Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Cardiovasc Dev Dis ; 9(7)2022 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-35877579

RESUMEN

Cardiovascular diseases (CVDs) are a significant burden globally and are especially prevalent in obese and/or diabetic populations. Epicardial adipose tissue (EAT) surrounding the heart has been implicated in the development of CVDs as EAT can shift from a protective to a maladaptive phenotype in diseased states. In diabetic and obese patients, an elevated EAT mass both secretes pro-fibrotic/pro-inflammatory adipokines and forms intramyocardial fibrofatty infiltrates. This narrative review considers the proposed pathophysiological roles of EAT in CVDs. Diabetes is associated with a disordered energy utilization in the heart, which promotes intramyocardial fat and structural remodeling. Fibrofatty infiltrates are associated with abnormal cardiomyocyte calcium handling and repolarization, increasing the probability of afterdepolarizations. The inflammatory phenotype also promotes lateralization of connexin (Cx) proteins, undermining unidirectional conduction. These changes are associated with conduction heterogeneity, together creating a substrate for atrial fibrillation (AF). EAT is also strongly implicated in coronary artery disease (CAD); inflammatory adipokines from peri-vascular fat can modulate intra-luminal homeostasis through an "outside-to-inside" mechanism. EAT is also a significant source of sympathetic neurotransmitters, which promote progressive diastolic dysfunction with eventual cardiac failure. Further investigations on the behavior of EAT in diabetic/obese patients with CVD could help elucidate the pathogenesis and uncover potential therapeutic targets.

2.
Cells ; 10(9)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34572150

RESUMEN

Atrial fibrillation is very common among the elderly and/or obese. While myocardial fibrosis is associated with atrial fibrillation, the exact mechanisms within atrial myocytes and surrounding non-myocytes are not fully understood. This review considers the potential roles of myocardial fibroblasts and myofibroblasts in fibrosis and modulating myocyte electrophysiology through electrotonic interactions. Coupling with (myo)fibroblasts in vitro and in silico prolonged myocyte action potential duration and caused resting depolarization; an optogenetic study has verified in vivo that fibroblasts depolarized when coupled myocytes produced action potentials. This review also introduces another non-myocyte which may modulate both myocardial (myo)fibroblasts and myocytes: epicardial adipose tissue. Epicardial adipocytes are in intimate contact with myocytes and (myo)fibroblasts and may infiltrate the myocardium. Adipocytes secrete numerous adipokines which modulate (myo)fibroblast and myocyte physiology. These adipokines are protective in healthy hearts, preventing inflammation and fibrosis. However, adipokines secreted from adipocytes may switch to pro-inflammatory and pro-fibrotic, associated with reactive oxygen species generation. Pro-fibrotic adipokines stimulate myofibroblast differentiation, causing pronounced fibrosis in the epicardial adipose tissue and the myocardium. Adipose tissue also influences myocyte electrophysiology, via the adipokines and/or through electrotonic interactions. Deeper understanding of the interactions between myocytes and non-myocytes is important to understand and manage atrial fibrillation.


Asunto(s)
Tejido Adiposo/metabolismo , Fibrilación Atrial/patología , Fibrosis Endomiocárdica/patología , Potenciales de Acción/fisiología , Adipocitos/fisiología , Adipoquinas/fisiología , Tejido Adiposo/patología , Fibrilación Atrial/metabolismo , Cardiomiopatías/patología , Fenómenos Electrofisiológicos , Fibrosis Endomiocárdica/metabolismo , Mapeo Epicárdico/métodos , Fibroblastos/metabolismo , Fibroblastos/fisiología , Fibrosis/patología , Corazón/fisiología , Atrios Cardíacos/patología , Humanos , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Miofibroblastos , Pericardio/patología
3.
Heliyon ; 6(1): e03031, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31909243

RESUMEN

Connexin 43 expression (Cx43) is increased in cardiac fibroblasts (CFs) following myocardial infarction. Here, potential mediators responsible for increasing Cx43 expression and effects of differential CF phenotype on cardiac myocyte (CM) function were investigated. Stimulating adult rat CFs with proinflammatory mediators revealed that interleukin 1ß (IL-1ß) significantly enhanced Cx43 levels through the IL-1ß pathway. Additionally, IL-1ß reduced mRNA levels of the myofibroblast (MF) markers: (i) connective tissue growth factor (CTGF) and (ii) α smooth muscle actin (αSMA), compared to control CFs. A co-culture adult rat CM:CF model was utilised to examine cell-to-cell interactions. Transfer of calcein from CMs to underlying CFs suggested functional gap junction formation. Functional analysis revealed contraction duration (CD) of CMs was shortened in co-culture with CFs, while treatment of CFs with IL-1ß reduced this mechanical effect of co-culture. No effect on action potential rise time or duration of CMs cultured with control or IL-1ß-treated CFs was observed. These data demonstrate that stimulating CFs with IL-1ß increases Cx43 and reduces MF marker expression, suggesting altered cell phenotype. These changes may underlie the reduced mechanical effects of IL-1ß treated CFs on CD of co-cultured CMs and therefore have an implication for our understanding of heterocellular interactions in cardiac disease.

4.
J Pharmacol Toxicol Methods ; 100: 106601, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31255746

RESUMEN

INTRODUCTION: A volume-pressure sensor and tail-cuff method for monitoring blood pressure is non-invasive and inexpensive. This method requires animals to be restrained or subjected to anesthesia, but comparative effects of these manipulations on hemodynamic parameters have not been documented. METHODS: Using a volume-pressure sensor and tail-cuff, we serially measured blood pressure and heart rate in normotensive adult male Lewis rats after light isoflurane-induced anesthesia (5% induction, 1% maintenance) and, following untrained restraint. Blood pressure was recorded until the acquisition of three complete measurements without the range of replicate mean arterial pressures exceeding 15 mmHg (steady-state). RESULTS: Averages for the entire series of consecutive measurements indicated that restraint yielded significantly higher systolic and diastolic blood pressure than anesthesia (P < .05), but heart rate was not affected. Following stabilization at steady-state, there were no significant differences in intra- or inter-day hemodynamic values between the restraint and isoflurane groups. The inter-day coefficient of variation for systolic pressure was 13-23% for isoflurane and 9-14% for restraint. Bland-Altman analysis showed wide limits of agreement (±59 mmHg systolic; ±49 mmHg diastolic pressure) between restraint and isoflurane measurements. DISCUSSION: Isoflurane caused more variability but there was agreement in BP evaluation by the isoflurane and restraint methods. Using the VPR system, light isoflurane-induced anesthesia and restraint could effectively be used to screen and quantify overt changes in hemodynamic parameters for cardiovascular research utilizing laboratory rodents.


Asunto(s)
Presión Sanguínea/fisiología , Hemodinámica/fisiología , Isoflurano/farmacología , Restricción Física/fisiología , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/farmacología , Animales , Presión Arterial/fisiología , Determinación de la Presión Sanguínea/métodos , Determinación de la Presión Sanguínea/veterinaria , Frecuencia Cardíaca/fisiología , Isoflurano/administración & dosificación , Masculino , Ratas , Ratas Endogámicas Lew , Cola (estructura animal)/irrigación sanguínea
5.
Autoimmunity ; 49(8): 563-570, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27562362

RESUMEN

Rheumatic fever and rheumatic heart disease (RF/RHD) develop following repeated infection with group A streptococci (GAS). We used the Rat Autoimmune Valvulitis (RAV) model of RF/RHD to demonstrate that repetitive booster immunization with GAS-derived recombinant M protein (rM5) resulted in an enhanced anti-cardiac myosin antibody response that may contribute to the breaking of immune tolerance leading to RF/RHD and increased infiltration of heart valves by mononuclear cells. With each boost, more inflammatory cells were observed infiltrating heart tissue which could lead to severe cardiac damage. We also found evidence that both complement and anti-M protein antibodies in serum from rM5-immunized rats have the potential to contribute to inflammation in heart valves by activating cardiac endothelium. More importantly, we have demonstrated by electrocardiography for the first time in the RAV model that elongation of P-R interval follows repetitive boost with rM5. Our observations provide experimental evidence for cardiac alterations following repeated exposure to GAS M protein with immunological and electrophysiological features resembling that seen in humans following recurrent GAS infection.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Endocardio/inmunología , Endocardio/patología , Cardiopatía Reumática/etiología , Cardiopatía Reumática/patología , Animales , Autoantígenos/inmunología , Autoinmunidad , Miosinas Cardíacas/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Electrocardiografía , Endocardio/metabolismo , Células Endoteliales/metabolismo , Femenino , Inmunización , Inmunización Secundaria , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratas , Cardiopatía Reumática/diagnóstico , Cardiopatía Reumática/metabolismo , Streptococcus pyogenes/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
6.
PLoS One ; 10(9): e0138048, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26393919

RESUMEN

Oxytocin and its receptor are synthesised in the heart and blood vessels but effects of chronic activation of this peripheral oxytocinergic system on cardiovascular function are not known. In acute studies, systemic administration of low dose oxytocin exerted a protective, preconditioning effect in experimental models of myocardial ischemia and infarction. In this study, we investigated the effects of chronic administration of low dose oxytocin following angiotensin II-induced hypertension, cardiac hypertrophy and renal damage. Angiotensin II (40 µg/Kg/h) only, oxytocin only (20 or 100 ng/Kg/h), or angiotensin II combined with oxytocin (20 or 100 ng/Kg/h) were infused subcutaneously in adult male Sprague-Dawley rats for 28 days. At day 7, oxytocin or angiotensin-II only did not change hemodynamic parameters, but animals that received a combination of oxytocin and angiotensin-II had significantly elevated systolic, diastolic and mean arterial pressure compared to controls (P < 0.01). Hemodynamic changes were accompanied by significant left ventricular cardiac hypertrophy and renal damage at day 28 in animals treated with angiotensin II (P < 0.05) or both oxytocin and angiotensin II, compared to controls (P < 0.01). Prolonged oxytocin administration did not affect plasma concentrations of renin and atrial natriuretic peptide, but was associated with the activation of calcium-dependent protein phosphatase calcineurin, a canonical signalling mechanism in pressure overload-induced cardiovascular disease. These data demonstrate that oxytocin accelerated angiotensin-II induced hypertension and end-organ renal damage, suggesting caution should be exercised in the chronic use of oxytocin in individuals with hypertension.


Asunto(s)
Angiotensina II/efectos adversos , Cardiomegalia/inducido químicamente , Hipertensión/inducido químicamente , Riñón/efectos de los fármacos , Oxitocina/administración & dosificación , Animales , Factor Natriurético Atrial/sangre , Calcineurina/metabolismo , Inyecciones Subcutáneas , Riñón/patología , Masculino , Ratas , Ratas Sprague-Dawley , Renina/sangre
7.
Biochem Soc Trans ; 43(3): 513-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26009200

RESUMEN

Gap junctions are channels which allow electrical signals to propagate through the heart from the sinoatrial node and through the atria, conduction system and onwards to the ventricles, and hence are essential for co-ordinated cardiac contraction. Twelve connexin (Cx) proteins make up one gap junction channel, of which there are three main subtypes in the heart; Cx40, Cx43 and Cx45. In the cardiac myocyte, gap junctions are present mainly at the intercalated discs between neighbouring myocytes, and assist in rapid electrical conduction throughout the ventricular myocardium. Fibroblasts provide the structural skeleton of the myocardium and fibroblast numbers significantly increase in heart disease. Fibroblasts also express connexins and this may facilitate heterocellular electrical coupling between myocytes and fibroblasts in the setting of cardiac disease. Interestingly, cardiac fibroblasts have been demonstrated to increase Cx43 expression in experimental models of myocardial infarction and functional gap junctions between myocytes and fibroblasts have been reported. Therefore, in the setting of heart disease enhanced cardiac myocyte: fibroblast coupling may influence the electrical activity of the myocyte and contribute to arrhythmias.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Conexina 43/biosíntesis , Uniones Comunicantes/genética , Miocardio/patología , Miocitos Cardíacos/metabolismo , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Síndrome de Brugada , Trastorno del Sistema de Conducción Cardíaco , Comunicación Celular/genética , Conexina 43/metabolismo , Conductividad Eléctrica , Fibroblastos/metabolismo , Fibroblastos/patología , Uniones Comunicantes/metabolismo , Sistema de Conducción Cardíaco/anomalías , Sistema de Conducción Cardíaco/fisiopatología , Humanos , Miocardio/metabolismo , Miocitos Cardíacos/patología
8.
Cardiovasc Res ; 92(1): 169-77, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21697146

RESUMEN

AIMS: We investigated the role of the inward rectifier K(+) channel (K(IR)) in the renal interlobular artery (ILA). The ILA supplies the afferent arteriole and ranges in diameter from >100 µm near its origin at the arcuate artery to <30 µm at its most distal segment. METHODS AND RESULTS: Vasodilatory responses to elevated extracellular K(+) (15 mmol/L) and vasoconstrictor responses due to K(IR) blockade by Ba(2+) (10-100 µmol/L) were assessed in in vitro perfused hydronephrotic rat kidneys. The distal ILA (26 ± 1 µm) exhibited K(+)-induced dilation and Ba(2+)-induced vasoconstriction, whereas neither response was observed in the proximal ILA (108 ± 3 µm). The intermediate ILA (55 ± 1 µm) exhibited a modest K(+)-induced vasodilatation, but no Ba(2+)-induced vasoconstriction. The K(+)-induced dilations were blocked by Ba(2+), but not by ouabain. Ba(2+)-induced depolarization, measured in ILA segments from normal kidneys, decreased with the increasing diameter. Patch-clamp studies demonstrated that the K(IR) current (I(KIR)) density also was inversely correlated with ILA segment diameter. Myocytes from afferent arterioles and distal ILAs exhibited similarly large I(KIR), whereas this current was absent in proximal ILA myocytes. Finally, we found that Ba(2+) attenuated myogenic vasoconstriction, suggesting an involvement of I(KIR). The previously shown pattern of myogenic reactivity of the ILA (distal > intermediate > proximal) mirrors the distribution of I(KIR) reported in the present study, further supporting a role for I(KIR). CONCLUSION: Our findings indicate differences in the magnitude of I(KIR) along the ILA and suggest that the influence of K(IR) on reactivity increases as vessel diameter decreases from proximal to distal regions.


Asunto(s)
Canales de Potasio de Rectificación Interna/fisiología , Arteria Renal/fisiología , Animales , Arteriolas/fisiología , Bario/farmacología , Masculino , Músculo Liso Vascular/fisiología , Técnicas de Placa-Clamp , Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Vasoconstricción , Vasodilatación/efectos de los fármacos
9.
Am J Physiol Heart Circ Physiol ; 301(2): H514-22, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21572008

RESUMEN

Chemotactic movement of myofibroblasts is recognized as a common means for their sequestration to the site of tissue injury. Following myocardial infarction (MI), recruitment of cardiac myofibroblasts to the infarct scar is a critical step in wound healing. Contractile myofibroblasts express embryonic smooth muscle myosin, α-smooth muscle actin, as well as collagens I and III. We examined the effects of cardiotrophin-1 (CT-1) in the induction of primary rat ventricular myofibroblast motility. Changes in membrane potential (E(m)) and Ca(2+) entry were studied to reveal the mechanisms for induction of myofibroblast migration. CT-1-induced cardiac myofibroblast cell migration, which was attenuated through the inhibition of JAK2 (25 µM AG490), and myosin light chain kinase (20 µM ML-7). Inhibition of K(+) channels (1 mM tetraethylammonium or 100 µM 4-aminopyridine) and nonselective cation channels by 10 µM gadolinium (Gd(3+)) significantly reduced migration in the presence of CT-1. CT-1 treatment caused a significant increase in myosin light chain phosphorylation, which could be inhibited by incubation in Ca(2+)-free conditions or by application of AG490, ML-7, and W7 (100 µM; calmodulin inhibitor). Monitoring myofibroblast membrane potential with potentiometric fluorescent DiBAC(4)(3) dye revealed a biphasic response to CT-1 consisting of an initial depolarization followed by hyperpolarization. Increased intracellular Ca(2+), as assessed by fluo 3, occurred immediately after membrane depolarization and attenuated at the time of maximal hyperpolarization. CT-1 exerts chemotactic effects via multiple parallel signaling modalities in ventricular myofibroblasts, including changes in membrane potential, alterations in intracellular calcium, and activation of a number of intracellular signaling pathways. Further study is warranted to determine the precise role of K(+) currents in this process.


Asunto(s)
Quimiotaxis , Citocinas/metabolismo , Miofibroblastos/enzimología , Quinasa de Cadena Ligera de Miosina/metabolismo , Análisis de Varianza , Animales , Calcio/metabolismo , Calmodulina/antagonistas & inhibidores , Calmodulina/metabolismo , Miosinas Cardíacas/metabolismo , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Gadolinio/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/enzimología , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Masculino , Potenciales de la Membrana , Miofibroblastos/efectos de los fármacos , Cadenas Ligeras de Miosina/metabolismo , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Fosforilación , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Canales de Potasio/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Espectrometría de Fluorescencia , Factores de Tiempo
10.
J Am Soc Nephrol ; 19(1): 69-76, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18178799

RESUMEN

The afferent and efferent arterioles regulate the inflow and outflow resistance of the glomerulus, acting in concert to control the glomerular capillary pressure and glomerular filtration rate. The myocytes of these two vessels are remarkably different, especially regarding electromechanical coupling. This study investigated the expression and function of inward rectifier K(+) channels in these two vessels using perfused hydronephrotic rat kidneys and arterioles and myocytes isolated from normal rat kidneys. In afferent arterioles pre-constricted with angiotensin II, elevating [K(+)](0) from 5 to 15 mmol/L induced hyperpolarization (-27 +/- 2 to 41 +/- 3 mV) and vasodilation (6.6 +/- 0.9 to 13.1 +/- 0.6 microm). This manipulation also attenuated angiotensin II-induced Ca(2+) signaling, an effect blocked by 100 micromol/LBa(2+). By contrast, elevating [K(+)](o) did not alter angiotensin II-induced Ca2(+) signaling or vasoconstriction in efferent arterioles, even though a significant hyperpolarization was observed (from -30 +/- 1 to 37 +/- 3 mV, P = 0.003). Both vessels expressed mRNA for Kir2.1 and exhibited anti-Kir2.1 antibody labeling.Patch-clamp measurements revealed prominent inwardly rectifying and Ba(2+)-sensitive currents in afferent and efferent arteriolar myocytes. Our findings indicate that both arterioles express an inward rectifier K(+) current, but that modulation of this current alters responsiveness of only the a different arteriole. The expression of Kir in the efferent arteriole, a resistance vessel whose tone is not affected by membrane potential, is intriguing and may suggest a novel function of this channel in the renal microcirculation.


Asunto(s)
Arteriolas/fisiología , Regulación de la Expresión Génica , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/fisiología , Canales de Potasio de Rectificación Interna/genética , Angiotensina II/farmacología , Animales , Calcio/fisiología , Células Musculares/fisiología , Músculo Liso Vascular/fisiología , ARN Mensajero/genética , Ratas , Transducción de Señal , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
11.
J Physiol ; 583(Pt 1): 225-36, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17569734

RESUMEN

Intercellular coupling between ventricular myocytes and myofibroblasts was studied by co-culturing adult rabbit ventricular myocytes with previously prepared layers of cardiac myofibroblasts. Intercellular coupling was examined by: (i) tracking the movement of the fluorescent dye calcein; (ii) immunostaining for connexin 43 (Cx43); and (iii) measurement of intracellular [Ca2+] ([Ca2+]i). The effects of stimulating ventricular myocytes on the underlying myofibroblasts was examined by confocal measurements of [Ca2+]i using fluo-3. When ventricular myocytes were preloaded with calcein and co-cultured with myofibroblasts for 24 h, calcein fluorescence was detected in 52+/-4% (n=8 co-cultures) of surrounding myofibroblasts. Treatment with the gap junction uncoupler heptanol significantly reduced the movement of calcein (12+/-3%, n=6 co-cultures). Immunostaining showed expression of Cx43 in co-cultured myofibroblasts and myocytes. Field stimulation of ventricular myocytes co-cultured with myofibroblasts increased myofibroblast [Ca2+]i, no response was observed after treatment with heptanol or stimulation of fibroblasts in the absence of ventricular myocytes. Action potential parameters of ventricular myocytes in co-culture were similar to control values. However, application of the hormone sphingosine-1-phosphate (S-1-P) to the co-culture caused a depolarization of ventricular myocytes to approximately -20 mV. Sphingosine-1-phosphate had no effect on ventricular myocytes alone. Voltage-clamp measurements of isolated myofibroblasts indicated that S-1-P activated a significant quasi-linear current with a reversal potential of approximately -40 mV. In conclusion, this study shows that stimulation of the ventricular myocyte influences the intracellular Ca2+ of the linked myofibroblast via connexons. These intercellular links also allow the myofibroblasts to influence the electrical activity of the myocyte. This work indicates the nature of the gap junction-mediated bi-directional interactions that occur between ventricular myocyte and myofibroblast.


Asunto(s)
Comunicación Celular/fisiología , Fibroblastos/citología , Uniones Intercelulares/fisiología , Miocitos Cardíacos/citología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Calcio/metabolismo , Comunicación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Colorantes , Conexina 43/metabolismo , Electrofisiología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Heptanol/farmacología , Uniones Intercelulares/efectos de los fármacos , Lisofosfolípidos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Conejos , Esfingosina/análogos & derivados , Esfingosina/farmacología
12.
Biophys J ; 92(11): 4121-32, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17307821

RESUMEN

Functional intercellular coupling has been demonstrated among networks of cardiac fibroblasts, as well as between fibroblasts and atrial or ventricular myocytes. In this study, the consequences of these interactions were examined by implementing the ten Tusscher model of the human ventricular action potential, and coupling it to our electrophysiological models for mammalian ventricular fibroblasts. Our simulations reveal significant electrophysiological consequences of coupling between 1 and 4 fibroblasts to a single ventricular myocyte. These include alterations in plateau height and/or action potential duration (APD) and changes in underlying ionic currents. Two series of simulations were carried out. First, fibroblasts were modeled as a spherical cell with a capacitance of 6.3 pF and an ohmic membrane resistance of 10.7 G Omega. When these "passive" fibroblasts were coupled to a myocyte, they caused slight prolongation of APD with no changes in the plateau, threshold for firing, or rate of initial depolarization. In contrast, when the same myocyte-fibroblast complexes were modeled after addition of the time- and voltage-gated K(+) currents that are expressed in fibroblasts, much more pronounced effects were observed: the plateau height of the action potential was reduced and the APD shortened significantly. In addition, each fibroblast exhibited significant electrotonic depolarizations in response to each myocyte action potential and the resting potential of the fibroblasts closely approximated the resting potential of the coupled ventricular myocyte.


Asunto(s)
Comunicación Celular/fisiología , Fibroblastos/fisiología , Modelos Biológicos , Miocitos Cardíacos/fisiología , Potenciales de Acción/fisiología , Ventrículos Cardíacos/citología , Humanos
13.
Circ Res ; 90(12): 1316-24, 2002 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-12089070

RESUMEN

The kinetic attributes of the afferent arteriole myogenic response were investigated using the in vitro perfused hydronephrotic rat kidney. Equations describing the time course for pressure-dependent vasoconstriction and vasodilation, and steady-state changes in diameter were combined to develop a mathematical model of autoregulation. Transfer functions were constructed by passing sinusoidal pressure waves through the model. These findings were compared with results derived using data from instrumented conscious rats. In each case, a reduction in gain and increase in phase were observed at frequencies of 0.2 to 0.3 Hz. We then examined the impact of oscillating pressure signals. The model predicted that pressure signals oscillating at frequencies above the myogenic operating range would elicit a sustained vasoconstriction the magnitude of which was dependent on peak pressure. These predictions were directly confirmed in the hydronephrotic kidney. Pressure oscillations presented at frequencies of 1 to 6 Hz elicited sustained afferent vasoconstrictions and the magnitude of the response depended exclusively on the peak pressure. Elevated systolic pressure elicited vasoconstriction even if mean pressure was reduced. These findings challenge the view that the renal myogenic response exists to maintain glomerular capillary pressure constant, but rather imply a primary role in protecting against elevated systolic pressures. Thus, the kinetic features of the afferent arteriole allow this vessel to adjust tone in response to changes in systolic pressures presented at the pulse rate. We suggest that the primary function of this mechanism is to protect the glomerulus from the blood pressure power that is normally present at the pulse frequency.


Asunto(s)
Arteria Renal/fisiopatología , Vasoconstricción , Animales , Arteriolas/anatomía & histología , Arteriolas/fisiopatología , Homeostasis , Hidronefrosis/fisiopatología , Riñón/anatomía & histología , Riñón/irrigación sanguínea , Cinética , Masculino , Modelos Teóricos , Técnicas de Cultivo de Órganos , Presión , Ratas , Ratas Sprague-Dawley , Arteria Renal/anatomía & histología , Circulación Renal , Vasodilatación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...