Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 644: 70-78, 2023 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-36634584

RESUMEN

During myocardial infarction, cellular debris is released, causing a sterile inflammation via pattern recognition receptors. These reactions amplify damage and promotes secondary heart failure. The pattern recognition receptor, Toll-like receptor 9 (TLR9) detects immunogenic fragments of endogenous DNA, inducing inflammation by NFκB. The p66ShcA adaptor protein plays an important role in both ischemic myocardial damage and immune responses. We hypothesized that p66ShcA adaptor protein promotes DNA-sensing signaling via the TLR9 pathway after myocardial infarction. TLR9 protein expression increased in cardiac tissue from patients with end-stage heart failure due to ischemic heart disease. Myocardial ischemia in mice in vivo induced gene expression of key TLR9 pathway proteins (MyD88 and Unc93b1). In this model, a functional link between TLR9 and p66ShcA was revealed as; (i) ischemia-induced upregulation of TLR9 protein was abrogated in myocardium of p66ShcA knockout mice; (ii) when p66ShcA was overexpressed in NFkB reporter cells stably expressing TLR9, NFkB-activation increased during stimulation with the TLR9 agonist CpG B; (iii) in cardiac fibroblasts, p66ShcA overexpression caused TLR9 upregulation. Co-immunoprecipitation showed that ShcA proteins and TLR9 may be found in the same protein complex, which was dissipated upon TLR9 stimulation in vivo. A proximity assay confirmed the co-localization of TLR9 and ShcA proteins. The systemic immune response after myocardial ischemia was dampened in p66ShcA knockout mice as interleukin-4, -17 and -22 expression in mononuclear cells isolated from spleens was reduced. In conclusion, p66ShcA adaptor may be an interaction partner and a regulator of the TLR9 pathway post-infarction.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Isquemia Miocárdica , Animales , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inflamación , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo , FN-kappa B/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Receptor Toll-Like 9/metabolismo
2.
Scand J Clin Lab Invest ; 77(5): 321-331, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28460577

RESUMEN

We aimed to study the cardiac expression of bone morphogenetic protein 2, its receptor 1 b, and connective tissue growth factor, factors implicated in cardiac embryogenesis, following ischemia/hypoxia, heart failure, and in remodeling hearts from humans and mice. Biopsies from the left ventricle of patients with end-stage heart failure due to dilated cardiomyopathy or coronary artery disease were compared with donor hearts and biopsies from patients with normal heart function undergoing coronary artery bypass grafting. Mouse model of post-infarction remodeling was made by permanent ligation of the left coronary artery. Hearts were analyzed by real-time polymerase chain reaction and Western blotting after 24 hours and after 2 and 4 weeks. Patients with dilated cardiomyopathy and mice post-infarction had increased cardiac expression of connective tissue growth factor. Bone morphogenetic protein 2 was increased in human hearts failing due to coronary artery disease and in mice post-infarction. Gene expression of bone morphogenetic protein receptor 1 beta was reduced in hearts of patients with failure, but increased two weeks following permanent ligation of the left coronary artery in mice. In conclusion, connective tissue growth factor is upregulated in hearts of humans with dilated cardiomyopathy, bone morphogenetic protein 2 is upregulated in remodeling due to myocardial infarction while its receptor 1 b in human failing hearts is downregulated. A potential explanation might be an attempt to engage regenerative processes, which should be addressed by further, mechanistic studies.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Cardiomiopatía Dilatada/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Enfermedad de la Arteria Coronaria/genética , Insuficiencia Cardíaca/genética , Adulto , Anciano , Animales , Proteína Morfogenética Ósea 2/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Puente de Arteria Coronaria , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Pruebas de Función Cardíaca , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miocardio/metabolismo , Miocardio/patología , Transducción de Señal
3.
Basic Res Cardiol ; 111(4): 42, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27164906

RESUMEN

Acute myocardial infarction (AMI) causes sterile inflammation, which exacerbates tissue injury. Elevated levels of circulating mitochondrial DNA (mtDNA) have been associated with AMI. We hypothesized that mtDNA triggers an innate immune response via TLR9 and NF-κB activation, causing cardiomyocyte injury. Murine cardiomyocytes express TLR9 mRNA and protein and were able to internalize fluorescently labeled mouse mtDNA. Incubation of human embryonic kidney cells with serum from AMI patients containing naturally elevated levels of mtDNA induced TLR9-dependent NF-κB activity. This effect was mimicked by isolated mtDNA. mtDNA activated NF-κB in reporter mice both in vivo and in isolated cardiomyocytes. Moreover, incubation of isolated cardiomyocytes with mtDNA induced cell death after 4 and 24 h. Laser confocal microscopy showed that incubation of cardiomyocytes with mtDNA accelerated mitochondrial depolarization induced by reactive oxygen species. In contrast to mtDNA, isolated total DNA did not activate NF-κB nor induce cell death. In conclusion, mtDNA can induce TLR9-dependent NF-κB activation in reporter cells and activate NF-κB in cardiomyocytes. In cardiomyocytes, mtDNA causes mitochondrial dysfunction and death. Endogenous mtDNA in the extracellular space is a danger signal with direct detrimental effects on cardiomyocytes.


Asunto(s)
ADN Mitocondrial/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , FN-kappa B/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Muerte Celular/fisiología , Femenino , Humanos , Immunoblotting , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Infarto del Miocardio/metabolismo , Reacción en Cadena de la Polimerasa
4.
Basic Res Cardiol ; 110(2): 13, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25680868

RESUMEN

Heart rupture and heart failure are deleterious complications of myocardial infarction. The ShcA gene encodes for three protein isoforms, p46-, p52- and p66ShcA. p66ShcA induces oxidative stress. We studied the role of p66ShcA post-infarction. Expression of p66ShcA was analyzed in myocardium of patients with stable angina (n = 11), in explanted hearts with end-stage ischemic heart failure (n = 9) and compared to non-failing hearts not suitable for donation (n = 7). p66ShcA was increased in the patients with stable angina, but not in the patients with end-stage heart failure. Mice (n = 105) were subjected to coronary artery ligation. p66ShcA expression and phosphorylation were evaluated over a 6-week period. p66ShcA expression increased transiently during the first weeks post-infarction. p66ShcA knockout mice (KO) were compared to wild type (n = 82 in total). KO had improved survival and reduced occurrence of heart rupture post-infarction. Expression of cardiac matrix metalloproteinase 2 (MMP-2) was reduced; fibroblast activation and collagen accumulation were facilitated, while oxidative stress was attenuated in KO early post-infarction. 6 weeks post-infarction, reactive fibrosis and left ventricular dilatation were diminished in KO. p66ShcA regulation of MMP-2 was demonstrated in cultured fibroblasts: lack or overexpression of p66ShcA in vitro altered expression of MMP-2. Myocardial infarction induced cardiac p66ShcA. Deletion of p66ShcA improved early survival, myocardial healing and reduced cardiac fibrosis. Upon myocardial infarction p66ShcA regulates MMP-2 activation. The role of p66ShcA in human cardiac disease deserves further study as a potential target for reducing adverse cardiac remodeling post-infarction.


Asunto(s)
Infarto del Miocardio/metabolismo , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Anciano , Animales , Western Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Masculino , Metaloproteinasa 2 de la Matriz/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Estrés Oxidativo/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Remodelación Ventricular/fisiología
5.
Stem Cells ; 32(8): 2123-34, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24578312

RESUMEN

Cardioprotection can be evoked through extracardiac approaches. This prompted us to investigate whether remote transplantation of stem cells confers protection of the heart against ischemic injury. The cardioprotective effect of subcutaneous transplantation of naïve versus heme oxygenase-1 (HMOX-1)-overexpressing mouse mesenchymal stem cells (MSC) to mice was investigated in hearts subjected to ischemia-reperfusion in a Langendorff perfusion system. Mice were transplanted into the interscapular region with naïve or HMOX-1 transfected MSC isolated from transgenic luciferase reporter mice and compared to sham-treated animals. The fate of transplanted cells was followed by in vivo bioluminescence imaging, revealing that MSC proliferated, but did not migrate detectably from the injection site. Ex vivo analysis of the hearts showed that remote transplantation of mouse adipose-derived MSC (mASC) resulted in smaller infarcts and improved cardiac function after ischemia-reperfusion compared to sham-treated mice. Although HMOX-1 overexpression conferred cytoprotective effects on mASC against oxidative stress in vitro, no additive beneficial effect of HMOX-1 transfection was noted on the ischemic heart. Subcutaneous transplantation of MSC also improved left ventricular function when transplanted in vivo after myocardial infarction. Plasma analysis and gene expression profile of naïve- and HMOX-1-mASC after transplantation pointed toward pentraxin 3 as a possible factor involved in the remote cardioprotective effect of mASC. These results have significant implications for understanding the behavior of stem cells after transplantation and development of safe and noninvasive cellular therapies with clinical applications. Remote transplantation of MSC can be considered as an alternative procedure to induce cardioprotection.


Asunto(s)
Terapia Genética/métodos , Hemo-Oxigenasa 1/administración & dosificación , Proteínas de la Membrana/administración & dosificación , Trasplante de Células Madre Mesenquimatosas/métodos , Infarto del Miocardio/complicaciones , Daño por Reperfusión/prevención & control , Animales , Western Blotting , Proteína C-Reactiva/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Inmunohistoquímica , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Estrés Oxidativo/fisiología , Transfección
6.
Basic Res Cardiol ; 109(6): 450, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25344086

RESUMEN

Activation of heme oxygenase-1 (HO-1), a heme-degrading enzyme responsive to a wide range of cellular stress, is traditionally considered to convey adaptive responses to oxidative stress, inflammation and vasoconstriction. These diversified effects are achieved through the degradation of heme to carbon monoxide (CO), biliverdin (which is rapidly converted to bilirubin by biliverdin reductase) and ferric iron. Recent findings have added antiproliferative and angiogenic effects to the list of HO-1/CO actions. HO-1 along with its reaction products bilirubin and CO are protective against ischemia-induced injury (myocardial infarction, ischemia-reperfusion (IR)-injury and post-infarct structural remodelling). Moreover, HO-1, and CO in particular, possess acute antihypertensive effects. As opposed to these curative potentials, the long-believed protective effect of HO-1 in cardiac remodelling in response to pressure overload and type 2 diabetes mellitus (DM) has been questioned by recent work. These challenges, coupled with emerging regulatory mechanisms, motivate further in-depth studies to help understand untapped layers of HO-1 regulation and action. The outcomes of these efforts may shed new light on critical mechanisms that could be used to harness the protective potential of this enzyme for the therapeutic benefit of patients suffering from such highly prevalent cardiovascular disorders.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/etiología , Hemo-Oxigenasa 1/fisiología , Humanos
7.
J Mol Cell Cardiol ; 56: 22-33, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23238222

RESUMEN

Aquaporin-1 (AQP1) is expressed in human and mouse hearts, but little is known about its cellular and subcellular localization and regulation. The aim of this study was to investigate the localization of AQP1 in the mouse heart and to determine the effects of ischemia and hypoxia on its expression. Mouse myocardial cells were freshly isolated and split into cardiomyocyte and non-cardiomyocyte fractions. Isolated, Langendorff-perfused C57Bl6 mouse hearts (n=46) were harvested with no intervention, subjected to 35min of ischemia or ischemia followed by 60min of reperfusion. Eleven mouse hearts were perfusion-fixed for electron microscopy. Forty C57Bl6 mice were exposed to normobaric hypoxia for one or two weeks (n=12). Needle biopsies of human left ventricular myocardium were sampled (n=30) during coronary artery bypass surgery before cardioplegia and after 30min of reperfusion. Human umbilical vein endothelial cells (HUVECs) were subjected to 4h of hypoxia with reoxygenation for either 4 or 24h. AQP1 expression was studied by electron microscopy with immunogold labeling, Western blot, and qPCR. Expression of miR-214 and miR-320 in HUVECs with hypoxia was studied with qPCR. HUVECs were then transfected with precursors and inhibitors of miR-214. AQP1 expression was confined to cardiac endothelial cells, with no signal in cardiomyocytes or cardiac fibroblasts. Immunogold electron microscopy showed AQP1 expression in endothelial caveolae with equal distribution along the basal and apical membranes. Ischemia and reperfusion tended to decrease AQP1 mRNA expression in mouse hearts by 37±9% (p=0.06), while glycosylated AQP1 protein was reduced by 16±9% (p=0.03). No difference in expression was found between ischemia alone and ischemia-reperfusion. In human left ventricles AQP1 mRNA expression was reduced following cardioplegia and reperfusion (p=0.008). Hypoxia in mice reduced AQP1 mRNA expression by 20±7% (p<0.0001), as well as both glycosylated (-47±10%, p=0.03) and glycan-free protein (-34±16%, p=0.05). Hypoxia and reoxygenation in HUVECs downregulated glycan-free AQP1 protein (-34±24%, p=0.04) and upregulated miR-214 (+287±52%, p<0.05). HUVECs transfected with anti-miR-214 had increased glycosylated (1.5 fold) and glycan-free (2 fold) AQP1. AQP1 in mouse hearts is localized to endothelial cell membranes and caveolae. Cardioplegia, ischemia and hypoxia decrease AQP1 mRNA as well as total protein expression and glycosylation, possibly regulated by miR-214.


Asunto(s)
Acuaporina 1/metabolismo , Regulación hacia Abajo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Isquemia Miocárdica/metabolismo , Miocardio/patología , Animales , Acuaporina 1/genética , Caveolas/metabolismo , Hipoxia de la Célula , Fibroblastos/metabolismo , Expresión Génica , Glicosilación , Paro Cardíaco Inducido , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Procesamiento Proteico-Postraduccional , Interferencia de ARN
8.
Mol Pharmacol ; 84(3): 372-83, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23778361

RESUMEN

Myocardial connective tissue growth factor (CTGF/CCN2) is induced in heart failure, a condition associated with diminution of ß-adrenergic receptor (ß-AR) responsiveness. Accordingly, we aimed to investigate whether CTGF could play a mechanistic role in regulation of ß-AR responsiveness. Concentration-response curves of isoproterenol-stimulated cAMP generation in cardiomyocytes from transgenic mice with cardiac-restricted overexpression of CTGF (Tg-CTGF) or cardiomyocytes pretreated with recombinant human CTGF (rec-hCTGF) revealed marked reduction of both ß1-AR and ß2-AR responsiveness. Consistently, ventricular muscle strips from Tg-CTGF mice stimulated with isoproterenol displayed attenuation of maximal inotropic responses. However, no differences of maximal inotropic responses of myocardial fibers from Tg-CTGF mice and nontransgenic littermate control (NLC) mice were discerned when stimulated with supramaximal concentrations of dibutyryl-cAMP, indicating preserved downstream responsiveness to cAMP. Congruent with a mechanism of desensitization of ß-ARs, mRNA and protein levels of G protein-coupled receptor kinase 5 (GRK5) were found isoform-selective upregulated in both cardiomyocytes from Tg-CTGF mice and cardiomyocytes exposed to rec-hCTGF. Corroborating a mechanism of GRK5 in CTGF-mediated control of ß-AR sensitivity, Chinese hamster ovary cells pretreated with rec-hCTGF displayed increased agonist- and biased ligand-stimulated ß-arrestin binding to ß-ARs. Despite increased sensitivity of cardiomyocytes from GRK5-knockout (KO) mice to ß-adrenergic agonists, pretreatment of GRK5-KO cardiomyocytes with rec-hCTGF, as opposed to cardiomyocytes from wild-type mice, did not alter ß-AR responsiveness. Finally, Tg-CTGF mice subjected to chronic exposure (14 days) to isoproterenol revealed blunted myocardial hypertrophy and preserved cardiac function versus NLC mice. In conclusion, this study uncovers a novel mechanism controlling ß-AR responsiveness in cardiomyocytes involving CTGF-mediated regulation of GRK5.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/biosíntesis , Corazón/efectos de los fármacos , Isoproterenol/toxicidad , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Agonistas Adrenérgicos/farmacología , Animales , Arrestinas/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cardiomegalia/inducido químicamente , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Cricetinae , Cricetulus , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Expresión Génica , Corazón/fisiopatología , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Contracción Miocárdica/efectos de los fármacos , Fosfoproteínas/metabolismo , Fosforilación , Ratas , Proteínas Recombinantes/farmacología , beta-Arrestinas
9.
Basic Res Cardiol ; 108(6): 393, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24158693

RESUMEN

Aquaporins are a group of proteins with high-selective permeability for water. A subgroup called aquaglyceroporins is also permeable to glycerol, urea and a few other solutes. Aquaporin function has mainly been studied in the brain, kidney, glands and skeletal muscle, while the information about aquaporins in the heart is still scarce. The current review explores the recent advances in this field, bringing aquaporins into focus in the context of myocardial ischemia, reperfusion, and blood osmolarity disturbances. Since the amount of data on aquaporins in the heart is still limited, examples and comparisons from better-studied areas of aquaporin biology have been used. The human heart expresses aquaporin-1, -3, -4 and -7 at the protein level. The potential roles of aquaporins in the heart are discussed, and some general phenomena that the myocardial aquaporins share with aquaporins in other organs are elaborated. Cardiac aquaporin-1 is mostly distributed in the microvasculature. Its main role is transcellular water flux across the endothelial membranes. Aquaporin-4 is expressed in myocytes, both in cardiac and in skeletal muscle. In addition to water flux, its function is connected to the calcium signaling machinery. It may play a role in ischemia-reperfusion injury. Aquaglyceroporins, especially aquaporin-7, may serve as a novel pathway for nutrient delivery into the heart. They also mediate toxicity of various poisons. Aquaporins cannot influence permeability by gating, therefore, their function is regulated by changes of expression-on the levels of transcription, translation (by microRNAs), post-translational modification, membrane trafficking, ubiquitination and subsequent degradation. Studies using mice genetically deficient for aquaporins have shown rather modest changes in the heart. However, they might still prove to be attractive targets for therapy directed to reduce myocardial edema and injury caused by ischemia and reperfusion.


Asunto(s)
Acuaporinas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Animales , Corazón , Humanos , Isquemia Miocárdica/metabolismo
10.
Basic Res Cardiol ; 108(1): 323, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23266787

RESUMEN

Liver X receptor (LXR)-α and -ß play a major role in lipid and glucose homeostasis. Their expression and function in the heart is not well characterized. Our aim was to describe the expression of LXRs in the murine heart, and to determine effects of cardiac LXR activation on target gene expression, lipid homeostasis and ischemia. Both LXRα and -ß were expressed in heart tissues, HL-1 cells and isolated cardiomyocytes as determined by qRT-PCR. Elevated cardiac expression of LXR target genes and LXRß was observed 24 h after in vivo permanent coronary artery ligation. The synthetic LXR agonist GW3965 induced mRNA expression of the LXR target genes in HL-1 cells and isolated cardiomyocytes. This was associated with a buildup of intracellular triglycerides and expanding lipid droplets as quantified by confocal microscopy. Mice injected with GW3965 had cardiac LXR activation as judged by increased target gene expression and lipid droplet accumulation. GW3965 in vivo and in vitro increased expression of genes inducing triglyceride synthesis, and altered expression of lipid droplet-binding protein genes. GW3965 protected HL-1 cells against hypoxia-reoxygenation induced apoptosis. LXR activation by GW3965 in vivo prior to heart isolation and perfusion with induced global ischemia and reperfusion improved left ventricular contractile function and decreased infarct size. In conclusion, LXRs are expressed in the murine heart in the basal state, and are activated by myocardial infarction. Activation of LXR by the synthetic agonist GW3965 is associated with intracardiac accumulation of lipid droplets and protection against myocardial ischemia-reperfusion injury.


Asunto(s)
Metabolismo de los Lípidos , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Receptores Nucleares Huérfanos/fisiología , Animales , Apoptosis/efectos de los fármacos , Benzoatos/farmacología , Bencilaminas/farmacología , Células Cultivadas , Espacio Intracelular/metabolismo , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Triglicéridos/metabolismo
11.
Biochem Biophys Res Commun ; 425(1): 70-5, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22820194

RESUMEN

PURPOSE: Hyperosmolarity is a common complication in intensive care patients, dysregulating water balance in many organs including brain and heart. The aquaporin (AQP) water channels, in particular AQP1 and -4, have been suggested to play an important role in fluid homeostasis of the myocardium. In many organs AQP expression is regulated by osmolarity, drastically altering water permeability of the cell membranes. The aim of our study was to investigate if plasma hyperosmolality may regulate cardiac expression of AQP1 and -4, and if so, at which magnitude and time frame such regulation takes place. METHODS: C57Bl6 mice were injected intraperitoneally with either 1.5 ml 0.154 Mol (isoosmotic), 0.5 ml 1 Mol (mild hyperosmotic) or 0.5 ml 2 Mol (strong hyperosmotic) NaCl. Plasma, hearts, and forebrains were harvested before injection ("time 0"), and after 1, 4, 8 and 24 h. AQP1 and -4 expression were analyzed using qPCR and Western blot. RESULTS: Isoosmotic and mild hyperosmotic injections caused no important changes in cardiac AQP expression. Strong hyperosmotic NaCl injections induced an upregulation of AQP1 mRNA and glycosylated fraction of AQP1 protein in the heart without changes of the total protein. AQP4 mRNA and protein decreased in the heart and increased in the brain after hyperosmotic NaCl. The change in AQP4 protein content in the brain preceded the increase of mRNA. CONCLUSION: As in the brain, expression of AQP1 and -4 in the heart is influenced by changes in plasma osmolality. Changes in AQP expression may alter cardiac function in hyperosmotic states.


Asunto(s)
Acuaporina 1/biosíntesis , Acuaporina 4/biosíntesis , Miocardio/metabolismo , Plasma/fisiología , Animales , Encéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Concentración Osmolar
12.
Basic Res Cardiol ; 107(5): 280, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22777185

RESUMEN

Aquaporins (AQPs) are channel-forming membrane proteins highly permeable to water. AQP4 is found in mammalian hearts; however, its expression sites, regulation and function are largely unknown. The aim was to investigate cardiac AQP4 expression in humans and mice, its regulation by ischemia and hypoxia, and in particular its role in cardiac ischemic injury using AQP4 knockout (KO) mice. Comparable levels of AQP4 were detected by Western blot and qPCR in biopsies from human donor hearts and wild type C57Bl6 mouse hearts. In mice, AQP4 was expressed on cardiomyocyte plasmalemma (qPCR, Western blot, immunogold), and its mRNA decreased following ischemia/reperfusion (isolated hearts, p = 0.02) and after normobaric hypoxia in vivo (oxygen fraction 10 % for 1 week, p < 0.001). Isolated hearts from AQP4 KO mice undergoing global ischemia and reperfusion had reduced infarct size (p = 0.05) and attenuated left ventricular end-diastolic pressure during reperfusion (p = 0.04). Infarct size was also reduced in AQP4 KO mice 24 h after left coronary artery ligation in vivo (p = 0.036). AQP4 KO hearts had no compensatory change in AQP1 protein expression. AQP4 KO cardiomyocytes were partially resisted to hypoosmotic stress in the presence of hypercontracture. AQP4 is expressed in human and mouse hearts, in the latter confined to the cardiomyocyte plasmalemma. AQP4 mRNA expression is downregulated by hypoxia and ischemia. Deletion of AQP4 is protective in acute myocardial ischemia-reperfusion, and this molecule might be a future target in the treatment of acute myocardial infarction.


Asunto(s)
Acuaporina 4/fisiología , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Animales , Acuaporina 1/genética , Acuaporina 4/análisis , Acuaporina 4/genética , Supervivencia Celular , Regulación hacia Abajo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Inmunoelectrónica , Miocitos Cardíacos/metabolismo , ARN Mensajero/análisis
13.
Am J Physiol Heart Circ Physiol ; 300(4): H1291-302, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21186275

RESUMEN

CCN2/connective tissue growth factor (CTGF), a CCN family matricellular protein repressed in healthy hearts after birth, is induced in heart failure of various etiologies. Multiple cellular and biological functions have been assigned to CCN2/CTGF depending on cellular context. However, the functions and mechanisms of action of CCN2/CTGF in the heart as well as its roles in cardiac physiology and pathophysiology remain unknown. Transgenic mice with cardiac-restricted overexpression of CTGF (Tg-CTGF) were generated and compared with nontransgenic littermate control (NLC) mice. Tg-CTGF mice displayed slightly lower cardiac mass and inconspicuous increase of myocardial collagen compared with NLC mice but no evidence of contractile dysfunction. Analysis of the myocardial transcriptome by DNA microarray revealed activation of several distinct gene programs in Tg-CTGF hearts involved in cardioprotection and growth inhibition. Indeed, Tg-CTGF mice subjected to ischemia-reperfusion injury by in situ transient occlusion of the left anterior descending coronary artery in vivo displayed reduced vulnerability with markedly diminished infarct size. These findings were recapitulated in isolated hearts perfused with recombinant human (h)CTGF before the ischemia-reperfusion procedure. Consistently, Tg-CTGF hearts, as well as isolated adult cardiac myocytes exposed to recombinant hCTGF, displayed enhanced phosphorylation and activity of the Akt/p70S6 kinase/GSK-3ß salvage kinase pathway and induction of several genes with reported cardioprotective functions. Inhibition of Akt activities also prevented the cardioprotective phenotype of hearts from Tg-CTGF mice. This report provides novel evidence that CTGF confers cardioprotection by salvage phosphokinase signaling leading to inhibition of GSK-3ß activities, activation of phospho-SMAD2, and reprogramming of gene expression.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/farmacología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/fisiopatología , Animales , Cardiotónicos/farmacología , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Perfilación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones , Ratones Transgénicos , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Proteína Smad2/metabolismo
14.
Heart Fail Rev ; 16(1): 71-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20694832

RESUMEN

A wide variety of cardiac disease states can induce remodelling and lead to the functional consequence of heart failure. These complex disease states involve a plethora of parallel signal transduction events, which may be associated with tissue injury or tissue repair. Innate immunity is activated in hearts injured in different ways, evident as cytokine release from the heart, activation of toll-like receptors involved in recognizing danger, and activation of the transcription factor nuclear factor kappa B. Nuclear factor kappa B regulates gene programmes involved in inflammation as well as the resolution of inflammation. The impact of this is an enigma; while cytokines, toll-like receptors, and nuclear factor kappa B appear to elicit myocardial protection in studies of preconditioning, the literature strongly indicates a detrimental role for activation of innate immunity in studies of acute ischaemia-reperfusion injury. The impact of activation of cardiac innate immunity on the long-term outcome in in vivo models of hypertrophy and remodelling is less clear, with conflicting results as to whether it is beneficial or detrimental. More research using genetically engineered mice as tools, different models of evoking remodelling, and long-term follow-up is required for us to conclude whether activation of the innate immune system is good, bad, or unimportant in chronic injury models.


Asunto(s)
Insuficiencia Cardíaca/inmunología , Inmunidad Innata , Miocardio/inmunología , Remodelación Ventricular , Citocinas/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Miocardio/patología , FN-kappa B/inmunología , Receptores Toll-Like/metabolismo
15.
Basic Res Cardiol ; 104(1): 22-32, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18931966

RESUMEN

Myocardial adaptation to ischemia in the form of ischemic preconditioning is clinically attractive, but not directly usable until molecular mimics are discovered. A growing body of evidence indicates that events underlying myocardial adaptation to ischemia may either involve, or be parallel to, signaling of the innate immune response. Preconditioning-like protection of the heart can be evoked through giving cytokines or fragments of bacterial walls. A possible role for cytokines, toll-like receptors, and nuclear factor kappa B for evoking ischemic preconditioning are discussed. Through stimulating innate immunity, there is potential to bring preconditioning into the clinics in a reasonable time frame. The possibility that the underlying protective response may involve adaptive immunity through danger signaling is briefly reviewed.


Asunto(s)
Corazón/fisiopatología , Inmunidad Innata/fisiología , Isquemia Miocárdica/inmunología , Isquemia Miocárdica/fisiopatología , Aclimatación , Corazón/fisiología , Humanos , Precondicionamiento Isquémico Miocárdico/efectos adversos , Modelos Cardiovasculares , Factor 88 de Diferenciación Mieloide/fisiología , Isquemia Miocárdica/prevención & control , FN-kappa B/fisiología , Transducción de Señal , Receptores Toll-Like/fisiología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/fisiología
16.
Br J Pharmacol ; 176(22): 4360-4372, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31412132

RESUMEN

BACKGROUND AND PURPOSE: Cellular debris causes sterile inflammation after myocardial infarction. Mitochondria constitute about 30 percent of the human heart. Mitochondrial DNA (mtDNA) is a damage-associated-molecular-pattern that induce injurious sterile inflammation. Little is known about mtDNA's inflammatory signalling pathways in cardiomyocytes and how mtDNA is internalized to associate with its putative receptor, toll-like receptor 9 (TLR9). EXPERIMENTAL APPROACH: We hypothesized that mtDNA can be internalized in cardiomyocytes and induce an inflammatory response. Adult mouse cardiomyocytes were exposed to hypoxia-reoxygenation and extracellular DNA. Microscale thermophoresis was used to demonstrate binding between nucleolin and DNA. KEY RESULTS: Expression of the pro-inflammatory cytokines IL-1ß and TNFα were upregulated by mtDNA, but not by nuclear DNA (nDNA), in cardiomyocytes exposed to hypoxia-reoxygenation. Blocking the RNA/DNA binding protein nucleolin with midkine reduced expression of IL-1ß/TNFα and the nucleolin inhibitor AS1411 reduced interleukin-6 release in adult mouse cardiomyocytes. mtDNA bound 10-fold stronger than nDNA to nucleolin. In HEK293-NF-κB reporter cells, mtDNA induced NF-κB activity in normoxia, while CpG-DNA and hypoxia-reoxygenation, synergistically induced TLR9-dependent NF-κB activity. Protein expression of nucleolin was found in the plasma membrane of cardiomyocytes and inhibition of nucleolin with midkine inhibited cellular uptake of CpG-DNA. Inhibition of endocytosis did not reduce CpG-DNA uptake in cardiomyocytes. CONCLUSION AND IMPLICATIONS: mtDNA, but not nDNA, induce an inflammatory response in mouse cardiomyocytes during hypoxia-reoxygenation. In cardiomyocytes, nucleolin is expressed on the membrane and blocking nucleolin reduce inflammation. Nucleolin might be a therapeutic target to prevent uptake of immunogenic DNA and reduce inflammation. LINKED ARTICLES: This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.


Asunto(s)
ADN/metabolismo , Hipoxia/metabolismo , Miocitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Islas de CpG , Fibroblastos/metabolismo , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Oxígeno/farmacología , Fosfoproteínas/genética , Proteínas de Unión al ARN/genética , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Nucleolina
17.
J Cardiovasc Transl Res ; 12(3): 184-192, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30542983

RESUMEN

During heart surgery with cardiopulmonary bypass (CPB), the release of mitochondrial (mtDNA) and nuclear DNA (nDNA) and their association to extracellular vesicles were investigated. In patients undergoing elective coronary artery bypass grafting (CABG, n = 12), blood was sampled before, during, and after surgery from peripheral artery, pulmonary artery, and the coronary sinus. Plasma was separated in three fractions: microvesicles, exosomes, and supernatant. mtDNA and nDNA were measured by qPCR. mtDNA and nDNA levels increased after start of surgery, but before CPB, and increased further during CPB. mtDNA copy number was about 1000-fold higher than nDNA. mtDNA was predominantly localized to the vesicular fractions in plasma, whereas nDNA was predominantly in the supernatant. The amount of free mtDNA increased after surgery. There was no net release or disappearance of DNAs across the pulmonary, systemic, or coronary circulation. Extracellular DNAs, in particular mtDNA, may be important contributors to the whole-body inflammation during CPB.


Asunto(s)
Puente Cardiopulmonar , Ácidos Nucleicos Libres de Células/sangre , Puente de Arteria Coronaria , ADN Mitocondrial/sangre , Exosomas/metabolismo , Ácidos Nucleicos Libres de Células/genética , ADN Mitocondrial/genética , Exosomas/genética , Humanos , Cinética
18.
Interact Cardiovasc Thorac Surg ; 28(5): 803-811, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30602018

RESUMEN

OBJECTIVES: Vascular wall calcification is a major pathophysiological component of atherosclerotic disease with many similarities to osteogenesis. Mechanical stress of the vascular wall may theoretically contribute to the proliferative processes by endothelial and interstitial cells. The aim of the study was to investigate the effect of mechanical stress on the expression of some calcification-related genes in primary human endothelial and interstitial cells, and how endothelial cells may stimulate the fibroblast and smooth muscle cells. METHODS: Human umbilical vein endothelial and interstitial cells were subjected to cyclic stretch using a FlexCell® bioreactor, and interstitial cells were also subjected to tensile strain in cultures embedded in 3-dimensional collagen gels. The medium from endothelial cells was used to stimulate the gel-cultured interstitial cells, or the endothelium was sown directly on top. For comparison, human endothelial and smooth muscle cells were isolated from aortic wall fragments of patients with and without the aortic aneurysm. The expression of genes was measured using quantitative PCR. RESULTS: Four hours of cyclic stretch applied to cultured endothelial cells upregulated the mRNA expression of bone morphogenetic protein 2 (BMP-2), a major procalcific growth factor. When applied to a 3-dimensional culture of vascular interstitial cells, the medium from prestretched endothelial cells decreased the expression of BMP-2 and periostin mRNA in the fibroblasts. The static tension in gel-cultured interstitial cells upregulated BMP-2 mRNA expression. The addition of endothelial cells on the top of this culture also reduced mRNA of anticalcific genes, periostin and osteopontin. Similar changes were observed in smooth muscle cells from human aortic aneurysms compared to cells from the healthy aorta. Aortic aneurysm endothelial cells also showed an increased expression of BMP-2 mRNA. CONCLUSIONS: Endothelial cells respond to mechanical stress by upregulation of pro-osteogenic factor BMP-2 mRNA and modulate the expression of other osteogenic factors in vascular interstitial cells. Endothelial cells may, thus, contribute to vascular calcification when exposed to mechanical stress.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Estrés Mecánico , Túnica Íntima/metabolismo , Calcificación Vascular/genética , Animales , Proteína Morfogenética Ósea 2/biosíntesis , Células Cultivadas , Células Endoteliales/patología , Endotelio Vascular/patología , Humanos , ARN Mensajero/genética , Túnica Íntima/patología , Regulación hacia Arriba , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
19.
Eur J Cardiothorac Surg ; 33(3): 355-63, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18221880

RESUMEN

OBJECTIVE: Corticosteroids and hyperoxia protect the heart against ischemia-reperfusion injury and may attenuate vascular reactivity. We hypothesized that (1) combining these two pretreatments induces an additive cardioprotection, (2) protection depends on activation of survival kinases and/or heat shock proteins, and (3) these interventions would change vascular reactivity into a more relaxed state. METHODS: Male rats were randomized (n=10 in each group): 1. control, 2. dexamethasone (3mg/kg) injected 24 and 12 h before harvesting the hearts, 3. 60 min of hyperoxia (90-95% O(2)) immediately before harvest, 4. combination of dexamethasone and hyperoxia as in groups 2 and 3. The hearts were Langendorff-perfused and exposed to 30 min of global ischemia and reperfused for 120 min. Cardiac function was monitored and infarct size determined. Isometric tension to vasoconstrictive and vasodilatory agents was measured in femoral artery rings. Phosphorylation of survival kinases (protein kinase B/AKT, extracellular signal-regulated kinases (ERK1/2), the stress-activated/c-Jun NH2 terminal kinases (SAPK/JNK) and p38 MAPK), adenosine monophosphate dependent kinase (AMPK) and expression of heat shock protein 72 (HSP72) in hearts was evaluated by immunoblotting. RESULTS: Infarct size was attenuated in all pretreated groups versus controls: 29% reduction in the combined group (p<0.01), 23% in hyperoxia group (p<0.05) and 31% in dexamethasone group (p<0.01). There was no significant difference between the treated groups. Combined pretreatment improved postischemic left ventricular end diastolic pressure compared to all other groups (p<0.001 vs controls, p=0.002 vs dexamethasone, p=0.005 vs hyperoxia). Combined pretreatment improved left ventricular developed pressure and coronary flow compared to controls (p<0.001 for both) and hyperoxia (p=0.0047 and p=0.0024, respectively). Combined pretreatment enhanced endothelium-independent relaxation (p=0.0032) compared to controls. Excepting ERK1/2 phosphorylation in the combined group during early reperfusion, there was no increased phosphorylation of the survival kinases AKT, p38, JNK, or AMPK and no increase of HSP72 expression. CONCLUSION: Combined pretreatment by hyperoxia and dexamethasone improved postischemic heart function, but did not reduce infarct size compared to single pretreatment groups. Except of a possible role of ERK1/2, protection depended neither on survival kinases nor heat shock protein 72.


Asunto(s)
Corticoesteroides/uso terapéutico , Dexametasona/uso terapéutico , Precondicionamiento Isquémico Miocárdico/métodos , Daño por Reperfusión Miocárdica/fisiopatología , Oxígeno/administración & dosificación , Cuidados Preoperatorios/métodos , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Presión Sanguínea/fisiología , Dexametasona/farmacología , Activación Enzimática , Proteínas del Choque Térmico HSP72/metabolismo , Immunoblotting , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Fosfotransferasas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Distribución Aleatoria , Ratas
20.
Scand Cardiovasc J ; 42(5): 327-36, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18609045

RESUMEN

OBJECTIVES: To investigate whether human veins responded to surgical handling with acute remodelling by measuring matrix metalloproteinase 9 (MMP9) expression and activity. DESIGN: Saphenous veins were collected from 24 patients (12 stable angina, 12 unstable angina) undergoing coronary artery bypass grafting. Expression of MMP9 and its regulators (plasminogen activators, plasminogen activator inhibitor-1, tissue inhibitor of metalloproteinase-1) was evaluated by semiquantitative RT-PCR in veins sampled at the start of and after surgical preparation, while protein was detected by western blotting. The proteolytic activity of MMP9 was analyzed by zymography. RESULTS: Gene (p=0.01) and protein (p=0.001) expression of MMP9 increased after surgical manipulation of vein grafts in all patients, accompanied by increased pro-MMP9 (p=0.04), but not active MMP9 (p=0.6). Grafts from stable patients had increased gene (p=0.05) and protein (p=0.006) expression, as well as increased pro- (p=0.04) and active (p=0.04) MMP9. Grafts from unstable patients increased only in MMP9 protein expression (p=0.05). The MMP9 regulators were unchanged. CONCLUSIONS: Surgical handling of vein grafts increased expression and activity of MMP9. However, the surgery-induced increase was attenuated in veins from unstable patients.


Asunto(s)
Angina de Pecho/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Vena Safena/metabolismo , Vena Safena/cirugía , Angina Inestable/metabolismo , Western Blotting , Puente de Arteria Coronaria , Femenino , Expresión Génica , Humanos , Masculino , Inhibidor 1 de Activador Plasminogénico/metabolismo , Activadores Plasminogénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA