Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(43): 27022-27033, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33051294

RESUMEN

The essential function of the circulatory system is to continuously and efficiently supply the O2 and nutrients necessary to meet the metabolic demands of every cell in the body, a function in which vast capillary networks play a key role. Capillary networks serve an additional important function in the central nervous system: acting as a sensory network, they detect neuronal activity in the form of elevated extracellular K+ and initiate a retrograde, propagating, hyperpolarizing signal that dilates upstream arterioles to rapidly increase local blood flow. Yet, little is known about how blood entering this network is distributed on a branch-to-branch basis to reach specific neurons in need. Here, we demonstrate that capillary-enwrapping projections of junctional, contractile pericytes within a postarteriole transitional region differentially constrict to structurally and dynamically determine the morphology of capillary junctions and thereby regulate branch-specific blood flow. We further found that these contractile pericytes are capable of receiving propagating K+-induced hyperpolarizing signals propagating through the capillary network and dynamically channeling red blood cells toward the initiating signal. By controlling blood flow at junctions, contractile pericytes within a functionally distinct postarteriole transitional region maintain the efficiency and effectiveness of the capillary network, enabling optimal perfusion of the brain.


Asunto(s)
Capilares/fisiología , Circulación Cerebrovascular , Microcirculación , Pericitos/fisiología , Animales , Arteriolas/fisiología , Canales de Calcio/metabolismo , Venas Cerebrales/fisiología , Ratones
2.
J Vasc Res ; 58(3): 159-171, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33706307

RESUMEN

INTRODUCTION: Studies in Cx40-GCaMP2 mice, which express calcium biosensor GCaMP2 in the endothelium under connexin 40 promoter, have identified the unique properties of endothelial calcium signals. However, Cx40-GCaMP2 mouse is associated with a narrow dynamic range and lack of signal in the venous endothelium. Recent studies have proposed many GCaMPs (GCaMP5/6/7/8) with improved properties although their performance in endothelium-specific calcium studies is not known. METHODS: We characterized a newly developed mouse line that constitutively expresses GCaMP8 in the endothelium under the VE-cadherin (Cdh5-GCaMP8) promoter. Calcium signals through endothelial IP3 receptors and TRP vanilloid 4 (TRPV4) ion channels were recorded in mesenteric arteries (MAs) and veins from Cdh5-GCaMP8 and Cx40-GCaMP2 mice. RESULTS: Cdh5-GCaMP8 mice showed lower baseline fluorescence intensity, higher dynamic range, and higher amplitudes of individual calcium signals than Cx40-GCaMP2 mice. Importantly, Cdh5-GCaMP8 mice enabled the first recordings of discrete calcium signals in the intact venous endothelium and revealed striking differences in IP3 receptor and TRPV4 channel calcium signals between MAs and mesenteric veins. CONCLUSION: Our findings suggest that Cdh5-GCaMP8 mice represent significant improvements in dynamic range, sensitivity for low-intensity signals, and the ability to record calcium signals in venous endothelium.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Calcio/metabolismo , Conexinas/metabolismo , Células Endoteliales/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Animales , Antígenos CD/genética , Técnicas Biosensibles , Cadherinas/genética , Proteínas de Unión al Calcio/genética , Conexinas/genética , Proteínas Fluorescentes Verdes/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Arterias Mesentéricas/citología , Arterias Mesentéricas/metabolismo , Venas Mesentéricas/citología , Venas Mesentéricas/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Regiones Promotoras Genéticas , Canales Catiónicos TRPV/metabolismo , Proteína alfa-5 de Unión Comunicante
3.
Am J Physiol Heart Circ Physiol ; 316(6): H1309-H1322, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30848676

RESUMEN

Histone proteins are elevated in the circulation after traumatic injury owing to cellular lysis and release from neutrophils. Elevated circulating histones in trauma contribute to coagulopathy and mortality through a mechanism suspected to involve endothelial cell (EC) dysfunction. However, the functional consequences of histone exposure on intact blood vessels are unknown. Here, we sought to understand the effects of clinically relevant concentrations of histones on the endothelium in intact, resistance-sized, mesenteric arteries (MAs). EC Ca2+ was measured with high spatial and temporal resolution in MAs from mice selectively expressing the EC-specific, genetically encoded ratiometric Ca2+ indicator, Cx40-GCaMP-GR, and vessel diameter was measured by edge detection. Application of purified histone protein directly to the endothelium of en face mouse and human MA preparations produced large Ca2+ signals that spread within and between ECs. Surprisingly, luminal application of histones had no effect on the diameter of pressurized arteries. Instead, after prolonged exposure (30 min), it reduced dilations to endothelium-dependent vasodilators and ultimately caused death of ~25% of ECs, as evidenced by markedly elevated cytosolic Ca2+ levels (793 ± 75 nM) and uptake of propidium iodide. Removal of extracellular Ca2+ but not depletion of intracellular Ca2+ stores prevented histone-induced Ca2+ signals. Histone-induced signals were not suppressed by transient receptor potential vanilloid 4 (TRPV4) channel inhibition (100 nM GSK2193874) or genetic ablation of TRPV4 channels or Toll-like receptor receptors. These data demonstrate that histones are robust activators of noncanonical EC Ca2+ signaling, which cause vascular dysfunction through loss of endothelium-dependent dilation in resistance-sized MAs. NEW & NOTEWORTHY We describe the first use of the endothelial cell (EC)-specific, ratiometric, genetically encoded Ca2+ indicator, Cx40-GCaMP-GR, to study the effect of histone proteins on EC Ca2+ signaling. We found that histones induce an influx of Ca2+ in ECs that does not cause vasodilation but instead causes Ca2+ overload, EC death, and vascular dysfunction in the form of lost endothelium-dependent dilation.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Histonas/toxicidad , Arterias Mesentéricas/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Animales , Presión Arterial , Muerte Celular , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Receptor Toll-Like 4/metabolismo , Resistencia Vascular
4.
Nature ; 489(7415): 322-5, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22864415

RESUMEN

Transplantation studies in mice and rats have shown that human embryonic-stem-cell-derived cardiomyocytes (hESC-CMs) can improve the function of infarcted hearts, but two critical issues related to their electrophysiological behaviour in vivo remain unresolved. First, the risk of arrhythmias following hESC-CM transplantation in injured hearts has not been determined. Second, the electromechanical integration of hESC-CMs in injured hearts has not been demonstrated, so it is unclear whether these cells improve contractile function directly through addition of new force-generating units. Here we use a guinea-pig model to show that hESC-CM grafts in injured hearts protect against arrhythmias and can contract synchronously with host muscle. Injured hearts with hESC-CM grafts show improved mechanical function and a significantly reduced incidence of both spontaneous and induced ventricular tachycardia. To assess the activity of hESC-CM grafts in vivo, we transplanted hESC-CMs expressing the genetically encoded calcium sensor, GCaMP3 (refs 4, 5). By correlating the GCaMP3 fluorescent signal with the host ECG, we found that grafts in uninjured hearts have consistent 1:1 host­graft coupling. Grafts in injured hearts are more heterogeneous and typically include both coupled and uncoupled regions. Thus, human myocardial grafts meet physiological criteria for true heart regeneration, providing support for the continued development of hESC-based cardiac therapies for both mechanical and electrical repair.


Asunto(s)
Arritmias Cardíacas/terapia , Fenómenos Electrofisiológicos , Células Madre Embrionarias/citología , Lesiones Cardíacas/fisiopatología , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/trasplante , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/fisiopatología , Calcio/análisis , Calcio/metabolismo , Estimulación Eléctrica , Colorantes Fluorescentes/análisis , Cobayas , Lesiones Cardíacas/complicaciones , Lesiones Cardíacas/patología , Humanos , Mediciones Luminiscentes , Masculino , Contracción Miocárdica/fisiología , Miocardio/citología , Miocitos Cardíacos/fisiología , Taquicardia Ventricular/etiología , Taquicardia Ventricular/fisiopatología , Taquicardia Ventricular/terapia
5.
Arterioscler Thromb Vasc Biol ; 36(8): 1627-37, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27312222

RESUMEN

OBJECTIVE: Calcific aortic valve (AoV) disease is a significant clinical problem for which the regulatory mechanisms are poorly understood. Enhanced cell-cell adhesion is a common mechanism of cellular aggregation, but its role in calcific lesion formation is not known. Cadherin-11 (Cad-11) has been associated with lesion formation in vitro, but its function during adult valve homeostasis and pathogenesis is not known. This study aims to elucidate the specific functions of Cad-11 and its downstream targets, RhoA and Sox9, in extracellular matrix remodeling and AoV calcification. APPROACH AND RESULTS: We conditionally overexpressed Cad-11 in murine heart valves using a novel double-transgenic Nfatc1(Cre);R26-Cad11(TglTg) mouse model. These mice developed hemodynamically significant aortic stenosis with prominent calcific lesions in the AoV leaflets. Cad-11 overexpression upregulated downstream targets, RhoA and Sox9, in the valve interstitial cells, causing calcification and extensive pathogenic extracellular matrix remodeling. AoV interstitial cells overexpressing Cad-11 in an osteogenic environment in vitro rapidly form calcific nodules analogous to in vivo lesions. Molecular analyses revealed upregulation of osteoblastic and myofibroblastic markers. Treatment with a Rho-associated protein kinase inhibitor attenuated nodule formation, further supporting that Cad-11-driven calcification acts through the small GTPase RhoA/Rho-associated protein kinase signaling pathway. CONCLUSIONS: This study identifies one of the underlying molecular mechanisms of heart valve calcification and demonstrates that overexpression of Cad-11 upregulates RhoA and Sox9 to induce calcification and extracellular matrix remodeling in adult AoV pathogenesis. The findings provide a potential molecular target for clinical treatment.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Cadherinas/metabolismo , Calcinosis/metabolismo , Matriz Extracelular/metabolismo , Animales , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/patología , Cadherinas/genética , Calcinosis/genética , Calcinosis/patología , Estudios de Casos y Controles , Adhesión Celular , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Matriz Extracelular/patología , Predisposición Genética a la Enfermedad , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Factor de Transcripción SOX9/metabolismo , Índice de Severidad de la Enfermedad , Fibras de Estrés/metabolismo , Fibras de Estrés/patología , Regulación hacia Arriba , Proteínas de Unión al GTP rho/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
6.
Circ Res ; 115(3): 339-47, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24871564

RESUMEN

RATIONALE: Intracellular Ca(2+) concentration ([Ca(2+)]i) is regulated and signals differently in various subcellular microdomains, which greatly enhances its second messenger versatility. In the heart, sarcoplasmic reticulum Ca(2+) release and signaling are controlled by local [Ca(2+)]i in the junctional cleft ([Ca(2+)]Cleft), the small space between sarcolemma and junctional sarcoplasmic reticulum. However, methods to measure [Ca(2+)]Cleft directly are needed. OBJECTIVE: To construct novel sensors that allow direct measurement of [Ca(2+)]Cleft. METHODS AND RESULTS: We constructed cleft-targeted [Ca(2+)] sensors by fusing Ca(2+)-sensor GCaMP2.2 and a new lower Ca(2+)-affinity variant GCaMP2.2Low to FKBP12.6, which binds with high affinity and selectivity to ryanodine receptors. The fluorescence pattern, affinity for ryanodine receptors, and competition by untagged FKBP12.6 demonstrated that FKBP12.6-tagged sensors are positioned to measure local [Ca(2+)]Cleft in adult rat myocytes. Using GCaMP2.2Low-FKBP12.6, we showed that [Ca(2+)]Cleft reaches higher levels with faster kinetics than global [Ca(2+)]i during excitation-contraction coupling. Diastolic sarcoplasmic reticulum Ca(2+) leak or sarcolemmal Ca(2+) entry may raise local [Ca(2+)]Cleft above bulk cytosolic [Ca(2+)]i ([Ca(2+)]Bulk), an effect that may contribute to triggered arrhythmias and even transcriptional regulation. We measured this diastolic standing [Ca(2+)]Cleft-[Ca(2+)]Bulk gradient with GCaMP2.2-FKBP12.6 versus GCaMP2.2, using [Ca(2+)] measured without gradients as a reference point. This diastolic difference ([Ca(2+)]Cleft=194 nmol/L versus [Ca(2+)]Bulk=100 nmol/L) is dictated mainly by the sarcoplasmic reticulum Ca(2+) leak rather than sarcolemmal Ca(2+) flux. CONCLUSIONS: We have developed junctional cleft-targeted sensors to measure [Ca(2+)]Cleft versus [Ca(2+)]Bulk and demonstrated dynamic differences during electric excitation and a standing diastolic [Ca(2+)]i gradient, which could influence local Ca(2+)-dependent signaling within the junctional cleft.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Miocitos Cardíacos/metabolismo , Imagen Óptica/métodos , Retículo Sarcoplasmático/metabolismo , Adenoviridae/genética , Animales , Calmodulina/genética , Células Cultivadas , Citosol/metabolismo , Acoplamiento Excitación-Contracción/fisiología , Proteínas Fluorescentes Verdes/genética , Uniones Intercelulares/metabolismo , Mutagénesis , Miocitos Cardíacos/citología , Quinasa de Cadena Ligera de Miosina/genética , Ratas , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
7.
Circ Res ; 114(10): 1623-39, 2014 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-24812351

RESUMEN

Genetically encoded Ca(2+) indicators constitute a powerful set of tools to investigate functional aspects of Ca(2+) signaling in isolated cardiomyocytes, cardiac tissue, and whole hearts. Here, we provide an overview of the concepts, experiences, state of the art, and ongoing developments in the use of genetically encoded Ca(2+) indicators for cardiac cells and heart tissue. This review is supplemented with in vivo viral gene transfer experiments and comparisons of available genetically encoded Ca(2+) indicators with each other and with the small molecule dye Fura-2. In the context of cardiac myocytes, we provide guidelines for selecting a genetically encoded Ca(2+) indicator. For future developments, we discuss improvements of a broad range of properties, including photophysical properties such as spectral spread and biocompatibility, as well as cellular and in vivo applications.


Asunto(s)
Señalización del Calcio/genética , Colorantes Fluorescentes , Miocitos Cardíacos/química , Miocitos Cardíacos/fisiología , Transgenes , Animales , Diagnóstico por Imagen/métodos , Técnicas de Transferencia de Gen , Humanos , Miocitos Cardíacos/metabolismo
8.
Am J Physiol Heart Circ Physiol ; 309(5): H958-68, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26209057

RESUMEN

Arterial myocytes express α1-catalytic subunit isoform Na(+) pumps (75-80% of total), which are ouabain resistant in rodents, and high ouabain affinity α2-Na(+) pumps. Mice with globally reduced α2-pumps (but not α1-pumps), mice with mutant ouabain-resistant α2-pumps, and mice with a smooth muscle (SM)-specific α2-transgene (α2 (SM-Tg)) that induces overexpression all have altered blood pressure (BP) phenotypes. We generated α2 (SM-DN) mice with SM-specific α2 (not α1) reduction (>50%) using nonfunctional dominant negative (DN) α2. We compared α2 (SM-DN) and α2 (SM-Tg) mice to controls to determine how arterial SM α2-pumps affect vasoconstriction and BP. α2 (SM-DN) mice had elevated basal mean BP (mean BP by telemetry: 117 ± 4 vs. 106 ± 1 mmHg, n = 7/7, P < 0.01) and enhanced BP responses to chronic ANG II infusion (240 ng·kg(-1)·min(-1)) and high (6%) NaCl. Several arterial Ca(2+) transporters, including Na(+)/Ca(2+) exchanger 1 (NCX1) and sarcoplasmic reticulum and plasma membrane Ca(2+) pumps [sarco(endo)plasmic reticulum Ca(2+)-ATPase 2 (SERCA2) and plasma membrane Ca(2+)-ATPase 1 (PMCA1)], were also reduced (>50%). α2 (SM-DN) mouse isolated small arteries had reduced myogenic reactivity, perhaps because of reduced Ca(2+) transporter expression. In contrast, α2 (SM-Tg) mouse aortas overexpressed α2 (>2-fold), NCX1, SERCA2, and PMCA1 (43). α2 (SM-Tg) mice had reduced basal mean BP (104 ± 1 vs. 109 ± 2 mmHg, n = 15/9, P < 0.02) and attenuated BP responses to chronic ANG II (300-400 ng·kg(-1)·min(-1)) with or without 2% NaCl but normal myogenic reactivity. NCX1 expression was inversely related to basal BP in SM-α2 engineered mice but was directly related in SM-NCX1 engineered mice. NCX1, which usually mediates arterial Ca(2+) entry, and α2-Na(+) pumps colocalize at plasma membrane-sarcoplasmic reticulum junctions and functionally couple via the local Na(+) gradient to help regulate cell Ca(2+). Altered Ca(2+) transporter expression in SM-α2 engineered mice apparently compensates to minimize Ca(2+) overload (α2 (SM-DN)) or depletion (α2 (SM-Tg)) and attenuate BP changes. In contrast, Ca(2+) transporter upregulation, observed in many rodent hypertension models, should enhance Ca(2+) entry and signaling and contribute significantly to BP elevation.


Asunto(s)
Arterias/metabolismo , Presión Sanguínea , Músculo Liso Vascular/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Angiotensina II/farmacología , Animales , Arterias/fisiología , Ratones , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Intercambiador de Sodio-Calcio/genética , Intercambiador de Sodio-Calcio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética
9.
Stem Cells ; 32(7): 1701-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24585704

RESUMEN

Ischemic heart disease is the number one cause of morbidity and mortality in the developed world due to the inability of the heart to replace lost myocytes. The cause of postinfarction myogenic failure has been a subject of intense scientific investigation and much controversy. Recent data indicate a brief perinatal developmental window exists during which postinfarction myogenesis, and substantial heart regeneration, occurs. By contrast, repair of an equivalent injury of the adult heart results in prominent revascularization without myogenesis. Here, we review recent experiments on neonatal postinjury myogenesis, examine the mechanistic hypotheses of dedifferentiation and precursor expansion, and discuss experiments indicating that postinfarction revascularization derives primarily from cardiac vascular precursors. These data have profound consequences for the understanding of human heart repair, as they address the long standing question as to whether human postinfarction myogenic failure is due to the loss of precursors existent at the neonatal stage or to a context-dependent inhibition of these precursors within the infarct, and suggest strategies for the recapitulation of neonatal myogenic capacity and the augmentation of revascularization.


Asunto(s)
Células Madre Adultas/fisiología , Corazón/fisiopatología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Desdiferenciación Celular , Vasos Coronarios/fisiopatología , Cardiopatías/fisiopatología , Humanos , Neovascularización Fisiológica , Regeneración
10.
Proc Natl Acad Sci U S A ; 109(33): 13380-5, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22847442

RESUMEN

We examined the myogenic response to infarction in neonatal and adult mice to determine the role of c-kit(+) cardiovascular precursor cells (CPC) that are known to be present in early heart development. Infarction of postnatal day 1-3 c-kit(BAC)-EGFP mouse hearts induced the localized expansion of (c-kit)EGFP(+) cells within the infarct, expression of the c-kit and Nkx2.5 mRNA, myogenesis, and partial regeneration of the infarction, with (c-kit)EGFP(+) cells adopting myogenic and vascular fates. Conversely, infarction of adult mice resulted in a modest induction of (c-kit)EGFP(+) cells within the infarct, which did not express Nkx2.5 or undergo myogenic differentiation, but adopted a vascular fate within the infarction, indicating a lack of authentic CPC. Explantation of infarcted neonatal and adult heart tissue to scid mice, and adoptive transfer of labeled bone marrow, confirmed the cardiac source of myogenic (neonate) and angiogenic (neonate and adult) cells. FACS-purified (c-kit)EGFP(+)/(αMHC)mCherry(-) (noncardiac) cells from microdissected infarcts within 6 h of infarction underwent cardiac differentiation, forming spontaneously beating myocytes in vitro; cre/LoxP fate mapping identified a noncardiac population of (c-kit)EGFP(+) myocytes within infarctions, indicating that the induction of undifferentiated precursors contributes to localized myogenesis. Thus, adult postinfarct myogenic failure is likely not due to a context-dependent restriction of precursor differentiation, and c-kit induction following injury of the adult heart does not define precursor status.


Asunto(s)
Envejecimiento/patología , Desarrollo de Músculos , Infarto del Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/citología , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Linaje de la Célula , Células Endoteliales/metabolismo , Células Endoteliales/patología , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Infarto del Miocardio/metabolismo , Regeneración , Células Madre/metabolismo
11.
Circ Res ; 111(2): e19-31, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22647876

RESUMEN

RATIONALE: Nkx2.5 is one of the most widely studied cardiac-specific transcription factors, conserved from flies to man, with multiple essential roles in both the developing and adult heart. Specific dominant mutations in NKX2.5 have been identified in adult congenital heart disease patients presenting with conduction system anomalies and recent genome-wide association studies implicate the NKX2.5 locus, as causative for lethal arrhythmias ("sudden cardiac death") that occur at a frequency in the population of 1 in 1000 per annum worldwide. Haploinsufficiency for Nkx2.5 in the mouse phenocopies human conduction disease pathology yet the phenotypes, described in both mouse and man, are highly pleiotropic, implicit of unknown modifiers and/or factors acting in epistasis with Nkx2.5/NKX2.5. OBJECTIVE: To identify bone fide upstream genetic modifier(s) of Nkx2.5/NKX2.5 function and to determine epistatic effects relevant to the manifestation of NKX2.5-dependent adult congenital heart disease. METHODS AND RESULTS: A study of cardiac function in prospero-related homeobox protein 1 (Prox1) heterozygous mice, using pressure-volume loop and micromannometry, revealed rescue of hemodynamic parameters in Nkx2.5(Cre/+); Prox1(loxP/+) animals versus Nkx2.5(Cre/+) controls. Anatomic studies, on a Cx40(EGFP) background, revealed Cre-mediated knock-down of Prox1 restored the anatomy of the atrioventricular node and His-Purkinje network both of which were severely hypoplastic in Nkx2.5(Cre/+) littermates. Steady state surface electrocardiography recordings and high-speed multiphoton imaging, to assess Ca(2+) handling, revealed atrioventricular conduction and excitation-contraction were also normalized by Prox1 haploinsufficiency, as was expression of conduction genes thought to act downstream of Nkx2.5. Chromatin immunoprecipitation on adult hearts, in combination with both gain and loss-of-function reporter assays in vitro, revealed that Prox1 recruits the corepressor HDAC3 to directly repress Nkx2.5 via a proximal upstream enhancer as a mechanism for regulating Nkx2.5 function in adult cardiac conduction. CONCLUSIONS: Here we identify Prox1 as a direct upstream modifier of Nkx2.5 in the maintenance of the adult conduction system and rescue of Nkx2.5 conduction disease phenotypes. This study is the first example of rescue of Nkx2.5 function and establishes a model for ensuring electrophysiological function within the adult heart alongside insight into a novel Prox1-HDAC3-Nkx2.5 signaling pathway for therapeutic targeting in conduction disease.


Asunto(s)
Epistasis Genética/genética , Sistema de Conducción Cardíaco/fisiopatología , Cardiopatías/genética , Cardiopatías/metabolismo , Histona Desacetilasas/genética , Proteínas de Homeodominio/genética , Fenotipo , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Animales , Cardiopatías/fisiopatología , Histona Desacetilasas/fisiología , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/fisiología , Ratones , Ratones Transgénicos , Células 3T3 NIH , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología
12.
Nucleic Acids Res ; 40(5): e39, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22189104

RESUMEN

Green Fluorescent Protein (GFP) and related fluorescent proteins (FPs) have been widely used to tag proteins, allowing their expression and subcellular localization to be examined in real time in living cells and animals. Similar fluorescent methods are highly desirable to detect and track RNA and other biological molecules in living cells. For this purpose, we have developed a group of RNA aptamers that bind GFP and related proteins, which we term Fluorescent Protein-Binding Aptamers (FPBA). These aptamers bind GFP, YFP and CFP with low nanomolar affinity and binding decreases GFP fluorescence, whereas slightly augmenting YFP and CFP brightness. Aptamer binding results in an increase in the pKa of EGFP, decreasing the 475 nm excited green fluorescence at a given pH. We report the secondary structure of FPBA and the ability to synthesize functional multivalent dendrimers. FPBA expressed in live cells decreased GFP fluorescence in a valency-dependent manner, indicating that the RNA aptamers function within cells. The development of aptamers that bind fluorescent proteins with high affinity and alter their function, markedly expands their use in the study of biological pathways.


Asunto(s)
Aptámeros de Nucleótidos/química , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/química , Aptámeros de Nucleótidos/metabolismo , Secuencia de Bases , Dendrímeros/química , Proteínas Fluorescentes Verdes/análisis , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/química , Datos de Secuencia Molecular , Conformación de Ácido Nucleico
13.
Proc Natl Acad Sci U S A ; 108(23): 9478-83, 2011 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-21606356

RESUMEN

In the epithelium of the lower airways, a cell type of unknown function has been termed "brush cell" because of a distinctive ultrastructural feature, an apical tuft of microvilli. Morphologically similar cells in the nose have been identified as solitary chemosensory cells responding to taste stimuli and triggering trigeminal reflexes. Here we show that brush cells of the mouse trachea express the receptors (Tas2R105, Tas2R108), the downstream signaling molecules (α-gustducin, phospholipase C(ß2)) of bitter taste transduction, the synthesis and packaging machinery for acetylcholine, and are addressed by vagal sensory nerve fibers carrying nicotinic acetylcholine receptors. Tracheal application of an nAChR agonist caused a reduction in breathing frequency. Similarly, cycloheximide, a Tas2R108 agonist, evoked a drop in respiratory rate, being sensitive to nicotinic receptor blockade and epithelium removal. This identifies brush cells as cholinergic sensors of the chemical composition of the lower airway luminal microenvironment that are directly linked to the regulation of respiration.


Asunto(s)
Células Quimiorreceptoras/metabolismo , Receptores Nicotínicos/metabolismo , Respiración , Tráquea/fisiología , Animales , Colina O-Acetiltransferasa/genética , Colina O-Acetiltransferasa/metabolismo , Femenino , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Unión al GTP Heterotriméricas/genética , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microscopía Confocal , Microscopía Electrónica , Microvellosidades/metabolismo , Microvellosidades/ultraestructura , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Gusto , Tráquea/citología , Tráquea/metabolismo , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
14.
JCI Insight ; 9(5)2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38319719

RESUMEN

Cauterization of the root of the left coronary artery (LCA) in the neonatal heart on postnatal day 1 (P1) resulted in large, reproducible lesions of the left ventricle (LV), and an attendant marked adaptive response in the right ventricle (RV). The response of both chambers to LV myocardial infarction involved enhanced cardiomyocyte (CM) division and binucleation, as well as LV revascularization, leading to restored heart function within 7 days post surgery (7 dps). By contrast, infarction of P3 mice resulted in cardiac scarring without a significant regenerative and adaptive response of the LV and the RV, leading to subsequent heart failure and death within 7 dps. The prominent RV myocyte expansion in P1 mice involved an acute increase in pulmonary arterial pressure and a unique gene regulatory response, leading to an increase in RV mass and preserved heart function. Thus, distinct adaptive mechanisms in the RV, such as CM proliferation and RV expansion, enable marked cardiac regeneration of the infarcted LV at P1 and full functional recovery.


Asunto(s)
Ventrículos Cardíacos , Infarto del Miocardio , Animales , Ratones , Ventrículos Cardíacos/patología , Miocitos Cardíacos/patología , Animales Recién Nacidos , Infarto del Miocardio/patología , Regeneración
15.
Neuron ; 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38781972

RESUMEN

Brain arterioles are active, multicellular complexes whose diameters oscillate at ∼ 0.1 Hz. We assess the physiological impact and spatiotemporal dynamics of vaso-oscillations in the awake mouse. First, vaso-oscillations in penetrating arterioles, which source blood from pial arterioles to the capillary bed, profoundly impact perfusion throughout neocortex. The modulation in flux during resting-state activity exceeds that of stimulus-induced activity. Second, the change in perfusion through arterioles relative to the change in their diameter is weak. This implies that the capillary bed dominates the hydrodynamic resistance of brain vasculature. Lastly, the phase of vaso-oscillations evolves slowly along arterioles, with a wavelength that exceeds the span of the cortical mantle and sufficient variability to establish functional cortical areas as parcels of uniform phase. The phase-gradient supports traveling waves in either direction along both pial and penetrating arterioles. This implies that waves along penetrating arterioles can mix, but not directionally transport, interstitial fluids.

16.
Am J Physiol Heart Circ Physiol ; 305(6): H829-42, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23832701

RESUMEN

Redox status has emerged as critical in modulating stemness and lineage commitment in several precursor cell types. However, a role for redox genes, specifically NADPH oxidases (Nox), in cardiac precursor cells (CPCs) has not been established. We tested whether CPCs marked by type III receptor tyrosine kinase c-kit (c-kit(+)) exhibit a unique NADPH oxidase signature that confers precursor status and whether alterations in this profile are functionally linked to changes in lineage specification. Dihydroethidium (DHE) microfluorography indicated reduced basal reactive oxygen species (ROS) formation within early postnatal c-kit(+) CPCs. Real-time quantitative PCR revealed downregulation of ROS generator Nox2 and its subunit p67(phox) in c-kit(+) CPCs under basal conditions but upregulation of Nox2 and Nox4 over the course of differentiation. Adenoviral silencing of Nox2 and Nox4 increased expression of CPC markers c-kit and Flk-1 and blunted smooth and cardiac muscle differentiation, respectively, while overexpression of Nox2 and Nox4 significantly reduced c-kit expression. These changes were accompanied by altered expression of transcription factors regulating cardiac lineage commitment, Gata6 and Gata4, and cytokine transforming growth factor (TGF)-ß1. Similar to other precursor cell types, RT(2)Profiler PCR Arrays revealed that c-kit(+) CPCs also exhibit enhanced antioxidant capacity at the mRNA level. In conclusion, we report that c-kit(+) CPCs demonstrate reduced Nox2 expression and ROS levels and that increases in Nox2 and Nox4 influence their differentiation into mature cells. We speculate that ROS generators Nox2 and Nox4, along with the antioxidant genes identified by PCR Arrays, may be novel targets in CPCs that could prove useful in cell-based therapy of the heart.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , NADPH Oxidasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular , Células Cultivadas , Ratones , Ratones Transgénicos , NADPH Oxidasa 2 , NADPH Oxidasa 4 , Especies Reactivas de Oxígeno/metabolismo
17.
Nature ; 450(7171): 819-24, 2007 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-18064002

RESUMEN

Ventricular tachyarrhythmias are the main cause of sudden death in patients after myocardial infarction. Here we show that transplantation of embryonic cardiomyocytes (eCMs) in myocardial infarcts protects against the induction of ventricular tachycardia (VT) in mice. Engraftment of eCMs, but not skeletal myoblasts (SMs), bone marrow cells or cardiac myofibroblasts, markedly decreased the incidence of VT induced by in vivo pacing. eCM engraftment results in improved electrical coupling between the surrounding myocardium and the infarct region, and Ca2+ signals from engrafted eCMs expressing a genetically encoded Ca2+ indicator could be entrained during sinoatrial cardiac activation in vivo. eCM grafts also increased conduction velocity and decreased the incidence of conduction block within the infarct. VT protection is critically dependent on expression of the gap-junction protein connexin 43 (Cx43; also known as Gja1): SMs genetically engineered to express Cx43 conferred a similar protection to that of eCMs against induced VT. Thus, engraftment of Cx43-expressing myocytes has the potential to reduce life-threatening post-infarct arrhythmias through the augmentation of intercellular coupling, suggesting autologous strategies for cardiac cell-based therapy.


Asunto(s)
Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/prevención & control , Conexina 43/metabolismo , Infarto del Miocardio/complicaciones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/trasplante , Animales , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Conexina 43/genética , Embrión de Mamíferos/citología , Corazón/fisiología , Corazón/fisiopatología , Humanos , Ratones , Ratones Transgénicos , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/citología , Miocardio/patología , Perfusión
18.
Am J Physiol Heart Circ Physiol ; 302(3): H594-602, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22140050

RESUMEN

It is generally accepted that the endothelium regulates vascular tone independent of the activity of the sympathetic nervous system. Here, we tested the hypothesis that the activation of sympathetic nerves engages the endothelium to oppose vasoconstriction. Local inositol 1,4,5-trisphosphate (IP(3))-mediated Ca(2+) signals ("pulsars") in or near endothelial projections to vascular smooth muscle (VSM) were measured in an en face mouse mesenteric artery preparation. Electrical field stimulation of sympathetic nerves induced an increase in endothelial cell (EC) Ca(2+) pulsars, recruiting new pulsar sites without affecting activity at existing sites. This increase in Ca(2+) pulsars was blocked by bath application of the α-adrenergic receptor antagonist prazosin or by TTX but was unaffected by directly picospritzing the α-adrenergic receptor agonist phenylephrine onto the vascular endothelium, indicating that nerve-derived norepinephrine acted through α-adrenergic receptors on smooth muscle cells. Moreover, EC Ca(2+) signaling was not blocked by inhibitors of purinergic receptors, ryanodine receptors, or voltage-dependent Ca(2+) channels, suggesting a role for IP(3), rather than Ca(2+), in VSM-to-endothelium communication. Block of intermediate-conductance Ca(2+)-sensitive K(+) channels, which have been shown to colocalize with IP(3) receptors in endothelial projections to VSM, enhanced nerve-evoked constriction. Collectively, our results support the concept of a transcellular negative feedback module whereby sympathetic nerve stimulation elevates EC Ca(2+) signals to oppose vasoconstriction.


Asunto(s)
Señalización del Calcio/fisiología , Arterias Mesentéricas/inervación , Arterias Mesentéricas/fisiología , Sistema Nervioso Simpático/fisiología , Vasoconstricción/fisiología , Animales , Calcio/metabolismo , Conexinas/genética , Endotelio Vascular/metabolismo , Retroalimentación Fisiológica/fisiología , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Receptores Adrenérgicos alfa/metabolismo , Proteína alfa-5 de Unión Comunicante
19.
Arterioscler Thromb Vasc Biol ; 31(6): 1291-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21372299

RESUMEN

OBJECTIVE: Although vascular smooth muscle cells (VSMCs) provide the major structural integrity of atherosclerotic plaques, their origin has been questioned. In particular, although some studies identified plaque VSMCs originating from bone marrow or peripheral blood, their frequency is controversial and their function unknown. We used genetic tracking of cell fate through smooth muscle cell (SMC)-specific LacZ reporter activity and VSMC-selective apoptosis to investigate the frequency, distribution, and role of marrow-derived VSMCs in atherogenesis. METHODS AND RESULTS: Cultured mouse bone marrow-derived smooth muscle-like cells expressed SMC markers and functional SMC promoter-driven transgenes over time. Transplantation of apolipoprotein E (ApoE)(-/-) mice with smooth muscle myosin heavy chain-Cre/ROSA26R/ApoE(-/-) marrow showed that 0.7±0.14% cells expressed LacZ in atherosclerotic plaques, located superficially in early plaques, and in necrotic cores but not fibrous caps of advanced lesions. Cells expressing both progenitor and SMC markers showed a similar distribution and frequency. Apoptosis of marrow-derived SMC-like cells transplanted from SM22α-human diphtheria toxin receptor/ApoE(-/-) mice retarded atherogenesis, with reduced plaque macrophage content. Cultured marrow-derived SMC-like cells secreted proinflammatory cytokines and promoted macrophage migration, VSMC proliferation, and collagen synthesis. CONCLUSION: Bone marrow-derived SMC-like cells are infrequent in advanced primary atherosclerotic plaques and absent in fibrous caps. However, these cells secrete proinflammatory cytokines and mitogens and promote atherosclerosis.


Asunto(s)
Aterosclerosis/etiología , Células de la Médula Ósea/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Animales , Apolipoproteínas E/fisiología , Apoptosis/efectos de los fármacos , Aterosclerosis/patología , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Toxina Diftérica/farmacología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Péptidos y Proteínas de Señalización Intercelular/fisiología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL
20.
Proc Natl Acad Sci U S A ; 106(6): 1808-13, 2009 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-19193854

RESUMEN

Directed differentiation of embryonic stem cells indicates that mesodermal lineages in the mammalian heart (cardiac, endothelial, and smooth muscle cells) develop from a common, multipotent cardiovascular precursor. To isolate and characterize the lineage potential of a resident pool of cardiovascular progenitor cells (CPcs), we developed BAC transgenic mice in which enhanced green fluorescent protein (EGFP) is placed under control of the c-kit locus (c-kit(BAC)-EGFP mice). Discrete c-kit-EGFP(+) cells were observed at different stages of differentiation in embryonic hearts, increasing in number to a maximum at about postnatal day (PN) 2; thereafter, EGFP(+) cells declined and were rarely observed in the adult heart. EGFP(+) cells purified from PN 0-5 hearts were nestin(+) and expanded in culture; 67% of cells were fluorescent after 9 days. Purified cells differentiated into endothelial, cardiac, and smooth muscle cells, and differentiation could be directed by specific growth factors. CPc-derived cardiac myocytes displayed rhythmic beating and action potentials characteristic of multiple cardiac cell types, similar to ES cell-derived cardiomyocytes. Single-cell dilution studies confirmed the potential of individual CPcs to form all 3 cardiovascular lineages. In adult hearts, cryoablation resulted in c-kit-EGFP(+) expression, peaking 7 days postcryolesion. Expression occurred in endothelial and smooth muscle cells in the revascularizing infarct, and in terminally differentiated cardiomyocytes in the border zone surrounding the infarct. Thus, c-kit expression marks CPc in the neonatal heart that are capable of directed differentiation in vitro; however, c-kit expression in cardiomyocytes in the adult heart after injury does not identify cardiac myogenesis.


Asunto(s)
Células Madre Multipotentes/citología , Miocardio/citología , Proteínas Proto-Oncogénicas c-kit/análisis , Animales , Animales Recién Nacidos , Sistema Cardiovascular/citología , Diferenciación Celular , Linaje de la Célula , Vasos Coronarios/citología , Criocirugía , Embrión de Mamíferos , Proteínas Fluorescentes Verdes/genética , Mesodermo/citología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Miocitos Cardíacos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA