Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Mol Cell Biochem ; 476(8): 3111-3126, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33837873

RESUMEN

The widespread environmental pollutant 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153) is a non-dioxin-like toxicant. It is a potential carcinogen compound able to induce gap junction (GJ) intercellular communication impairment, probably the first non-genomic event leading to tumor promotion. Although PCBs have been known for many years, the molecular mode of PCB153 action is still unclear. Recent studies from our research group have shown that the toxicant elicits a transient modulation of connexin (Cx) 43-formed GJs in hepatic stem-like WB-F344 cells involving sphingosine 1-phosphate (S1P) path. Taking into account that other strictly related bioactive sphingolipids, such as ceramide (Cer), may have different effects from S1P, here we aim to clarify the signaling paths engaged by PCB153 in the control of GJs, focusing primarily on the role of Cer. Accordingly, we have achieved a combined biomolecular and electrophysiological analysis of GJs in cultured WB-F344 cells treated with PCB153 at different time points. We have found that the toxicant elicited a time-dependent regulation of GJs formed by different Cx isoforms, through a transient modulation of Cer/Cer kinase (CerK) axis and, in turn, of protein phosphatase 2A (PP2A). Our new findings demonstrate the existence of a specific molecular mechanism downstream to Cer, which distinctly affects the voltage-dependent and -independent GJs in liver stem-like cells, and open new opportunities for the identification of additional potential targets of these environmental toxicants.


Asunto(s)
Ceramidas/metabolismo , Uniones Comunicantes/patología , Hígado/patología , Bifenilos Policlorados/farmacología , Proteína Fosfatasa 2/metabolismo , Células Madre/patología , Animales , Comunicación Celular , Células Cultivadas , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteína Fosfatasa 2/genética , Ratas , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/metabolismo
2.
Mol Cell Neurosci ; 75: 50-62, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27370937

RESUMEN

Over the past decades, studies in both Huntington's disease animal models and pilot clinical trials have demonstrated that replacement of degenerated striatum and repair of circuitries by grafting fetal striatal primordium is feasible, safe and may counteract disease progression. However, a better comprehension of striatal ontogenesis is required to assess the fetal graft regenerative potential. During neuronal development, neurotrophins exert pleiotropic actions in regulating cell fate and synaptic plasticity. In this regard, brain-derived neurotrophic factor (BDNF) and fibroblast growth factor 2 (FGF2) are crucially implicated in the control of fate choice of striatal progenitor cells. In this study, we intended to refine the functional features of human striatal precursor (HSP) cells isolated from ganglionic eminence of 9-12week old human fetuses, by studying with electrophysiological methods the effect of BDNF and FGF2 on the membrane biophysical properties and the voltage-dependent Ca(2+) currents. These features are particularly relevant to evaluate neuronal cell functioning and can be considered reliable markers of the developmental phenotype of human striatal primordium. Our results have demonstrated that BDNF and FGF2 induced membrane hyperpolarization, increased the membrane capacitance and reduced the resting total and specific conductance values, suggesting a more efficient control of resting ionic fluxes. Moreover, the treatment with both neurotrophins enhanced N-type Ca(2+) current amplitude and reduced L- and T-type ones. Overall, our data indicate that BDNF and FGF2 may help HSP cells to attain a more functionally mature phenotype.


Asunto(s)
Potenciales de Acción , Factor Neurotrófico Derivado del Encéfalo/farmacología , Canales de Calcio/metabolismo , Cuerpo Estriado/fisiología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células-Madre Neurales/fisiología , Neurogénesis , Células Cultivadas , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Humanos , Células-Madre Neurales/efectos de los fármacos
3.
Exp Physiol ; 101(8): 1086-100, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27307205

RESUMEN

What is the central question of this study? Hyponatraemia, an electrolyte disorder encountered in hospitalized patients, can cause neurological symptoms usually attributed to a reduction in plasma osmolarity. Here, we investigated whether low [Na(+) ] per se can cause neuronal changes independent of osmolarity, focusing on involvement of the Na(+) -Ca(2+) exchanger. What is the main finding and its importance? We show that hyponatraemia per se causes alterations of neuronal properties. The novel finding of Na(+) -Ca(2+) exchanger involvement helps us to elucidate the volume regulation following hyponatraemia. This might have relevance in a translational perspective because Na(+) -Ca(2+) exchanger could be a target for novel therapies. Hyponatraemia is the most frequent electrolyte disorder encountered in hospitalized patients, and it can cause a wide variety of neurological symptoms. Most of the negative effects of this condition on neuronal cells are attributed to cell swelling because of the reduction of plasma osmolarity, although in hyponatraemia different membrane proteins are supposed to be involved in the conservation of neuronal volume. We have recently reported detrimental effects of hyponatraemia on two different neuronal cell lines, SK-N-AS and SH-SY5Y, independent of osmotic alterations. In this study we investigated, in the same cell lines, whether hyponatraemic conditions per se can cause electrophysiological alterations and whether these effects vary over time. Accordingly, we carried out experiments in low-sodium medium in either hyposmotic [Osm(-)] or isosmotic [Osm(+)] conditions, for a short (24 h) or long time (7 days). Using a patch pipette in voltage-clamp conditions, we recorded possible modifications of cell capacitance (Cm ) and membrane conductance (Gm ). Our results indicate that in both Osm(-) and Osm(+) medium, Cm and Gm show a similar increase, but such effects are dependent on the time in culture in different ways. Notably, regarding the possible mechanisms involved in the maintenance of Cm , Gm and Gm /Cm in Osm(+) conditions, we observed a greater contribution of the Na(+) -Ca(2+) exchanger compared with Osm(-) and control conditions. Overall, these novel electrophysiological results help us to understand the mechanisms of volume regulation after ionic perturbation. Our results might also have relevance in a translational perspective because the Na(+) -Ca(2+) exchanger can be considered a target for planning novel therapies.


Asunto(s)
Membrana Celular/fisiología , Hiponatremia/fisiopatología , Neuronas/fisiología , Calcio/metabolismo , Recuento de Células/métodos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Humanos , Hiponatremia/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Níquel/farmacología , Concentración Osmolar , Técnicas de Placa-Clamp/métodos , Sodio/metabolismo , Intercambiador de Sodio-Calcio/metabolismo
4.
Exp Physiol ; 100(6): 652-66, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25786395

RESUMEN

NEW FINDINGS: What is the central question of this study? Fibroblast-to-myofibroblast transition is a key mechanism in the reparative response to tissue damage, but myofibroblast persistence in the wound leads to fibrosis and organ failure. The role of relaxin as an antifibrotic agent capable of counteracting the acquisition of biophysical features of differentiated myofibroblasts deserves further investigation. What is the main finding and its importance? Electrophysiological analysis showed that relaxin, administered during profibrotic treatment, hyperpolarizes the membrane potential and attenuates delayed rectifier and inwardly rectifying K(+) currents, which usually increase in the transition to myofibroblasts. These findings provide further clues to the therapeutic potential of relaxin in fibrosis. The hormone relaxin (RLX) is produced by the heart and may be involved in endogenous mechanisms of cardiac protection against ischaemic injury and fibrosis. Recent findings in cultured cardiac stromal cells suggest that RLX can inhibit fibroblast-to-myofibroblast transition, thereby counteracting fibrosis. In order to explore its efficiency as an antifibrotic agent further, we designed the present study to investigate whether RLX may influence the electrophysiological events associated with differentiation of cardiac stromal cells to myofibroblasts. Primary cardiac proto-myofibroblasts and NIH/3T3 fibroblasts were induced to myofibroblasts by transforming growth factor-ß1, and the electrophysiological features of both cell populations were investigated by whole-cell patch clamp. We demonstrated that proto-myofibroblasts and myofibroblasts express different membrane passive properties and K(+) currents. Here, we have shown, for the first time, that RLX (100 ng ml(-1) ) significantly reduced both voltage- and Ca(2+) -dependent delayed-rectifier and inward-rectifying K(+) currents that are typically increased in myofibroblasts compared with proto-myofibroblasts, suggesting that this hormone can antagonize the biophysical effects of transforming growth factor-ß1 in inducing myofibroblast differentiation. These newly recognized effects of RLX on the electrical properties of cardiac stromal cell membrane correlate well with its well-known ability to suppress myofibroblast differentiation, further supporting the possibility that RLX may be used for the treatment of cardiac fibrosis.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Canales de Potasio de Rectificación Interna/metabolismo , Relaxina/farmacología , Animales , Biomarcadores/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Humanos , Potenciales de la Membrana , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/patología , Células 3T3 NIH , Fenotipo , Potasio/metabolismo , Proteínas Recombinantes/farmacología , Factor de Crecimiento Transformador beta1/farmacología
5.
J Cell Physiol ; 228(1): 172-81, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22628164

RESUMEN

Mesenchymal stromal cells (MSCs) are a promising cell candidate in tissue engineering and regenerative medicine. Their proliferative potential can be increased by low-level laser irradiation (LLLI), but the mechanisms involved remain to be clarified. With the aim of expanding the therapeutic application of LLLI to MSC therapy, in the present study we investigated the effects of 635 nm diode laser on mouse MSC proliferation and investigated the underlying cellular and molecular mechanisms, focusing the attention on the effects of laser irradiation on Notch-1 signal activation and membrane ion channel modulation. It was found that MSC proliferation was significantly enhanced after laser irradiation, as judged by time lapse videomicroscopy and EdU incorporation. This phenomenon was associated with the up-regulation and activation of Notch-1 pathway, and with increased membrane conductance through voltage-gated K(+) , BK and Kir, channels and T- and L-type Ca(2+) channels. We also showed that MSC proliferation was mainly dependent on Kir channel activity, on the basis that the cell growth and Notch-1 up-regulation were severely decreased by the pre-treatment with the channel inhibitor Ba(2+) (0.5 mM). Interestingly, the channel inhibition was also able to attenuate the stimulatory effects of diode laser on MSCs, thus providing novel evidence to expand our knowledge on the mechanisms of biostimulation after LLLI. In conclusions, our findings suggest that diode laser may be a valid approach for the preconditioning of MSCs in vitro prior cell transplantation.


Asunto(s)
Células de la Médula Ósea/efectos de la radiación , Láseres de Semiconductores , Células Madre Mesenquimatosas/efectos de la radiación , Animales , Células de la Médula Ósea/fisiología , Proliferación Celular/efectos de la radiación , Supervivencia Celular , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Fenómenos Electrofisiológicos , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/fisiología , Ratones , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje , Receptor Notch1/genética , Receptor Notch1/metabolismo , Coloración y Etiquetado
6.
Am J Physiol Gastrointest Liver Physiol ; 305(9): G628-37, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23989009

RESUMEN

Obestatin is a hormone released from the stomach deriving from the same peptide precursor as ghrelin. It is known to act as an anorectic hormone decreasing food intake, but contrasting results have been reported about the effects of obestatin on gastrointestinal motility. The aim of the present study was to investigate whether this peptide may act on the gastric longitudinal smooth muscle by using a combined mechanical and electrophysiological approach. When fundal strips from mice were mounted in organ baths for isometric recording of the mechanical activity, obestatin caused a tetrodotoxin-insensitive decrease of the basal tension and a reduction in amplitude of the neurally induced cholinergic contractile responses, even in the presence of the nitric oxide synthesis inhibitor N(G)-nitro-l-arginine. Obestatin reduced the amplitude of the response to the ganglionic stimulating agent dimethylphenyl piperazinium iodide but did not influence that to methacholine. In nonadrenergic, noncholinergic conditions, obestatin still decreased the basal tension of the preparations without influencing the neurally induced relaxant responses. For comparison, in circular fundal strips, obestatin had no effects. Notably, in the longitudinal antral ones, obestatin only caused a decrease of the basal tension. Electrophysiological experiments, performed by a single microelectrode inserted in a gastric longitudinal smooth muscle cell, showed that obestatin had similar effects in fundal and antral preparations: it decreased the resting specific membrane conductance, inhibited Ca(2+) currents, and positively shifted their voltage threshold of activation. In conclusion, the present results indicate that obestatin influences gastric smooth muscle exerting site-specific effects.


Asunto(s)
Fenómenos Electrofisiológicos , Ghrelina/farmacología , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Estómago/efectos de los fármacos , Animales , Fundus Gástrico/efectos de los fármacos , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Contracción Muscular/fisiología , Músculo Liso/fisiología , Estómago/fisiología , Tetrodotoxina/farmacología
7.
J Cell Mol Med ; 16(3): 507-19, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21554533

RESUMEN

The demonstration that the adult heart contains myocardial progenitor cells which can be recruited in an attempt to replace the injured myocardium has sparkled interest towards novel molecules capable of improving the differentiation of these cells. In this context, the peptide hormone relaxin (RLX), recently validated as a cardiovascular hormone, is a promising candidate. This study was designed to test the hypothesis that RLX may promote the growth and maturation of mouse neonatal immature cardiomyocytes in primary culture. The cultures were studied at 2, 12, 24 and 48 hrs after the addition of human recombinant H2 RLX (100 ng/ml), the main circulating form of the hormone, or plain medium by combining molecular biology, morphology and electrophysiology. RLX modulated cell proliferation, promoting it at 2 and 12 hrs and inhibiting it at 24 hrs; RLX also induced the expression of both cardiac-specific transcription factors (GATA-4 and Nkx2-5) and cardiac-specific structural genes (connexin 43, troponin T and HCN4 ion channel) at both the mRNA and protein level. Consistently, RLX induced the appearance of ultrastructural and electrophysiological signs of functionally competent, mature cardiomyocytes. In conclusion, this study provides novel circumstantial evidence that RLX specifically acts on immature cardiomyocytes by promoting their proliferation and maturation. This notion suggests that RLX, for which the heart is both a source and target organ, may be an endogenous regulator of cardiac morphogenesis during pre-natal life and could participate in heart regeneration and repair, both as endogenous myocardium-derived factor and exogenous cardiotropic drug, during adult life.


Asunto(s)
Corazón/fisiología , Miocitos Cardíacos/citología , ARN Mensajero/biosíntesis , Regeneración , Relaxina/farmacología , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Conexina 43/genética , Conexina 43/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Expresión Génica/efectos de los fármacos , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Troponina T/genética , Troponina T/metabolismo
8.
J Cell Mol Med ; 15(11): 2498-511, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21199328

RESUMEN

Skeletal muscle regeneration is severely compromised in the case of extended damage. The current challenge is to find factors capable of limiting muscle degeneration and/or potentiating the inherent regenerative program mediated by a specific type of myoblastic cells, the satellite cells. Recent studies from our groups and others have shown that the bioactive lipid, sphingosine 1-phosphate (S1P), promotes myoblast differentiation and exerts a trophic action on denervated skeletal muscle fibres. In the present study, we examined the effects of S1P on eccentric contraction (EC)-injured extensor digitorum longus muscle fibres and resident satellite cells. After EC, skeletal muscle showed evidence of structural and biochemical damage along with significant electrophysiological changes, i.e. reduced plasma membrane resistance and resting membrane potential and altered Na(+) and Ca(2+) current amplitude and kinetics. Treatment with exogenous S1P attenuated the EC-induced tissue damage, protecting skeletal muscle fibre from apoptosis, preserving satellite cell viability and affecting extracellular matrix remodelling, through the up-regulation of matrix metalloproteinase 9 (MMP-9) expression. S1P also promoted satellite cell renewal and differentiation in the damaged muscle. Notably, EC was associated with the activation of sphingosine kinase 1 (SphK1) and with increased endogenous S1P synthesis, further stressing the relevance of S1P in skeletal muscle protection and repair/regeneration. In line with this, the treatment with a selective SphK1 inhibitor during EC, caused an exacerbation of the muscle damage and attenuated MMP-9 expression. Together, these findings are in favour for a role of S1P in skeletal muscle healing and offer new clues for the identification of novel therapeutic approaches to counteract skeletal muscle damage and disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Músculo Esquelético/fisiología , Regeneración , Células Satélite del Músculo Esquelético/fisiología , Esfingosina/análogos & derivados , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Calcio/análisis , Caspasa 3 , Caspasa 7 , Diferenciación Celular/efectos de los fármacos , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/biosíntesis , Potenciales de la Membrana/efectos de los fármacos , Ratones , Contracción Muscular , Fibras Musculares Esqueléticas , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Células Satélite del Músculo Esquelético/efectos de los fármacos , Células Satélite del Músculo Esquelético/metabolismo , Transducción de Señal , Sodio/análisis , Esfingosina/metabolismo , Esfingosina/farmacología , Cicatrización de Heridas
9.
J Physiol ; 589(Pt 21): 5231-46, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21911618

RESUMEN

Orexin A (OXA) has been reported to influence gastrointestinal motility, acting at both central and peripheral neural levels. The aim of the present study was to evaluate whether OXA also exerts direct effects on the duodenal smooth muscle. The possible mechanism of action involved was investigated by employing a combined mechanical and electrophysiological approach. Duodenal segments were mounted in organ baths for isometric recording of the mechanical activity. Ionic channel activity was recorded in current- and voltage-clamp conditions by a single microelectrode inserted in a duodenal longitudinal muscle cell. In the duodenal preparations, OXA (0.3 µM) caused a TTX-insensitive transient contraction. Nifedipine (1 µM), as well as 2-aminoethyl diphenyl borate (10 µM), reduced the amplitude and shortened the duration of the response to OXA, which was abolished by Ni(2+) (50 µM) or TEA (1 mM). Electrophysiological studies in current-clamp conditions showed that OXA caused an early depolarization, which paralleled in time the contractile response, followed by a long-lasting depolarization. Such a depolarization was triggered by activation of receptor-operated Ca(2+) channels and enhanced by activation of T- and L-type Ca(2+) channels and store-operated Ca(2+) channels and by inhibition of K(+) channels. Experiments in voltage-clamp conditions demonstrated that OXA affects not only receptor-operated Ca(2+) channels, but also the maximal conductance and kinetics of activation and inactivation of Na(+), T- and L-type Ca(2+) voltage-gated channels. The results demonstrate, for the first time, that OXA exerts direct excitatory effects on the mouse duodenal smooth muscle. Finally, this work demonstrates new findings related to the expression and kinetics of the voltage-gated channel types, as well as store-operated Ca(2+) channels.


Asunto(s)
Duodeno/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Músculo Liso/efectos de los fármacos , Neuropéptidos/farmacología , Animales , Compuestos de Boro/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Duodeno/fisiología , Femenino , Técnicas In Vitro , Contracción Isométrica/efectos de los fármacos , Ratones , Músculo Liso/fisiología , Nifedipino/farmacología , Orexinas , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
10.
Cell Mol Life Sci ; 67(24): 4269-85, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20614160

RESUMEN

We recently demonstrated that skeletal muscle differentiation induced by sphingosine 1-phosphate (S1P) requires gap junctions and transient receptor potential canonical 1 (TRPC1) channels. Here, we searched for the signaling pathway linking the channel activity with Cx43 expression/function, investigating the involvement of the Ca(2+)-sensitive protease, m-calpain, and its targets in S1P-induced C2C12 myoblast differentiation. Gene silencing and pharmacological inhibition of TRPC1 significantly reduced Cx43 up-regulation and Cx43/cytoskeletal interaction elicited by S1P. TRPC1-dependent functions were also required for the transient increase of m-calpain activity/expression and the subsequent decrease of PKCα levels. Remarkably, Cx43 expression in S1P-treated myoblasts was reduced by m-calpain-siRNA and enhanced by pharmacological inhibition of classical PKCs, stressing the relevance for calpain/PKCα axis in Cx43 protein remodeling. The contribution of this pathway in myogenesis was also investigated. In conclusion, these findings provide novel mechanisms by which S1P regulates myoblast differentiation and offer interesting therapeutic options to improve skeletal muscle regeneration.


Asunto(s)
Conexina 43/metabolismo , Lisofosfolípidos/metabolismo , Desarrollo de Músculos/fisiología , Músculo Esquelético/embriología , Músculo Esquelético/crecimiento & desarrollo , Transducción de Señal/fisiología , Esfingosina/análogos & derivados , Canales Catiónicos TRPC/metabolismo , Animales , Calpaína/genética , Calpaína/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Conexina 43/genética , Ratones , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/fisiología , Técnicas de Placa-Clamp , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Esfingosina/metabolismo , Canales Catiónicos TRPC/genética
11.
Cell Mol Life Sci ; 67(21): 3711-23, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20496097

RESUMEN

Glucagon-like peptide-1 (GLP-1) is an insulinotropic peptide with neurotrophic properties, as assessed in animal cell models. Exendin-4, a GLP-1 analogue, has been recently approved for the treatment of type 2 diabetes mellitus. The aim of this study was to morphologically, structurally, and functionally characterize the differentiating actions of exendin-4 using a human neuronal cell model (i.e., SH-SY5Y cells). We found that exendin-4 increased the number of neurites paralleled by dramatic changes in intracellular actin and tubulin distribution. Electrophysiological analyses showed an increase in cell membrane surface and in stretch-activated-channels sensitivity, an increased conductance of Na(+) channels and amplitude of Ca(++) currents (T- and L-type), typical of a more mature neuronal phenotype. To our knowledge, this is the first demonstration that exendin-4 promotes neuronal differentiation in human cells. Noteworthy, our data support the claimed favorable role of exendin-4 against diabetic neuropathy as well as against different neurodegenerative diseases.


Asunto(s)
Péptido 1 Similar al Glucagón/análogos & derivados , Neurogénesis/efectos de los fármacos , Neuronas/citología , Péptidos/farmacología , Ponzoñas/farmacología , Factores Despolimerizantes de la Actina/metabolismo , Actinas/ultraestructura , Línea Celular , Citoesqueleto/ultraestructura , Exenatida , Expresión Génica , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Canales Iónicos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación , Tretinoina/farmacología , Tubulina (Proteína)/ultraestructura
12.
FASEB J ; 23(10): 3494-505, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19584303

RESUMEN

Adipose tissue is a dynamic endocrine organ with a central role in metabolism regulation. Functional differences in adipose tissue seem associated with the regional distribution of fat depots, in particular in subcutaneous and visceral omental pads. Here, we report for the first time the isolation of human adipose-derived adult stem cells from visceral omental and subcutaneous fat (V-ASCs and S-ASCs, respectively) from the same subject. Immunophenotyping shows that plastic culturing selects homogeneous cell populations of V-ASCs and S-ASCs from the corresponding stromal vascular fractions (SVFs), sharing typical markers of mesenchymal stem cells. Electron microscopy and electrophysiological and real-time RT-PCR analyses confirm the mesenchymal stem nature of both V-ASCs and S-ASCs, while no significant differences in a limited pattern of cytokine/chemokine expression can be detected. Similar to S-ASCs, V-ASCs can differentiate in vitro toward adipogenic, osteogenic, chondrogenic, muscular, and neuronal lineages, as demonstrated by histochemical, immunofluorescence, real-time RT-PCR, and electrophysiological analyses, suggesting the multipotency of such adult stem cells. Our data demonstrate that both visceral and subcutaneous adipose tissues are a source of pluripotent stem cells with multigermline potential. However, the visceral rather than the subcutaneous ASC could represent a more appropriate in vitro cell model for investigating the molecular mechanisms implicated in the pathophysiology of metabolic disorders such as obesity.


Asunto(s)
Tejido Adiposo/citología , Células Madre Adultas/fisiología , Colon/citología , Células Madre Pluripotentes/fisiología , Piel/citología , Células Madre Adultas/citología , Diferenciación Celular , Linaje de la Célula , Separación Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes/citología , Vísceras/citología
13.
J Mol Cell Cardiol ; 47(2): 335-45, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19465027

RESUMEN

The possibility that resident myocardial progenitor cells may be re-activated by transplantation of exogenous stem cells into the post-infarcted heart has been suggested as a possible mechanism to explain the heart's functional improvement after stem cell therapy. Here we studied whether differentiation of mouse neonatal immature cardiomyocytes in vitro was influenced by mouse skeletal myoblasts C2C12, wild type or engineered to secrete the cardiotropic hormone relaxin. The cultured cardiomyocytes formed spontaneously beating clusters and temporally exhibited cardiac immunophenotypical (cKit, atrial natriuretic peptide, troponin T, connexin-43, HCN4) and electrical features (inward voltage-dependent Na(+), T- and L-type Ca(2+) currents, outward and inward K(+) currents, I(f) pacemaker current). These clusters were functionally connected through nanotubular structures and undifferentiated cardiac cells in the form of flattened stripes, bridging the clusters through connexin-43-containing gap junctions. These findings suggested the existence of long distance cell-to-cell communications among the cardiomyocyte aggregates involved in the intercellular transfer of Ca(2+) signals and organelles, likely required for coordination of myocardial differentiation. Co-presence of the myoblasts greatly increased cardiomyocyte differentiation and the amount of intercellular connections. In fact, these cells formed a structural support guiding elongation of nanotubules and stripe-like cells. The secretion of relaxin by the engineered myoblasts accelerated and enhanced the cardiomyogenic potential of the co-culture. These findings underscore the possibility that grafted myoblasts and cardiotropic factors, such as relaxin, may influence regeneration of resident immature cardiac cells, thus adding a tile to the mosaic of mechanisms involved in the functional benefits of cell transplantation for cardiac repair.


Asunto(s)
Comunicación Celular , Diferenciación Celular , Mioblastos Esqueléticos/metabolismo , Miocitos Cardíacos/citología , Relaxina/metabolismo , Animales , Animales Recién Nacidos , Membrana Celular/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Conexina 43/metabolismo , Fenómenos Electrofisiológicos , Inmunofenotipificación , Activación del Canal Iónico , Ratones , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/ultraestructura , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Factores de Tiempo
14.
Front Physiol ; 10: 1000, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31447692

RESUMEN

Some adipokines known to regulate food intake at a central level can also affect gastrointestinal motor responses. These are recognized to be peripheral signals able to influence feeding behavior as well. In this view, it has been recently observed that adiponectin (ADPN), which seems to have a role in sending satiety signals at the central nervous system level, actually affects the mechanical responses in gastric strips from mice. However, at present, there are no data in the literature about the electrophysiological effects of ADPN on gastric smooth muscle. To this aim, we achieved experiments on smooth muscle cells (SMCs) of gastric fundus to find out a possible action on SMC excitability and on membrane phenomena leading to the mechanical response. Experiments were made inserting a microelectrode in a single cell of a muscle strip of the gastric fundus excised from adult female mice. We found that ADPN was able to hyperpolarize the resting membrane potential, to enhance the delayed rectifier K+ currents and to reduce the voltage-dependent Ca2+ currents. Our overall results suggest an inhibitory action of ADPN on gastric SMC excitation-contraction coupling. In conclusion, the depressant action of ADPN on the gastric SMC excitability, here reported for the first time, together with its well-known involvement in metabolism, might lead us to consider a possible contribution of ADPN also as a peripheral signal in the hunger-satiety cycle and thus in feeding behavior.

15.
Neurol Res ; 30(2): 155-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18397607

RESUMEN

INTRODUCTION AND OBJECTIVES: Evidence showed that physical forces, as passive stretching or active contraction, may counteract various kinds of skeletal muscle atrophy due, for instance, to muscle immobilization, pathophysiology or denervation. Accordingly, active muscle contraction induced by functional electric stimulation is helpful to reduce the muscle atrophic state in denervated man. Moreover, there is evidence that also passive mechanical stimulation of the sarcolemnic membrane may reduce the atrophic muscle state. As to the mechanisms by which mechanical stimulation modulates muscle physiology and pathophysiology, there is a growing list of facts that signaling pathway to the nucleus involves stretch activated channels (SACs) of the sarcolemma and the cytoskeleton. SACs activation allowed a Ca(2+) inflow that activates Ca(2+)-dependent molecular signals. Cytoskeleton may be activated by Ca(2+)-dependent and -independent paths and its contraction and elongation represent not only a mechanical signal to the nucleus but also a stimulus for many molecular signals. The aim of this work was to evaluate in soleus muscle of the rat, the mechano-sensitivity of SACs before and after medium and long term denervation. METHODS: Electrophysiologic experiments were made in normal and denervated Soleus muscle of Wistar rats. Currents were recorded in voltage clamp by intracellular microelectrodes inserted in a single fiber. RESULTS: Our findings demonstrated that SACs were expressed in normal soleus muscle and that SAC currents were potentiated by muscle stretching. Another important result was that the sensitivity to stretching increased after denervation and was particularly evident in long term denervated muscles. DISCUSSION: The reported effects are in agreement with the effects of exercise on inducing muscle hypertrophy or with the positive effects on repairing the atrophic state of skeletal muscles by mechanical stimulation or, in denervated humans, by the functional electrical stimulation (FES).


Asunto(s)
Potenciales de la Membrana/fisiología , Desnervación Muscular/métodos , Músculo Esquelético/fisiología , Animales , Masculino , Técnicas de Placa-Clamp/métodos , Estimulación Física/métodos , Ratas , Ratas Wistar , Factores de Tiempo
16.
World J Gastroenterol ; 24(8): 882-893, 2018 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-29491682

RESUMEN

AIM: To investigate the signaling pathways involved in the relaxin (RLX) effects on ileal preparations from mice through mechanical and electrophysiological experiments. METHODS: For mechanical experiments, ileal preparations from female mice were mounted in organ baths containing Krebs-Henseleit solution. The mechanical activity was recorded via force-displacement transducers, which were coupled to a polygraph for continuous recording of isometric tension. Electrophysiological measurements were performed in current- and voltage-clamp conditions by a microelectrode inserted in a single smooth muscle cell (SMC) of the ileal longitudinal layer. Both the membrane passive properties and inward voltage-dependent L-type Ca2+ currents were recorded using suitable solutions and voltage stimulation protocols. RESULTS: Mechanical experiments showed that RLX induced a decay of the basal tension and a reduction in amplitude of the spontaneous contractions. The effects of RLX were partially reduced by 1H-[1,2,4]oxadiazolo[4,3-a]-quinoxalin-1-one (ODQ) or 9-cyclopentyladenine mesylate (9CPA), inhibitors of guanylate cyclase (GC) and adenylate cyclase (AC), respectively, and were abolished in the concomitant presence of both drugs. Electrophysiological experiments demonstrated that RLX directly influenced the biophysical properties of ileal SMCs, decreasing the membrane conductance, hyperpolarizing the resting membrane potential, reducing the L-type calcium current amplitude and affecting its kinetics. The voltage dependence of the current activation and inactivation time constant was significantly speeded by RLX. Each electrophysiological effect of RLX was reduced by ODQ or 9CPA, and abolished in the concomitant presence of both drugs as observed in mechanical experiments. CONCLUSION: Our new findings demonstrate that RLX influences ileal muscle through a dual mechanism involving both GC and AC.


Asunto(s)
Motilidad Gastrointestinal/fisiología , Íleon/fisiología , Relaxina/metabolismo , Transducción de Señal/fisiología , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Femenino , Potenciales de la Membrana/fisiología , Ratones , Microelectrodos , Modelos Animales , Contracción Muscular/fisiología , Músculo Liso/citología , Músculo Liso/fisiología , Miocitos del Músculo Liso/metabolismo , Técnicas de Placa-Clamp
17.
World J Gastroenterol ; 24(35): 4028-4035, 2018 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-30254407

RESUMEN

AIM: To investigate whether the adipocytes derived hormone adiponectin (ADPN) affects the mechanical responses in strips from the mouse gastric fundus. METHODS: For functional experiments, gastric strips from the fundal region were cut in the direction of the longitudinal muscle layer and placed in organ baths containing Krebs-Henseleit solution. Mechanical responses were recorded via force-displacement transducers, which were coupled to a polygraph for continuous recording of isometric tension. Electrical field stimulation (EFS) was applied via two platinum wire rings through which the preparation was threaded. The effects of ADPN were investigated on the neurally-induced contractile and relaxant responses elicited by EFS. The expression of ADPN receptors, Adipo-R1 and Adipo-R2, was also evaluated by touchdown-PCR analysis. RESULTS: In the functional experiments, EFS (4-16 Hz) elicited tetrodotoxin (TTX)-sensitive contractile responses. Addition of ADPN to the bath medium caused a reduction in amplitude of the neurally-induced contractile responses (P < 0.05). The effects of ADPN were no longer observed in the presence of the nitric oxide (NO) synthesis inhibitor L-NG-nitro arginine (L-NNA) (P > 0.05). The direct smooth muscle response to methacholine was not influenced by ADPN (P > 0.05). In carbachol precontracted strips and in the presence of guanethidine, EFS induced relaxant responses. Addition of ADPN to the bath medium, other than causing a slight and progressive decay of the basal tension, increased the amplitude of the neurally-induced relaxant responses (P < 0.05). Touchdown-PCR analysis revealed the expression of both Adipo-R1 and Adipo-R2 in the gastric fundus. CONCLUSION: The results indicate for the first time that ADPN is able to influence the mechanical responses in strips from the mouse gastric fundus.


Asunto(s)
Adiponectina/fisiología , Fundus Gástrico/fisiología , Músculo Liso/fisiología , Adipocitos/metabolismo , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/metabolismo , Animales , Estimulación Eléctrica , Femenino , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/fisiología , Relajación Muscular/fisiología , Receptores de Adiponectina/metabolismo
18.
J Chem Neuroanat ; 90: 87-97, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29274379

RESUMEN

Human striatal precursor cells (HSPs) isolated from ganglionic eminence may differentiate in electrophysiologically functional excitable neuron-like cells and a number of endogenous molecules such as hormones, neurotransmitters or growth factors can actually regulate neuronal growing and differentiation. The purpose of this research was to assess, by electrophysiological and immunocytochemical analysis, if the type of culture medium could specifically impact on the neuronal differentiation potential of HSPs. Accordingly, HSPs were maintained in different inductive media such as cortical and spinal cord conditioned media, and we estimated the possible changes in the main ion currents, excitability and expression of neuronal markers indicative of neuronal differentiation. Our results have shown that 36 h exposure to each of the conditioned media, with their blend of autocrine and paracrine growth factors, was able to modify significantly the electrophysiological membrane properties and the functional expression of inward ionic currents in selected neuronal HSPs. Moreover, although both types of conditioned media determined neuronal maturation (increased neuritogenesis and increased expression of neuronal and striatal markers), each of them leads to the occurrence of different functional features. Particularly, the spinal medium caused a stronger depolarization of the membrane potential and significantly increased the amplitude of Na+ current as well as L- and N- type Ca2+ currents, definitely modifying their kinetics. In contrast, the cortical medium mainly caused a significant and more marked increase of the membrane conductance and time constant values. These results strongly support the plasticity of our cellular model that, although already committed towards a specific phenotype, it can be differently affected by the conditioned media, thereby resulting functionally modifiable according to environmental cues.


Asunto(s)
Diferenciación Celular/fisiología , Células-Madre Neurales/citología , Encéfalo/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Médula Espinal/metabolismo
19.
Ann N Y Acad Sci ; 1041: 444-5, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15956744

RESUMEN

We have investigated the interaction between mouse skeletal myoblasts and rat cardiomyocytes in coculture and the influence of relaxin. Connexin43 expression, Lucifer yellow microinjection, Ca2+ propagation, and electrophysiological analysis have shown that myoblasts and cardiomyocytes are coupled by functional gap junctions. Cardiomyocytes and relaxin upregulated connexin43 expression and gap junctional communication in myoblasts. Relaxin also increased transjunctional current between myoblasts and between myoblasts and cardiomyocytes. In conclusion, relaxin potentiates the intercellular coupling, upregulating the transcellular exchange of regulatory molecules between myoblasts and cardiomyocytes, including Ca2+. This could favor the transdifferentiation of myoblasts toward a cardiac phenotype.


Asunto(s)
Envejecimiento/fisiología , Mioblastos Esqueléticos/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Relaxina/farmacología , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Técnicas de Cocultivo , Conexina 43/metabolismo , Uniones Comunicantes/efectos de los fármacos , Ratones , Mioblastos Esqueléticos/metabolismo , Miocitos Cardíacos/metabolismo , Ratas
20.
Eur J Pharmacol ; 758: 40-52, 2015 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-25843408

RESUMEN

Big efforts have been dedicated up to now to identify novel targets for cancer treatment. The peculiar biophysical profile and the atypical ionic channels activity shown by diverse types of human cancers suggest that ion channels may be possible targets in cancer therapy. Earlier studies have shown that melatonin exerts an oncostatic action on different tumors. In particular, it was shown that melatonin was able to inhibit growth/viability and proliferation, to reduce the invasiveness and metastatic properties of human estrogen-sensitive breast adenocarcinoma MCF-7 cell line cultured in growth medium, with substantial impairments of epidermal growth factor (EGF) and Notch-1-mediated signaling. The purpose of this work was to evaluate on MCF-7 cells the possible effects of melatonin on the biophysical features known to have a role in proliferation and differentiation, by using the patch-clamp technique. Our results show that in cells cultured in growth as well as in differentiation medium melatonin caused a hyperpolarization of resting membrane potential paralleled by significant changes of the inward Ca(2+) currents (T- and L-type), outward delayed rectifier K(+) currents and cell capacitance. All these effects are involved in MCF-7 growth and differentiation. These findings strongly suggest that melatonin, acting as a modulator of different voltage-dependent ion channels, might be considered a new promising tool for specifically disrupting cell viability and differentiation pathways in tumour cells with possible beneficial effects on cancer therapy.


Asunto(s)
Canales de Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Melatonina/farmacología , Potenciales de la Membrana/efectos de los fármacos , Canales de Potasio/metabolismo , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Células MCF-7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA