Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Cell Biochem ; 110(1): 70-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20213742

RESUMEN

Accumulating evidences point to a significant role for the chromogranin A (CgA)-derived peptide vasostatin 1 (VS-1) in the protective modulation of the cardiovascular activity, because of its ability to counteract the adrenergic signal. We have recently shown that VS-1 induces a PI3K-dependent-nitric oxide (NO) release by endothelial cells, contributing to explain the mechanism of its cardio-suppressive and vasodilator properties. However, the cellular processes upstream the eNOS activation exerted by this peptide are still unknown, as typical high-affinity receptors have not been identified. Here we hypothesize that in endothelial cells VS-1 acts, on the basis of its cationic and amphipathic properties, as a cell penetrating peptide, binding to heparan sulfate proteoglycans (HSPGs) and activating eNOS phosphorylation (Ser1179) through a PI3K-dependent, endocytosis-coupled mechanism. In bovine aortic endothelial cells (BAE-1 cells) endocytotic vesicles trafficking was quantified by confocal microscopy with a water-soluble membrane dye; caveolin 1 (Cav1) shift from plasma membrane was studied by immunofluorescence staining; VS-1-dependent eNOS phosphorylation was assessed by immunofluorescence and immunoblot analysis. Our experiments demonstrate that VS-1 induces a marked increase in the caveolae-dependent endocytosis, (115 +/- 23% endocytotic spots/cell/field in VS-1-treated cells with respect to control cells), that is significantly reduced by both heparinase III (HEP, 17 +/- 15% above control) and Wortmannin (Wm, 7 +/- 22% above control). Heparinase, Wortmannin, and methyl-beta-cyclodextrin (MbetaCD) abolish the VS-1-dependent eNOS phosphorylation (P(Ser1179)eNOS). These results suggest a novel signal transduction pathway for endogenous cationic and amphipathic peptides in endothelial cells: HSPGs interaction and caveolae endocytosis, coupled with a PI3K-dependent eNOS phosphorylation.


Asunto(s)
Cromogranina A/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fragmentos de Péptidos/farmacología , Proteoglicanos/metabolismo , Androstadienos/farmacología , Animales , Bovinos , Caveolas/efectos de los fármacos , Caveolas/metabolismo , Caveolina 1/metabolismo , Endocitosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Liasa de Heparina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Vesículas Transportadoras/efectos de los fármacos , Vesículas Transportadoras/metabolismo , Wortmanina
2.
Basic Res Cardiol ; 105(5): 609-20, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20467749

RESUMEN

Thrombopoietin (TPO) is a humoral growth factor that has been shown to increase platelet activation in response to several agonists. Patients with sepsis have increased circulating TPO levels, which may enhance platelet activation, potentially participating to the pathogenesis of multi-organ failure. Aim of this study was to investigate whether TPO affects myocardial contractility and participates to depress cardiac function during sepsis. We showed the expression of the TPO receptor c-Mpl on myocardial cells and tissue by RT-PCR, immunofluorescence and western blotting. We then evaluated the effect of TPO on the contractile function of rat papillary muscle and isolated heart. TPO did not change myocardial contractility in basal conditions, but, when followed by epinephrine (EPI) stimulation, it blunted the enhancement of contractile force induced by EPI both in papillary muscle and isolated heart. An inhibitor of TPO prevented TPO effect on cardiac inotropy. Treatment of papillary muscle with pharmacological inhibitors of phosphatidylinositol 3-kinase, NO synthase, and guanilyl cyclase abolished TPO effect, indicating NO as the final mediator. We finally studied the role of TPO in the negative inotropic effect exerted by human septic shock (HSS) serum and TPO cooperation with TNF-alpha and IL-1beta. Pre-treatment with the TPO inhibitor prevented the decrease in contractile force induced by HSS serum. Moreover, TPO significantly amplified the negative inotropic effect induced by TNF-alpha and IL-1beta in papillary muscle. In conclusion, TPO negatively modulates cardiac inotropy in vitro and contributes to the myocardial depressing activity of septic shock serum.


Asunto(s)
Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Choque Séptico/fisiopatología , Trombopoyetina/metabolismo , Adolescente , Adulto , Animales , Proteínas Sanguíneas/farmacología , Línea Celular , Femenino , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Óxido Nítrico/metabolismo , Músculos Papilares/citología , Músculos Papilares/efectos de los fármacos , Músculos Papilares/fisiología , Ratas , Ratas Wistar , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Choque Séptico/sangre , Trombopoyetina/farmacología , Adulto Joven
3.
Cell Mol Biol Lett ; 14(1): 100-12, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18839068

RESUMEN

Ischemic diseases are characterized by the presence of pro-apoptotic stimuli, which initiate a cascade of processes that lead to cell injury and death. Several molecules and events represent detectable indicators of the different stages of apoptosis. Among these indicators is phosphatidylserine (PS) translocation from the inner to the outer leaflet of the plasma membrane, which can be detected by annexinV (ANXA5) conjugation. This is a widely used in vivo and in vitro assay marking the early stages of apoptosis. We report here on an original method that employs PS-ANXA5 conjugation to target stem cells to apoptotic cells. Mesenchymal stem cells (MSCs) from GFP-positive transgenic rats were biotinylated on membrane surfaces with sulfosuccinimidyl-6-(biotinamido) hexanoate (sulfo-NHS-LC-biot) and then bound to avidin. The avidin-biotinylated MSCs were labeled with biotin conjugated ANXA5. Bovine aortic endothelial cells (BAE-1 cells) were exposed to UVC to induce caspasedependent apoptosis. Finally, we tested the ability of ANXA5-labeled MSCs to bind BAE-1 apoptotic cells: suspended ANXA5-labeled MSCs were seeded for 1 hour on a monolayer of UV-treated or control BAE-1 cells. After washing, the number of MSCs bound to BAE-1 cells was evaluated by confocal microscopy. Statistical analysis demonstrated a significant increase in the number of MSCs tagged to apoptotic BAE-1 cells. Therefore, stem cell ANXA5 tagging via biotin-avidin bridges could be a straightforward method of improving homing to apoptotic tissues.


Asunto(s)
Anexina A5/metabolismo , Apoptosis , Movimiento Celular , Células Endoteliales/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Coloración y Etiquetado , Animales , Biotinilación , Bovinos , Línea Celular , Células Endoteliales/metabolismo , Cetonas/metabolismo , Ratas
4.
Biomed Res Int ; 2014: 783623, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25136621

RESUMEN

The chromogranin-A peptide catestatin modulates a wide range of processes, such as cardiovascular functions, innate immunity, inflammation, and metabolism. We recently found that the cardiac antiadrenergic action of catestatin requires a PI3K-dependent NO release from endothelial cells, although the receptor involved is yet to be identified. In the present work, based on the cationic properties of catestatin, we tested the hypothesis of its interaction with membrane heparan sulphate proteoglycans, resulting in the activation of a caveolae-dependent endocytosis. Experiments were performed on bovine aortic endothelial cells. Endocytotic vesicles trafficking was quantified by confocal microscopy using a water-soluble membrane dye; catestatin colocalization with heparan sulphate proteoglycans and caveolin 1 internalization were studied by fluorimetric measurements in live cells. Modulation of the catestatin-dependent eNOS activation was assessed by immunofluorescence and immunoblot analysis. Our results demonstrate that catestatin (5 nM) colocalizes with heparan sulphate proteoglycans and induces a remarkable increase in the caveolae-dependent endocytosis and caveolin 1 internalization, which were significantly reduced by both heparinase and wortmannin. Moreover, catestatin was unable to induce Ser(1179) eNOS phosphorylation after pretreatments with heparinase and methyl-ß-cyclodextrin. Taken together, these results highlight the obligatory role for proteoglycans and caveolae internalization in the catestatin-dependent eNOS activation in endothelial cells.


Asunto(s)
Cromogranina A/administración & dosificación , Endocitosis/efectos de los fármacos , Proteoglicanos de Heparán Sulfato/metabolismo , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/administración & dosificación , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Bovinos , Caveolas/efectos de los fármacos , Caveolas/ultraestructura , Caveolina 1/metabolismo , Cromogranina A/metabolismo , Cromogranina A/ultraestructura , Células Endoteliales/efectos de los fármacos , Células Endoteliales/ultraestructura , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/ultraestructura , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/ultraestructura
5.
Regul Pept ; 167(1): 5-8, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21237210

RESUMEN

Thrombopoietin (TPO) is known for its ability to stimulate platelet production. However, little is currently known whether TPO plays a physiological function in the heart. The potential vasodilatory role of TPO was tested on the isolated rat heart. The expression of TPO receptor (c-mpl) and the TPO-dependent eNOS phosphorylation (P(Ser1179)) were studied on Cardiac-derived normal Human Micro Vascular Endothelial Cells (HMVEC-C) by Western blot analysis. While TPO (10-200 pg/mL) did not modify coronary flow (CF) under basal conditions, it reduced the coronary constriction caused by endothelin-1 (ET-1; 10nM) in a dose-dependent manner. This effect was blocked by both Wortmannin (100 nM) and L-NAME (100 nM); on HMVEC-C, TPO induced eNOS phosphorylation through a Wortmannin sensitive mechanism. Taken together, our data suggest a potential role of TPO as a physiological regulator of CF. By acting on specific receptors present on endothelial cells, TPO may induce PI3K/Akt-dependent eNOS phosphorylation and NO release.


Asunto(s)
Circulación Coronaria/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores de Trombopoyetina/metabolismo , Trombopoyetina/metabolismo , Androstadienos/farmacología , Animales , Western Blotting , Células Cultivadas , Células Endoteliales/metabolismo , Endotelina-1/farmacología , Expresión Génica , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptores de Trombopoyetina/genética , Trombopoyetina/genética , Trombopoyetina/farmacología , Vasoconstricción/efectos de los fármacos , Wortmanina
6.
Cardiovasc Res ; 91(4): 617-24, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21543385

RESUMEN

AIMS: Catestatin (CST) is a chromogranin A (CgA)-derived peptide (hCgA352-372) with three identified human variants (G364S/P370L/R374Q-CST) that show differential potencies towards the inhibition of catecholamine release. Although CST affects several cardiovascular parameters, the mechanisms underlying CST action in the heart have remained elusive. Therefore, we sought to determine the mechanism of action of CST and its variants on ventricular myocardium and endothelial cells. METHODS AND RESULTS: Contractile force and Ca(2+) transients were measured, respectively, on rat papillary muscles and isolated cardiomyocytes (CC) under basal conditions and after ß-adrenergic stimulation. Nitric oxide (NO) production and endothelial nitric oxide synthase (eNOS) phosphorylation (P(Ser1179)eNOS) were studied in bovine aortic endothelial (BAE-1) cells. Under basal conditions, wild-type CST (WT-CST, 10-50 nM) transiently enhanced myocardial contractility. CST variants (G364S and P370L) exerted a comparable positive inotropic effect. The H(1) histamine receptor antagonist mepyramine abolished the increase of contractile force induced by WT-CST. Moreover, WT-CST dose-dependently (5-50 nM) reduced the effect of ß-adrenergic stimulation. This anti-adrenergic effect was not mediated by a direct action on CC, but involved a PI3K-dependent NO release from endocardial endothelial cells. Indeed, CST induced a wortmannin-sensitive, Ca(2+)-independent increase in NO production and eNOS phosphorylation on BAE-1 cells. While the anti-adrenergic and NO release effects of P370L-CST were comparable with those of WT-CST, the G364S variant was ineffective on the same parameters. CONCLUSION: Our results suggest that the anti-adrenergic action of CST depends on the endothelial PI3K-Akt-eNOS pathway and that its structural alterations entail functional features that correlate with the different anti-hypertensive potential described in humans.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Cromogranina A/farmacología , Corazón/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/fisiología , Fragmentos de Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Calcio/metabolismo , Bovinos , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Miocardio/metabolismo , Óxido Nítrico/biosíntesis , Músculos Papilares/efectos de los fármacos , Ratas
7.
Cardiovasc Res ; 88(3): 443-52, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20634213

RESUMEN

AIMS: The ErbB-neuregulin-1ß1 (Nrg1ß1) pathway is required for cardiac development and exerts chronic effects on the postnatal adult heart. Long-term application of Nrg1ß1 results in hypertrophy and protection against oxidative stress and cytotoxic agents. We performed experiments with acute Nrg1ß1 treatment to find evidence for a further protective role due to rapid modulation of adult cardiomyocyte function. METHODS AND RESULTS: In confocal fluorimetric measurements, Nrg1ß1 induced a calcium-independent increase in nitric oxide (NO) production in isolated adult rat ventricular myocytes (ARVCMs) that was blocked by the phosphoinositide-3-kinase (PI3K) inhibitor Wortmannin. Western blot analysis showed enhancement of endothelial nitric oxide synthase phosphorylation in Nrg1ß1-treated ARVCMs, which was attenuated by Wortmannin. Nrg1ß1 induced a significant increase in calcium transient amplitude (indo-1 ratiometric measurement) and accelerated the recovery of cytosolic calcium in the sarcoplasmic reticulum without affecting whole-cell L-type calcium current. Wortmannin or the protein kinase G inhibiting peptide (DT-2) abolished the increase in calcium transient amplitude and the acceleration of calcium recovery induced by Nrg1ß1 treatment. Immunofluorescence analysis revealed that Nrg1ß1 treatment increased phospholamban phosphorylation, and the effect was blocked by PI3K and protein kinase G inhibition. Caffeine-releasable sarcoplasmic reticulum calcium content was also higher during Nrg1ß1 administration. CONCLUSION: Rapid activation of PI3K, endothelial nitric oxide synthase and protein kinase G and a consequent improvement in diastolic calcium can be added to established Nrg1 protective roles.


Asunto(s)
Calcio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Neurregulina-1/farmacología , Óxido Nítrico/metabolismo , Androstadienos/farmacología , Animales , Proteínas de Unión al Calcio/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Femenino , Fluoresceínas/farmacología , Modelos Animales , Miocitos Cardíacos/citología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fragmentos de Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Endogámicas , Retículo Sarcoplasmático/metabolismo , Wortmanina
8.
Cardiovasc Res ; 83(2): 303-12, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19293247

RESUMEN

AIMS: The hypothalamic neuropeptide growth hormone-releasing hormone (GHRH) stimulates GH synthesis and release in the pituitary. GHRH also exerts proliferative effects in extrapituitary cells, whereas GHRH antagonists have been shown to suppress cancer cell proliferation. We investigated GHRH effects on cardiac myocyte cell survival and the underlying signalling mechanisms. METHODS AND RESULTS: Reverse transcriptase-polymerase chain reaction analysis showed GHRH receptor (GHRH-R) mRNA in adult rat ventricular myocytes (ARVMs) and in rat heart H9c2 cells. In ARVMs, GHRH prevented cell death and caspase-3 activation induced by serum starvation and by the beta-adrenergic receptor agonist isoproterenol. The GHRH-R antagonist JV-1-36 abolished GHRH survival action under both experimental conditions. GHRH-induced cardiac cell protection required extracellular signal-regulated kinase (ERK)1/2 and phosphoinositide-3 kinase (PI3K)/Akt activation and adenylyl cyclase/cAMP/protein kinase A signalling. Isoproterenol strongly upregulated the mRNA and protein of the pro-apoptotic inducible cAMP early repressor, whereas GHRH completely blocked this effect. Similar to ARVMs, in H9c2 cardiac cells, GHRH inhibited serum starvation- and isoproterenol-induced cell death and apoptosis through the same signalling pathways. Finally, GHRH improved left ventricular recovery during reperfusion and reduced infarct size in Langendorff-perfused rat hearts, subjected to ischaemia-reperfusion (I/R) injury. These effects involved PI3K/Akt signalling and were inhibited by JV-1-36. CONCLUSION: Our findings suggest that GHRH promotes cardiac myocyte survival through multiple signalling mechanisms and protects against I/R injury in isolated rat heart, indicating a novel cardioprotective role of this hormone.


Asunto(s)
Hormona Liberadora de Hormona del Crecimiento/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Transducción de Señal , Adenilil Ciclasas/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citoprotección , Hormona Liberadora de Hormona del Crecimiento/análogos & derivados , Hormona Liberadora de Hormona del Crecimiento/farmacología , Isoproterenol/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Contracción Miocárdica/efectos de los fármacos , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Perfusión , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/metabolismo , Ratas , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/antagonistas & inhibidores , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Recuperación de la Función , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos
9.
Am J Physiol Heart Circ Physiol ; 292(6): H2906-12, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17293489

RESUMEN

Vasostatins (VSs) are vasoactive peptides derived from chromogranin A (CgA), a protein contained in secretory granules of chromaffin and other cells. The negative inotropic effect and the reduction of isoproterenol (Iso)-dependent inotropism induced by VSs in the heart suggest that they have an antiadrenergic function. However, further investigation of the mechanisms of action of VSs is needed. The aim of the present study was to define the signaling pathways activated by VS-1 in mammalian ventricular myocardium and cultured endothelial cells that lead to the modulation of cardiac contractility. Ca(2+) and nitric oxide (NO) fluorometric confocal imaging was used to study the effects induced by recombinant human VS-1 [STA-CgA-(1-76)] on contractile force, L-type Ca(2+) current, and Ca(2+) transients under basal conditions and after beta-adrenergic stimulation in rat papillary muscles and ventricular cells and the effects on intracellular Ca(2+) concentration and NO production in cultured bovine aortic endothelial (BAE-1) cells. VS-1 had no effect on basal contractility of papillary muscle, but the effect of Iso stimulation was reduced by 27%. Removal of endocardial endothelium and inhibition of NO synthesis and phosphatidylinositol 3-kinase (PI3K) activity abolished the antiadrenergic effect of VS-1 on papillary muscle. In cardiomyocytes, 10 nM VS-1 was ineffective on basal and Iso (1 microM)-stimulated L-type Ca(2+) current and Ca(2+) transients. In BAE-1 cells, VS-1 induced a Ca(2+)-independent increase in NO production that was blocked by the PI3K inhibitor wortmannin. Our results suggest that the antiadrenergic effect of VS-1 is mainly due to a PI3K-dependent NO release by endothelial cells, rather than a direct action on cardiomyocytes.


Asunto(s)
Antagonistas Adrenérgicos beta/metabolismo , Señalización del Calcio , Cromogranina A/metabolismo , Células Endoteliales/metabolismo , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Músculos Papilares/metabolismo , Fragmentos de Péptidos/metabolismo , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Androstadienos/farmacología , Animales , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio/efectos de los fármacos , Bovinos , Células Cultivadas , Cromogranina A/farmacología , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Femenino , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Humanos , Técnicas In Vitro , Isoproterenol/antagonistas & inhibidores , Isoproterenol/farmacología , Fuerza Muscular , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Músculos Papilares/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Proteínas Recombinantes/metabolismo , Wortmanina
10.
J Cell Biochem ; 100(1): 86-99, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16888800

RESUMEN

In order to assess, in a controlled in vitro model, the differentiation potential of adult bone marrow derived stem cells we have developed a coculture procedure using adult rat cardiomyocytes and mesenchymal stem cells (MSCs) from transgenic GFP positive rats. We investigated in the cocultured MSCs the time course of cellular processes that are difficult to monitor in in vivo experiments. Adult rat cardiomyocytes and adult rat MSCs were cocultured for up to 7 days and analyzed by confocal microscopy. Several markers were studied by immunofluorescence technique. The fluorescent ST-BODIPY-Dihydropyridine was used to label calcium channels in living cells. Intracellular calcium was monitored with the fluorescent probe X-Rhod-1. Immunofluorescence experiments showed the presence of connexin-43 between cardiomyocytes and MSCs and between MSCs, while no sarcomeric structures were observed at any time of the coculture. We looked at the expression of calcium channels and development of voltage-dependent calcium signaling in cocultured MSCs. MSCs showed a time-dependent increase of labeling of ST-BODIPY-Dihydropyridine, reaching a relatively strong level after 72 h of coculture. The treatment with a non-fluorescent DHP, Nifedipine, completely abolished ST-BODIPY labeling. We investigated whether depolarization could modulate intracellular calcium. Depolarization-induced calcium transients increased in MSCs in relation to the coculture time. We conclude that MSCs cocultured with adult cardiomyocytes present preliminary evidence of voltage-dependent calcium modulation uncoupled with the development of nascent or adult myofibrils, thus showing a limited lineage specification and a low plasticity to differentiate in a full cardiomyocyte-like phenotype.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Miocitos Cardíacos/fisiología , Actinina/metabolismo , Animales , Animales Modificados Genéticamente , Compuestos de Boro , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/fisiología , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Conexina 43/metabolismo , Dihidropiridinas , Colorantes Fluorescentes , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Activación del Canal Iónico , Células Madre Mesenquimatosas/citología , Miocitos Cardíacos/citología , Miosinas/metabolismo , Nifedipino/farmacología , Ratas , Sarcómeros/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA