Sensoriales/fisiología , Tacto/genética
7.
Proc Natl Acad Sci U S A ; 111(28): 10347-52, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24958852

RESUMEN

Mechanosensation is perhaps the last sensory modality not understood at the molecular level. Ion channels that sense mechanical force are postulated to play critical roles in a variety of biological processes including sensing touch/pain (somatosensation), sound (hearing), and shear stress (cardiovascular physiology); however, the identity of these ion channels has remained elusive. We previously identified Piezo1 and Piezo2 as mechanically activated cation channels that are expressed in many mechanosensitive cell types. Here, we show that Piezo1 is expressed in endothelial cells of developing blood vessels in mice. Piezo1-deficient embryos die at midgestation with defects in vascular remodeling, a process critically influenced by blood flow. We demonstrate that Piezo1 is activated by shear stress, the major type of mechanical force experienced by endothelial cells in response to blood flow. Furthermore, loss of Piezo1 in endothelial cells leads to deficits in stress fiber and cellular orientation in response to shear stress, linking Piezo1 mechanotransduction to regulation of cell morphology. These findings highlight an essential role of mammalian Piezo1 in vascular development during embryonic development.


Asunto(s)
Sistema Cardiovascular/embriología , Desarrollo Embrionario/fisiología , Células Endoteliales/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Animales , Sistema Cardiovascular/citología , Células Endoteliales/citología , Canales Iónicos/genética , Ratones , Ratones Transgénicos
8.
Science ; 371(6530)2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33303684

RESUMEN

Mapping the gene-regulatory networks dysregulated in human disease would allow the design of network-correcting therapies that treat the core disease mechanism. However, small molecules are traditionally screened for their effects on one to several outputs at most, biasing discovery and limiting the likelihood of true disease-modifying drug candidates. Here, we developed a machine-learning approach to identify small molecules that broadly correct gene networks dysregulated in a human induced pluripotent stem cell (iPSC) disease model of a common form of heart disease involving the aortic valve (AV). Gene network correction by the most efficacious therapeutic candidate, XCT790, generalized to patient-derived primary AV cells and was sufficient to prevent and treat AV disease in vivo in a mouse model. This strategy, made feasible by human iPSC technology, network analysis, and machine learning, may represent an effective path for drug discovery.


Asunto(s)
Enfermedad de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Válvula Aórtica/patología , Calcinosis/tratamiento farmacológico , Redes Reguladoras de Genes/efectos de los fármacos , Aprendizaje Automático , Nitrilos/farmacología , Nitrilos/uso terapéutico , Tiazoles/farmacología , Tiazoles/uso terapéutico , Algoritmos , Animales , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/metabolismo , Válvula Aórtica/fisiopatología , Enfermedad de la Válvula Aórtica/genética , Enfermedad de la Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/fisiopatología , Calcinosis/genética , Calcinosis/fisiopatología , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Haploinsuficiencia , Humanos , Células Madre Pluripotentes Inducidas , Ratones Endogámicos C57BL , RNA-Seq , Receptor Notch1/genética , Bibliotecas de Moléculas Pequeñas
9.
Science ; 364(6443): 865-870, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31147515

RESUMEN

Complex genetic mechanisms are thought to underlie many human diseases, yet experimental proof of this model has been elusive. Here, we show that a human cardiac anomaly can be caused by a combination of rare, inherited heterozygous mutations. Whole-exome sequencing of a nuclear family revealed that three offspring with childhood-onset cardiomyopathy had inherited three missense single-nucleotide variants in the MKL2, MYH7, and NKX2-5 genes. The MYH7 and MKL2 variants were inherited from the affected, asymptomatic father and the rare NKX2-5 variant (minor allele frequency, 0.0012) from the unaffected mother. We used CRISPR-Cas9 to generate mice encoding the orthologous variants and found that compound heterozygosity for all three variants recapitulated the human disease phenotype. Analysis of murine hearts and human induced pluripotent stem cell-derived cardiomyocytes provided histologic and molecular evidence for the NKX2-5 variant's contribution as a genetic modifier.


Asunto(s)
Cardiomiopatías/genética , Heterocigoto , Proteína Homeótica Nkx-2.5/genética , Herencia Multifactorial , Factor Nuclear Tiroideo 1/genética , Animales , Proteína 9 Asociada a CRISPR , Miosinas Cardíacas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Exoma , Frecuencia de los Genes , Humanos , Células Madre Pluripotentes Inducidas , Ratones , Ratones Mutantes , Mutación Missense , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Herencia Paterna/genética , Factores de Transcripción/genética
10.
J Clin Invest ; 127(5): 1683-1688, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28346225

RESUMEN

Diseases caused by gene haploinsufficiency in humans commonly lack a phenotype in mice that are heterozygous for the orthologous factor, impeding the study of complex phenotypes and critically limiting the discovery of therapeutics. Laboratory mice have longer telomeres relative to humans, potentially protecting against age-related disease caused by haploinsufficiency. Here, we demonstrate that telomere shortening in NOTCH1-haploinsufficient mice is sufficient to elicit age-dependent cardiovascular disease involving premature calcification of the aortic valve, a phenotype that closely mimics human disease caused by NOTCH1 haploinsufficiency. Furthermore, progressive telomere shortening correlated with severity of disease, causing cardiac valve and septal disease in the neonate that was similar to the range of valve disease observed within human families. Genes that were dysregulated due to NOTCH1 haploinsufficiency in mice with shortened telomeres were concordant with proosteoblast and proinflammatory gene network alterations in human NOTCH1 heterozygous endothelial cells. These dysregulated genes were enriched for telomere-contacting promoters, suggesting a potential mechanism for telomere-dependent regulation of homeostatic gene expression. These findings reveal a critical role for telomere length in a mouse model of age-dependent human disease and provide an in vivo model in which to test therapeutic candidates targeting the progression of aortic valve disease.


Asunto(s)
Envejecimiento , Haploinsuficiencia , Defectos de los Tabiques Cardíacos , Enfermedades de las Válvulas Cardíacas , Receptor Notch1 , Homeostasis del Telómero/genética , Telómero , Envejecimiento/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Defectos de los Tabiques Cardíacos/genética , Defectos de los Tabiques Cardíacos/metabolismo , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Ratones , Ratones Mutantes , Regiones Promotoras Genéticas , Receptor Notch1/genética , Receptor Notch1/metabolismo , Telómero/genética , Telómero/metabolismo
11.
Neuron ; 87(6): 1162-1179, 2015 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-26402601

RESUMEN

Mechanotransduction, the conversion of physical forces into biochemical signals, is essential for various physiological processes such as the conscious sensations of touch and hearing, and the unconscious sensation of blood flow. Mechanically activated (MA) ion channels have been proposed as sensors of physical force, but the identity of these channels and an understanding of how mechanical force is transduced has remained elusive. A number of recent studies on previously known ion channels along with the identification of novel MA ion channels have greatly transformed our understanding of touch and hearing in both vertebrates and invertebrates. Here, we present an updated review of eukaryotic ion channel families that have been implicated in mechanotransduction processes and evaluate the qualifications of the candidate genes according to specified criteria. We then discuss the proposed gating models for MA ion channels and highlight recent structural studies of mechanosensitive potassium channels.


Asunto(s)
Activación del Canal Iónico/fisiología , Canales Iónicos/fisiología , Transporte Iónico/fisiología , Mecanotransducción Celular/fisiología , Sensación/fisiología , Animales , Humanos , Tacto/fisiología
12.
Elife ; 42015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-26001274

RESUMEN

Red blood cells (RBCs) experience significant mechanical forces while recirculating, but the consequences of these forces are not fully understood. Recent work has shown that gain-of-function mutations in mechanically activated Piezo1 cation channels are associated with the dehydrating RBC disease xerocytosis, implicating a role of mechanotransduction in RBC volume regulation. However, the mechanisms by which these mutations result in RBC dehydration are unknown. In this study, we show that RBCs exhibit robust calcium entry in response to mechanical stretch and that this entry is dependent on Piezo1 expression. Furthermore, RBCs from blood-cell-specific Piezo1 conditional knockout mice are overhydrated and exhibit increased fragility both in vitro and in vivo. Finally, we show that Yoda1, a chemical activator of Piezo1, causes calcium influx and subsequent dehydration of RBCs via downstream activation of the KCa3.1 Gardos channel, directly implicating Piezo1 signaling in RBC volume control. Therefore, mechanically activated Piezo1 plays an essential role in RBC volume homeostasis.


Asunto(s)
Calcio/metabolismo , Eritrocitos/fisiología , Canales Iónicos/fisiología , Mecanotransducción Celular/fisiología , Análisis de Varianza , Animales , Fenómenos Biomecánicos , Western Blotting , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Recuento de Eritrocitos , Eritrocitos/metabolismo , Eritrocitos/ultraestructura , Citometría de Flujo , Fluorescencia , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Mutación/genética , Bibliotecas de Moléculas Pequeñas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA