Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Circulation ; 149(17): 1375-1390, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38214189

RESUMO

BACKGROUND: Cardiac transverse tubules (T-tubules) are anchored to sarcomeric Z-discs by costameres to establish a regular spaced pattern. One of the major components of costameres is the dystrophin-glycoprotein complex (DGC). Nevertheless, how the assembly of the DGC coordinates with the formation and maintenance of T-tubules under physiological and pathological conditions remains unclear. METHODS: Given the known role of Ptpn23 (protein tyrosine phosphatase, nonreceptor type 23) in regulating membrane deformation, its expression in patients with dilated cardiomyopathy was determined. Taking advantage of Cre/Loxp, CRISPR/Cas9, and adeno-associated virus 9 (AAV9)-mediated in vivo gene editing, we generated cardiomyocyte-specific Ptpn23 and Actn2 (α-actinin-2, a major component of Z-discs) knockout mice. We also perturbed the DGC by using dystrophin global knockout mice (DmdE4*). MM 4-64 and Di-8-ANEPPS staining, Cav3 immunofluorescence, and transmission electron microscopy were performed to determine T-tubule structure in isolated cells and intact hearts. In addition, the assembly of the DGC with Ptpn23 and dystrophin loss of function was determined by glycerol-gradient fractionation and SDS-PAGE analysis. RESULTS: The expression level of Ptpn23 was reduced in failing hearts from dilated cardiomyopathy patients and mice. Genetic deletion of Ptpn23 resulted in disorganized T-tubules with enlarged diameters and progressive dilated cardiomyopathy without affecting sarcomere organization. AAV9-mediated mosaic somatic mutagenesis further indicated a cell-autonomous role of Ptpn23 in regulating T-tubule formation. Genetic and biochemical analyses showed that Ptpn23 was essential for the integrity of costameres, which anchor the T-tubule membrane to Z-discs, through interactions with α-actinin and dystrophin. Deletion of α-actinin altered the subcellular localization of Ptpn23 and DGCs. In addition, genetic inactivation of dystrophin caused similar T-tubule defects to Ptpn23 loss-of-function without affecting Ptpn23 localization at Z-discs. Last, inducible Ptpn23 knockout at 1 month of age showed Ptpn23 is also required for the maintenance of T-tubules in adult cardiomyocytes. CONCLUSIONS: Ptpn23 is essential for cardiac T-tubule formation and maintenance along Z-discs. During postnatal heart development, Ptpn23 interacts with sarcomeric α-actinin and coordinates the assembly of the DGC at costameres to sculpt T-tubule spatial patterning and morphology.

2.
J Mol Cell Cardiol ; 190: 35-47, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38593639

RESUMO

BACKGROUND: Exosomes released by cardiomyocytes are essential mediators of intercellular communications within the heart, and various exosomal proteins and miRNAs are associated with cardiovascular diseases. However, whether the endosomal sorting complex required for transport (ESCRT) and its key component Alix is required for exosome biogenesis within cardiomyocyte remains poorly understood. METHODS: Super-resolution imaging was performed to investigate the subcellular location of Alix and multivesicular body (MVB) in primary cardiomyocytes. Cardiomyocyte-specific Alix-knockout mice were generated using AAV9/CRISPR/Cas9-mediated in vivo gene editing. A stable Alix-knockdown H9c2 cardiomyocyte line was constructed through lentiviral-mediated delivery of short hairpin RNA. In order to determine the role of Alix in controlling exosome biogenesis, exosomes from cardiomyocyte-specific Alix-knockout mice plasma and Alix-knockdown H9c2 culture medium were isolated and examined by western blot, NTA analysis and transmission electron microscopy. Biochemical and immunofluorescence analysis were performed to determine the role of ESCRT machinery in regulating MVB formation. Lastly, transverse aortic constriction (TAC)-induced cardiac pressure overload model was established to further explore the role of Alix-mediated exosome biogenesis under stress conditions. RESULTS: A significant proportion of Alix localized to the MVB membrane within cardiomyocytes. Genetic deletion of Alix in murine heart resulted in a reduction of plasma exosome content without affecting cardiac structure or contractile function. Consistently, the downregulation of Alix in H9c2 cardiomyocyte line also suppressed the biogenesis of exosomes. We found the defective ESCRT machinery and suppressed MVB formation upon Alix depletion caused compromised exosome biogenesis. Remarkably, TAC-induced cardiac pressure overload led to increased Alix, MVB levels, and elevated plasma exosome content, which could be totally abolished by Alix deletion. CONCLUSION: These results establish Alix as an essential and stress-sensitive regulator of cardiac exosome biogenesis and the findings may yield valuable therapeutic implications.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte , Exossomos , Camundongos Knockout , Miócitos Cardíacos , Estresse Fisiológico , Miócitos Cardíacos/metabolismo , Animais , Exossomos/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Camundongos , Corpos Multivesiculares/metabolismo , Linhagem Celular , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/genética , Ratos
3.
Cell ; 134(2): 279-90, 2008 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-18662543

RESUMO

In quiescent cells, mitochondria are the primary source of reactive oxygen species (ROS), which are generated by leakiness of the electron transport chain (ETC). High levels of ROS can trigger cell death, whereas lower levels drive diverse and important cellular functions. We show here by employing a newly developed mitochondrial matrix-targeted superoxide indicator, that individual mitochondria undergo spontaneous bursts of superoxide generation, termed "superoxide flashes." Superoxide flashes occur randomly in space and time, exhibit all-or-none properties, and provide a vital source of superoxide production across many different cell types. Individual flashes are triggered by transient openings of the mitochondrial permeability transition pore stimulating superoxide production by the ETC. Furthermore, we observe a flurry of superoxide flash activity during reoxygenation of cardiomyocytes after hypoxia, which is inhibited by the cardioprotective compound adenosine. We propose that superoxide flashes could serve as a valuable biomarker for a wide variety of oxidative stress-related diseases.


Assuntos
Mitocôndrias/metabolismo , Superóxidos/metabolismo , Adenoviridae/genética , Animais , Hipóxia Celular , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Proteínas Luminescentes/metabolismo , Células Musculares/metabolismo , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
4.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37445978

RESUMO

Autophagy has stabilizing functions for cardiomyocytes. Recent studies indicate that an impairment in the autophagy pathway can seriously affect morphology and function, potentially leading to heart failure. However, the role and the underlying mechanism of the endosomal sorting complex required for transport (ESCRT) family protein, in particular the AAA-ATPase vacuolar protein sorting 4a (Vps4a), in regulating myocardial autophagy remains unclear. In the present study, cardiomyocyte-specific Vps4a knockout mice were generated by crossing Vps4aflox/flox (Vps4afl/fl) with Myh6-cre transgenic mice. As a result, we observed a partially dilated left ventricular (LV) chamber, a significant increase in heart weight to body weight ratio (HW/BW), and heart weight to tibial length ratio (HW/TL), hypertrophic cardiomyopathy and early lethality starting at 3 months of age. Hematoxylin-eosin (HE), immunofluorescence assay (IFA), and Western blot (WB) revealed autophagosome accumulation in cardiomyocytes. A transcriptome-based analysis and autophagic flux tracking by AAV-RFP-GFP-LC3 showed that the autophagic flux was blocked in Vps4a knockout cardiomyocytes. In addition, we provided in vitro evidence demonstrating that Vps4a and LC3 were partially co-localized in cardiomyocytes, and the knockdown of Vps4a led to the accumulation of autophagosomes in cardiomyocytes. Similarly, the transfection of cardiomyocytes with adenovirus (Adv) mCherry-GFP-LC3 further indicated that the autophagic flux was blocked in cells with deficient levels of Vps4a. Finally, an electron microscope (EM) showed that the compromised sealing of autophagosome blocked the autophagic flux in Vps4a-depleted cardiomyocytes. These findings revealed that Vps4a contributed to the sealing of autophagosomes in cardiomyocytes. Therefore, we demonstrated that Vps4a deletion could block the autophagic flux, leading to the accumulation of degradation substances and compromised cardiac function. Overall, this study provides insights into a new theoretical basis for which autophagy may represent a therapeutic target for cardiovascular diseases.


Assuntos
Autofagia , Cardiomiopatia Hipertrófica , Camundongos , Animais , Autofagia/genética , Miocárdio/metabolismo , Autofagossomos/metabolismo , Miócitos Cardíacos/metabolismo , Camundongos Transgênicos , Cardiomiopatia Hipertrófica/metabolismo , Camundongos Knockout , Transporte Proteico
5.
Nature ; 534(7609): 700-4, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27357797

RESUMO

Many organs are composed of complex tissue walls that are structurally organized to optimize organ function. In particular, the ventricular myocardial wall of the heart comprises an outer compact layer that concentrically encircles the ridge-like inner trabecular layer. Although disruption in the morphogenesis of this myocardial wall can lead to various forms of congenital heart disease and non-compaction cardiomyopathies, it remains unclear how embryonic cardiomyocytes assemble to form ventricular wall layers of appropriate spatial dimensions and myocardial mass. Here we use advanced genetic and imaging tools in zebrafish to reveal an interplay between myocardial Notch and Erbb2 signalling that directs the spatial allocation of myocardial cells to their proper morphological positions in the ventricular wall. Although previous studies have shown that endocardial Notch signalling non-cell-autonomously promotes myocardial trabeculation through Erbb2 and bone morphogenetic protein (BMP) signalling, we discover that distinct ventricular cardiomyocyte clusters exhibit myocardial Notch activity that cell-autonomously inhibits Erbb2 signalling and prevents cardiomyocyte sprouting and trabeculation. Myocardial-specific Notch inactivation leads to ventricles of reduced size and increased wall thickness because of excessive trabeculae, whereas widespread myocardial Notch activity results in ventricles of increased size with a single-cell-thick wall but no trabeculae. Notably, this myocardial Notch signalling is activated non-cell-autonomously by neighbouring Erbb2-activated cardiomyocytes that sprout and form nascent trabeculae. Thus, these findings support an interactive cellular feedback process that guides the assembly of cardiomyocytes to morphologically create the ventricular myocardial wall and more broadly provide insight into the cellular dynamics of how diverse cell lineages organize to create form.


Assuntos
Ventrículos do Coração/citologia , Ventrículos do Coração/embriologia , Morfogênese , Miócitos Cardíacos/citologia , Peixe-Zebra/embriologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem da Célula , Retroalimentação Fisiológica , Ventrículos do Coração/anatomia & histologia , Proteína Jagged-2 , Miócitos Cardíacos/metabolismo , Tamanho do Órgão , Organogênese , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/metabolismo , Receptores Notch/antagonistas & inibidores , Receptores Notch/metabolismo , Transdução de Sinais , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
6.
Nature ; 498(7455): 497-501, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23783515

RESUMO

Despite current treatment regimens, heart failure remains the leading cause of morbidity and mortality in the developed world due to the limited capacity of adult mammalian ventricular cardiomyocytes to divide and replace ventricular myocardium lost from ischaemia-induced infarct. Hence there is great interest to identify potential cellular sources and strategies to generate new ventricular myocardium. Past studies have shown that fish and amphibians and early postnatal mammalian ventricular cardiomyocytes can proliferate to help regenerate injured ventricles; however, recent studies have suggested that additional endogenous cellular sources may contribute to this overall ventricular regeneration. Here we have developed, in the zebrafish (Danio rerio), a combination of fluorescent reporter transgenes, genetic fate-mapping strategies and a ventricle-specific genetic ablation system to discover that differentiated atrial cardiomyocytes can transdifferentiate into ventricular cardiomyocytes to contribute to zebrafish cardiac ventricular regeneration. Using in vivo time-lapse and confocal imaging, we monitored the dynamic cellular events during atrial-to-ventricular cardiomyocyte transdifferentiation to define intermediate cardiac reprogramming stages. We observed that Notch signalling becomes activated in the atrial endocardium following ventricular ablation, and discovered that inhibiting Notch signalling blocked the atrial-to-ventricular transdifferentiation and cardiac regeneration. Overall, these studies not only provide evidence for the plasticity of cardiac lineages during myocardial injury, but more importantly reveal an abundant new potential cardiac resident cellular source for cardiac ventricular regeneration.


Assuntos
Transdiferenciação Celular , Reprogramação Celular , Coração/fisiologia , Miocárdio/citologia , Regeneração/fisiologia , Peixe-Zebra/fisiologia , Animais , Morte Celular , Coração/embriologia , Átrios do Coração/citologia , Átrios do Coração/embriologia , Ventrículos do Coração/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Peixe-Zebra/embriologia
7.
J Immunol ; 192(12): 5998-6008, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24835391

RESUMO

During inflammation, the proper inflammatory infiltration of neutrophils is crucial for the host to fight against infections and remove damaged cells and detrimental substances. IL-1ß and NADPH oxidase-mediated reactive oxygen species (ROS) have been implicated to play important roles in this process. However, the cellular and molecular basis underlying the actions of IL-1ß and ROS and their relationship during inflammatory response remains undefined. In this study, we use the zebrafish model to investigate these issues. We find that, similar to that of NADPH oxidase-mediated ROS signaling, the Il-1ß-Myd88 pathway is required for the recruitment of neutrophils, but not macrophages, to the injury-induced inflammatory site, whereas it is dispensable for bacterial-induced inflammation. Interestingly, the Il-1ß-Myd88 pathway is independent of NADPH oxidase-mediated ROS signaling and critical for the directional migration, but not the basal random movement, of neutrophils. In contrast, the NADPH oxidase-mediated ROS signaling is required for both basal random movement and directional migration of neutrophils. We further document that ectopic expression of Il-1ß in zebrafish induces an inflammatory disorder, which can be suppressed by anti-inflammatory treatment. Our findings reveal that the Il-1ß-Myd88 axis and NADPH oxidase-mediated ROS signaling are two independent pathways that differentially regulate neutrophil migration during sterile inflammation. In addition, Il-1ß overexpressing Tg(hsp70:(m)il-1ß_eGFP;lyz:DsRed2)hkz10t;nz50 transgenic zebrafish provides a useful animal model for the study of chronic inflammatory disorder and for anti-inflammatory drug discovery.


Assuntos
Movimento Celular/imunologia , Interleucina-1beta/imunologia , Neutrófilos/imunologia , Transdução de Sinais/imunologia , Proteínas de Peixe-Zebra/imunologia , Peixe-Zebra/imunologia , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-1beta/genética , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , NADPH Oxidases/genética , NADPH Oxidases/imunologia , Neutrófilos/patologia , Espécies Reativas de Oxigênio/imunologia , Transdução de Sinais/genética , Proteínas de Peixe-Zebra/genética
8.
Arterioscler Thromb Vasc Biol ; 34(10): 2268-75, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25147335

RESUMO

OBJECTIVE: Fluid shear stress intimately regulates vasculogenesis and endothelial homeostasis. The canonical Wnt/ß-catenin signaling pathways play an important role in differentiation and proliferation. In this study, we investigated whether shear stress activated angiopoietin-2 (Ang-2) via the canonical Wnt signaling pathway with an implication in vascular endothelial repair. APPROACH AND RESULTS: Oscillatory shear stress upregulated both TOPflash Wnt reporter activities and the expression of Ang-2 mRNA and protein in human aortic endothelial cells accompanied by an increase in nuclear ß-catenin intensity. Oscillatory shear stress-induced Ang-2 and Axin-2 mRNA expression was downregulated in the presence of a Wnt inhibitor, IWR-1, but was upregulated in the presence of a Wnt agonist, LiCl. Ang-2 expression was further downregulated in response to a Wnt signaling inhibitor, DKK-1, but was upregulated by Wnt agonist Wnt3a. Both DKK-1 and Ang-2 siRNA inhibited endothelial cell migration and tube formation, which were rescued by human recombinant Ang-2. Both Ang-2 and Axin-2 mRNA downregulation was recapitulated in the heat-shock-inducible transgenic Tg(hsp70l:dkk1-GFP) zebrafish embryos at 72 hours post fertilization. Ang-2 morpholino injection of Tg (kdrl:GFP) fish impaired subintestinal vessel formation at 72 hours post fertilization, which was rescued by zebrafish Ang-2 mRNA coinjection. Inhibition of Wnt signaling with IWR-1 also downregulated Ang-2 and Axin-2 expression and impaired vascular repair after tail amputation, which was rescued by zebrafish Ang-2 mRNA injection. CONCLUSIONS: Shear stress activated Ang-2 via canonical Wnt signaling in vascular endothelial cells, and Wnt-Ang-2 signaling is recapitulated in zebrafish embryos with a translational implication in vascular development and repair.


Assuntos
Angiopoietina-2/metabolismo , Mecanotransdução Celular , Neovascularização Fisiológica , Via de Sinalização Wnt , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Angiopoietina-2/genética , Animais , Animais Geneticamente Modificados , Proteína Axina/genética , Proteína Axina/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mecanotransdução Celular/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Interferência de RNA , RNA Mensageiro/metabolismo , Estresse Fisiológico , Fatores de Tempo , Transfecção , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt3A/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
9.
Basic Res Cardiol ; 108(3): 344, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23553082

RESUMO

The histidine-rich Ca(2+)-binding protein (HRC) is located in the lumen of the sarcoplasmic reticulum (SR) and exhibits high-capacity Ca(2+)-binding properties. Overexpression of HRC in the heart resulted in impaired SR Ca(2+) uptake and depressed relaxation through its interaction with SERCA2a. However, the functional significance of HRC in overall regulation of calcium cycling and contractility is not currently well defined. To further elucidate the role of HRC in vivo under physiological and pathophysiological conditions, we generated and characterized HRC-knockout (KO) mice. The KO mice were morphologically and histologically normal compared to wild-type (WT) mice. At the cellular level, ablation of HRC resulted in significantly enhanced contractility, Ca(2+) transients, and maximal SR Ca(2+) uptake rates in the heart. However, after-contractions were developed in 50 % of HRC-KO cardiomyocytes, compared to 11 % in WT mice under stress conditions of high-frequency stimulation (5 Hz) and isoproterenol application. A parallel examination of the electrical activity revealed significant increases in the occurrence of Ca(2+) spontaneous SR Ca(2+) release and delayed afterdepolarizations with ISO in HRC-KO, compared to WT cells. The frequency of Ca(2+) sparks was also significantly higher in HRC-KO cells with ISO, consistent with the elevated SR Ca(2+) load in the KO cells. Furthermore, HRC-KO cardiomyocytes showed significantly deteriorated cell contractility and Ca(2+)-cycling caused possibly by depressed SERCA2a expression after transverse-aortic constriction (TAC). Also HRC-null mice exhibited severe cardiac hypertrophy, fibrosis, pulmonary edema and decreased survival after TAC. Our results indicate that ablation of HRC is associated with poorly regulated SR Ca(2+)-cycling, and severe pathology under pressure-overload stress, suggesting an essential role of HRC in maintaining the integrity of cardiac function.


Assuntos
Sinalização do Cálcio , Proteínas de Ligação ao Cálcio/deficiência , Cardiomegalia/metabolismo , Hemodinâmica , Miócitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo , Animais , Proteínas de Ligação ao Cálcio/genética , Estimulação Cardíaca Artificial , Cardiomegalia/etiologia , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Fibrose , Genótipo , Isoproterenol , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , Miócitos Cardíacos/patologia , Fenótipo , Edema Pulmonar/etiologia , Edema Pulmonar/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Índice de Gravidade de Doença
10.
Sci Total Environ ; 904: 166926, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37689185

RESUMO

Evapotranspiration (ET) is a vital parameter in terrestrial water-energy cycles. The transpiration fraction (TF) is defined as the ratio of transpiration (T) to evapotranspiration (ET), representing the contribution rate of vegetation transpiration to ecosystem ET. Quantifying the relative contributions of vegetation and climate change on the ET and TF dynamic is of great significance to better understand the water budget between the land and atmosphere. Here, we chose Yellow River Basin (YRB) as the study area and analyzed the spatiotemporal changes of ET, T, and TF from 1982 to 2015 using the Priestley-Taylor Jet Propulsion Laboratory (PT-JPL) model. Meanwhile, the relative contributions of vegetation and climate change to ET, T and TF change were quantified. Model evaluation showed that the PT-JPL model performs well in the simulation of ET and T. During 1982-2015, the average annual ET, T, and TF increased at a rate of 3.20 mm/a, 0.77 mm/a and 0.003/a over the YRB during 1982-2015, respectively. The regions with significant increases in ET, T and TF almost covered the whole study area except for the upper reaches of the YRB. Vegetation greening was the main factor for the increase of ET and TF in the YRB and enhanced ET and TF at a rate of 0.72 mm/a and 0.57/a, respectively, which mainly observed in the entire Loess Plateau region (over 50 % of the study area). Precipitation (PRE) was also the dominated factor contributing to the increase in ET and TF, and temperature (TEM) showed a positive correlation with the changes in ET and TF in the most areas of YRB, which jointly dominated ET changes in the upper reaches of the YRB and TF changes in the southern part of the basin. Except for the total effects, leaf area index (LAI) also indirectly promoted ET changes by affecting PRE, TEM and relative humidity (RH). While wind speed (WS) and radiation (RAD) had a relatively weak regulatory effect on the changes in ET and TF. These findings were helpful for regional water resources management and formulating water resources-sustainable vegetation restoration strategies for local government.

11.
Circ Res ; 106(2): 317-27, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19926875

RESUMO

RATIONALE: Rad (Ras associated with diabetes) GTPase, a monomeric small G protein, binds to Ca(v)beta subunit of the L-type Ca(2+) channel (LCC) and thereby regulates LCC trafficking and activity. Emerging evidence suggests that Rad is an important player in cardiac arrhythmogenesis and hypertrophic remodeling. However, whether and how Rad involves in the regulation of excitation-contraction (EC) coupling is unknown. OBJECTIVE: This study aimed to investigate possible role of Rad in cardiac EC coupling and beta-adrenergic receptor (betaAR) inotropic mechanism. METHODS AND RESULTS: Adenoviral overexpression of Rad by 3-fold in rat cardiomyocytes suppressed LCC current (I(Ca)), [Ca(2+)](i) transients, and contractility by 60%, 42%, and 38%, respectively, whereas the "gain" function of EC coupling was significantly increased, due perhaps to reduced "redundancy" of LCC in triggering sarcoplasmic reticulum release. Conversely, approximately 70% Rad knockdown by RNA interference increased I(Ca) (50%), [Ca(2+)](i) transients (52%) and contractility (58%) without altering EC coupling efficiency; and the dominant negative mutant RadS105N exerted a similar effect on I(Ca). Rad upregulation caused depolarizing shift of LCC activation and hastened time-dependent LCC inactivation; Rad downregulation, however, failed to alter these attributes. The Na(+)/Ca(2+) exchange activity, sarcoplasmic reticulum Ca(2+) content, properties of Ca(2+) sparks and propensity for Ca(2+) waves all remained unperturbed regardless of Rad manipulation. Rad overexpression, but not knockdown, negated betaAR effects on I(Ca) and Ca(2+) transients. CONCLUSION: These results establish Rad as a novel endogenous regulator of cardiac EC coupling and betaAR signaling and support a parsimonious model in which Rad buffers Ca(v)beta to modulate LCC activity, EC coupling, and betaAR responsiveness.


Assuntos
Miócitos Cardíacos/fisiologia , Receptores Adrenérgicos beta/fisiologia , Transdução de Sinais , Proteínas ras/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Western Blotting , Cálcio/metabolismo , Células Cultivadas , Proteínas de Homeodomínio/metabolismo , Isoproterenol/farmacologia , Potenciais da Membrana , Contração Miocárdica , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Proteínas ras/genética
12.
Circ Res ; 107(1): 76-83, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20466981

RESUMO

RATIONALE: Unrepaired cardiomyocyte membrane injury causes irreplaceable cell loss, leading to myocardial fibrosis and eventually heart failure. However, the cellular and molecular mechanisms of cardiac membrane repair are largely unknown. MG53, a newly identified striated muscle-specific protein, is involved in skeletal muscle membrane repair. But the role of MG53 in the heart has not been determined. OBJECTIVE: We sought to investigate whether MG53 mediates membrane repair in cardiomyocytes and, if so, the cellular and molecular mechanism underlying MG53-mediated membrane repair in cardiomyocytes. Moreover, we determined possible cardioprotective effect of MG53-mediated membrane repair. METHODS AND RESULTS: We demonstrated that MG53 is crucial to the emergency membrane repair response in cardiomyocytes and protects the heart from stress-induced loss of cardiomyocytes. Disruption of the sarcolemmal membrane by mechanical, electric, chemical, or metabolic insults caused rapid and robust translocation of MG53 toward the injury sites. Ablation of MG53 prevented sarcolemmal resealing after infrared laser-induced membrane damage in intact heart, and exacerbated mitochondrial dysfunction and loss of cardiomyocytes during ischemia/reperfusion injury. Unexpectedly, the MG53-mediated cardiac membrane repair was mediated by a cholesterol-dependent mechanism: depletion of membrane cholesterol abolished, and its recovery restored injury-induced membrane translocation of MG53. The redox status of MG53 did not affect initiation of MG53 translocation, whereas MG53 oxidation conferred stability to the membrane repair patch. CONCLUSIONS: Thus, cholesterol-dependent MG53-mediated membrane repair is a vital, heretofore unappreciated cardioprotective mechanism against a multitude of insults and may bear important therapeutic implications.


Assuntos
Proteínas de Transporte/fisiologia , Membrana Celular/metabolismo , Colesterol/fisiologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/metabolismo , Animais , Proteínas de Transporte/genética , Membrana Celular/genética , Membrana Celular/patologia , Proteínas de Membrana , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/genética , Miócitos Cardíacos/patologia
13.
Proc Natl Acad Sci U S A ; 106(49): 20776-81, 2009 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-19920172

RESUMO

The HS-1 associated protein X-1 (HAX-1) is a ubiquitously expressed protein that protects cardiomyocytes from programmed cell death. Here we identify HAX-1 as a regulator of contractility and calcium cycling in the heart. HAX-1 overexpression reduced sarcoplasmic reticulum Ca-ATPase (SERCA2) pump activity in isolated cardiomyocytes and in vivo, leading to depressed myocyte calcium kinetics and mechanics. Conversely, downregulation of HAX-1 enhanced calcium cycling and contractility. The inhibitory effects of HAX-1 were abolished upon phosphorylation of phospholamban, which plays a fundamental role in controlling basal contractility and constitutes a key downstream effector of the beta-adrenergic signaling cascade. Mechanistically, HAX-1 promoted formation of phospholamban monomers, the active/inhibitory units of the calcium pump. Indeed, ablation of PLN rescued HAX-1 inhibition of contractility in vivo. Thus, HAX-1 represents a regulatory mechanism in cardiac calcium cycling and its responses to sympathetic stimulation, implicating its importance in calcium homeostasis and cell survival.


Assuntos
Apoptose , Testes de Função Cardíaca , Coração/fisiologia , Proteínas/metabolismo , Envelhecimento/metabolismo , Animais , Transporte Biológico , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/metabolismo , Regulação para Baixo , Transferência Ressonante de Energia de Fluorescência , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Transgênicos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Especificidade de Órgãos , Ligação Proteica , Ratos , Retículo Sarcoplasmático/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transgenes
14.
Am J Physiol Heart Circ Physiol ; 301(4): H1588-95, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21742996

RESUMO

The histidine-rich calcium binding protein (HRC) Ser96Ala polymorphism was shown to correlate with ventricular arrhythmias and sudden death only in dilated cardiomyopathy patients but not in healthy human carriers. In the present study, we assessed the molecular and cellular mechanisms underlying human arrhythmias by adenoviral expression of the human wild-type (HRC(WT)) or mutant HRC (HRC(S96A)) in adult rat ventricular cardiomyocytes. Total HRC protein was increased by ∼50% in both HRC(WT)- and HRC(S96A)-infected cells. The HRC(S96A) mutant exacerbated the inhibitory effects of HRC(WT) on the amplitude of Ca(2+) transients, prolongation of Ca(2+) decay time, and caffeine-induced sarcoplasmic reticulum Ca(2+) release. Consistent with these findings, HRC(S96A) reduced maximal sarcoplasmic reticulum calcium uptake rate to a higher extent than HRC(WT). Furthermore, the frequency of spontaneous Ca(2+) sparks, which was reduced by HRC(WT), was increased by mutant HRC(S96A) under resting conditions although there were no spontaneous Ca(2+) waves under stress conditions. However, expression of the HRC(S96A) genetic variant in cardiomyocytes from a rat model of postmyocardial infarction heart failure induced dramatic disturbances of rhythmic Ca(2+) transients. These findings indicate that the HRC Ser96Ala variant increases the propensity of arrhythmogenic Ca(2+) waves in the stressed failing heart, suggesting a link between this genetic variant and life-threatening ventricular arrhythmias in human carriers.


Assuntos
Arritmias Cardíacas/induzido quimicamente , Proteínas de Ligação ao Cálcio/genética , Catecolaminas , Insuficiência Cardíaca/induzido quimicamente , Miócitos Cardíacos/efeitos dos fármacos , Adenoviridae/genética , Substituição de Aminoácidos , Animais , Arritmias Cardíacas/genética , Western Blotting , Cálcio/metabolismo , Cálcio/fisiologia , Sinalização do Cálcio/genética , Sinalização do Cálcio/fisiologia , DNA Complementar/biossíntese , DNA Complementar/genética , Eletrocardiografia , Expressão Gênica , Células HEK293 , Insuficiência Cardíaca/genética , Humanos , Imunoprecipitação , Masculino , Mutação Puntual/genética , Mutação Puntual/fisiologia , Polimorfismo Genético/genética , Ratos , Ratos Wistar , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética
15.
Cell Regen ; 10(1): 4, 2021 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-33527149

RESUMO

Heart disease is the leading cause of mortality worldwide. Due to the limited proliferation rate of mature cardiomyocytes, adult mammalian hearts are unable to regenerate damaged cardiac muscle following injury. Instead, injured area is replaced by fibrotic scar tissue, which may lead to irreversible cardiac remodeling and organ failure. In contrast, adult zebrafish and neonatal mammalian possess the capacity for heart regeneration and have been widely used as experimental models. Recent studies have shown that multiple types of cells within the heart can respond to injury with the activation of distinct signaling pathways. Determining the specific contributions of each cell type is essential for our understanding of the regeneration network organization throughout the heart. In this review, we provide an overview of the distinct functions and coordinated cell behaviors of several major cell types including cardiomyocytes, endocardial cells, epicardial cells, fibroblasts, and immune cells. The topic focuses on their specific responses and cellular plasticity after injury, and potential therapeutic applications.

16.
Dev Cell ; 50(6): 729-743.e5, 2019 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-31402282

RESUMO

Pacemaker cardiomyocytes that create the sinoatrial node are essential for the initiation and maintenance of proper heart rhythm. However, illuminating developmental cues that direct their differentiation has remained particularly challenging due to the unclear cellular origins of these specialized cardiomyocytes. By discovering the origins of pacemaker cardiomyocytes, we reveal an evolutionarily conserved Wnt signaling mechanism that coordinates gene regulatory changes directing mesoderm cell fate decisions, which lead to the differentiation of pacemaker cardiomyocytes. We show that in zebrafish, pacemaker cardiomyocytes derive from a subset of Nkx2.5+ mesoderm that responds to canonical Wnt5b signaling to initiate the cardiac pacemaker program, including activation of pacemaker cell differentiation transcription factors Isl1 and Tbx18 and silencing of Nkx2.5. Moreover, applying these developmental findings to human pluripotent stem cells (hPSCs) notably results in the creation of hPSC-pacemaker cardiomyocytes, which successfully pace three-dimensional bioprinted hPSC-cardiomyocytes, thus providing potential strategies for biological cardiac pacemaker therapy.


Assuntos
Proteína Homeobox Nkx-2.5/metabolismo , Mesoderma/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Animais , Sequência de Bases , Bioimpressão , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mutação com Perda de Função/genética , Modelos Cardiovasculares , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco/metabolismo , Peixe-Zebra
17.
PLoS One ; 10(7): e0132567, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26186000

RESUMO

Mitochondrial flash (mitoflash) is a highly-conserved, universal, and physiological mitochondrial activity in isolated mitochondria, intact cells, and live organisms. Here we investigated developmental and disease-related remodeling of mitoflash activity in zebrafish skeletal muscles. In transgenic zebrafish expressing the mitoflash reporter cpYFP, in vivo imaging revealed that mitoflash frequency and unitary properties underwent multiphasic and muscle type-specific changes, accompanying mitochondrial morphogenesis from 2 to 14 dpf. In particular, short (S)-type mitoflashes predominated in early muscle formation, then S-, transitory (T)- and regular (R)-type mitoflashes coexisted during muscle maturation, followed by a switch to R-type mitoflashes in mature skeletal muscles. In early development of muscular dystrophy, we found accelerated S- to R-type mitoflash transition and reduced mitochondrial NAD(P)H amidst a remarkable cell-to-cell heterogeneity. This study not only unravels a profound functional and morphological remodeling of mitochondria in developing and diseased skeletal muscles, but also underscores mitoflashes as a useful reporter of mitochondrial function in milieu of live animals under physiological and pathophysiological conditions.


Assuntos
Proteínas de Membrana/genética , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Proteínas de Peixe-Zebra/genética , Actinas/genética , Actinas/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Mitocôndrias/patologia , Morfolinos/genética , Morfolinos/metabolismo , Desenvolvimento Muscular/genética , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , NADP/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Imagem com Lapso de Tempo , Peixe-Zebra , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/metabolismo
18.
Cell Res ; 24(9): 1091-107, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25124925

RESUMO

While the adult human heart has very limited regenerative potential, the adult zebrafish heart can fully regenerate after 20% ventricular resection. Although previous reports suggest that developmental signaling pathways such as FGF and PDGF are reused in adult heart regeneration, the underlying intracellular mechanisms remain largely unknown. Here we show that H2O2 acts as a novel epicardial and myocardial signal to prime the heart for regeneration in adult zebrafish. Live imaging of intact hearts revealed highly localized H2O2 (~30 µM) production in the epicardium and adjacent compact myocardium at the resection site. Decreasing H2O2 formation with the Duox inhibitors diphenyleneiodonium (DPI) or apocynin, or scavenging H2O2 by catalase overexpression markedly impaired cardiac regeneration while exogenous H2O2 rescued the inhibitory effects of DPI on cardiac regeneration, indicating that H2O2 is an essential and sufficient signal in this process. Mechanistically, elevated H2O2 destabilized the redox-sensitive phosphatase Dusp6 and hence increased the phosphorylation of Erk1/2. The Dusp6 inhibitor BCI achieved similar pro-regenerative effects while transgenic overexpression of dusp6 impaired cardiac regeneration. H2O2 plays a dual role in recruiting immune cells and promoting heart regeneration through two relatively independent pathways. We conclude that H2O2 potentially generated from Duox/Nox2 promotes heart regeneration in zebrafish by unleashing MAP kinase signaling through a derepression mechanism involving Dusp6.


Assuntos
Coração/fisiologia , Peróxido de Hidrogênio/farmacologia , Regeneração/efeitos dos fármacos , Animais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Coração/efeitos dos fármacos , Técnicas In Vitro , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Modelos Biológicos , Miocárdio/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
19.
Med Eng Phys ; 35(11): 1584-90, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23727155

RESUMO

Needle biopsy is a medical procedure to extract tissue for diagnosis of cancer and other diseases. The quality of tissue samples acquired by needle biopsy greatly depends on the cutting forces of the cannula. The reduction of cutting forces is crucial for obtaining good tissue samples. There exist many factors that influence the cutting forces, some of which include the cannula tip geometry, translation speed, and rotation speed. In the present paper, the effects of rotating the cannula on tissue cutting for needle biopsy are studied. A fracture-mechanics-based approach is used to analyze the cutting forces. Analysis has shown that the cutting forces decrease with the increases in the slice/push ratio defined as the ratio of speed component parallel to the cutting edge/speed perpendicular to the cutting edge. Experiments are performed to demonstrate this phenomenon. Mathematical models of the slice/push ratio for bevel tip cannulas are formulated. The results are used to determine the optimal cannula rotation/translation speed and the desired tip geometry for needle biopsy. It is shown that a minimal slice/push ratio of 2 is recommended. A cannula with a large bevel angle is more suitable for rotational needle biopsy.


Assuntos
Biópsia por Agulha/instrumentação , Rotação , Fenômenos Mecânicos , Modelos Teóricos
20.
Methods Mol Biol ; 1037: 463-73, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24029953

RESUMO

Despite current treatment regimens, heart failure still remains one of the leading causes of morbidity and mortality in the world due to failure to adequately replace lost ventricular myocardium from ischemia-induced infarct. Although adult mammalian ventricular cardiomyocytes have a limited capacity to divide, this proliferation is insufficient to overcome the significant loss of myocardium from ventricular injury. However, lower vertebrates, such as the zebrafish and newt, have the remarkable capacity to fully regenerate their hearts after severe injury. Thus, there is great interest in studying these animal model systems to discover new regenerative approaches that might be applied to injured mammalian hearts. To this end, the zebrafish has been utilized more recently to gain additional mechanistic insight into cardiac regeneration because of its genetic tractability. Here, we describe two cardiac injury methods, a mechanical and a genetic injury model, for studying cardiac regeneration in the zebrafish.


Assuntos
Traumatismos Cardíacos/etiologia , Regeneração , Peixe-Zebra , Animais , Modelos Animais de Doenças
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA