Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Am J Physiol Cell Physiol ; 317(6): C1093-C1106, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31461344

RESUMO

This study explored the mechanism by which Ca2+-activated Cl- channels (CaCCs) encoded by the Tmem16a gene are regulated by calmodulin-dependent protein kinase II (CaMKII) and protein phosphatases 1 (PP1) and 2A (PP2A). Ca2+-activated Cl- currents (IClCa) were recorded from HEK-293 cells expressing mouse TMEM16A. IClCa were evoked using a pipette solution in which free Ca2+ concentration was clamped to 500 nM, in the presence (5 mM) or absence of ATP. With 5 mM ATP, IClCa decayed to <50% of the initial current magnitude within 10 min after seal rupture. IClCa rundown seen with ATP-containing pipette solution was greatly diminished by omitting ATP. IClCa recorded after 20 min of cell dialysis with 0 ATP were more than twofold larger than those recorded with 5 mM ATP. Intracellular application of autocamtide-2-related inhibitory peptide (5 µM) or KN-93 (10 µM), two specific CaMKII inhibitors, produced a similar attenuation of TMEM16A rundown. In contrast, internal application of okadaic acid (30 nM) or cantharidin (100 nM), two nonselective PP1 and PP2A blockers, promoted the rundown of TMEM16A in cells dialyzed with 0 ATP. Mutating serine 528 of TMEM16A to an alanine led to a similar inhibition of TMEM16A rundown to that exerted by either one of the two CaMKII inhibitors tested, which was not observed for three putative CaMKII consensus sites for phosphorylation (T273, T622, and S730). Our results suggest that TMEM16A-mediated CaCCs are regulated by CaMKII and PP1/PP2A. Our data also suggest that serine 528 of TMEM16A is an important contributor to the regulation of IClCa by CaMKII.


Assuntos
Anoctamina-1/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Regulação da Expressão Gênica , Proteínas de Neoplasias/genética , Proteína Fosfatase 1/genética , Proteína Fosfatase 2/genética , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Sequência de Aminoácidos , Animais , Anoctamina-1/metabolismo , Benzilaminas/farmacologia , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cantaridina/farmacologia , Cloretos/metabolismo , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Células HEK293 , Humanos , Transporte de Íons/efeitos dos fármacos , Camundongos , Proteínas de Neoplasias/metabolismo , Ácido Okadáico/farmacologia , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 1/antagonistas & inibidores , Proteína Fosfatase 1/metabolismo , Proteína Fosfatase 2/antagonistas & inibidores , Proteína Fosfatase 2/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Sulfonamidas/farmacologia
2.
Am J Physiol Cell Physiol ; 303(12): C1229-43, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23034390

RESUMO

Pulmonary artery smooth muscle cells (PASMCs) are more depolarized and display higher Ca(2+) levels in pulmonary hypertension (PH). Whether the functional properties and expression of Ca(2+)-activated Cl- channels (Cl(Ca)), an important excitatory mechanism in PASMCs, are altered in PH is unknown. The potential role of Cl(Ca) channels in PH was investigated using the monocrotaline (MCT)-induced PH model in the rat. Three weeks postinjection with a single dose of MCT (50 mg/kg ip), the animals developed right ventricular hypertrophy (heart weight measurements) and changes in pulmonary arterial flow (pulse-waved Doppler imaging) that were consistent with increased pulmonary arterial pressure and PH. Whole cell patch experiments revealed an increase in niflumic acid (NFA)-sensitive Ca(2+)-activated Cl(-) current [I(Cl(Ca))] density in PASMCs from large conduit and small intralobar pulmonary arteries of MCT-treated rats vs. aged-matched saline-injected controls. Quantitative RT-PCR and Western blot analysis revealed that the alterations in I(Cl(Ca)) were accompanied by parallel changes in the expression of TMEM16A, a gene recently shown to encode for Cl(Ca) channels. The contraction to serotonin of conduit and intralobar pulmonary arteries from MCT-treated rats exhibited greater sensitivity to nifedipine (1 µM), an l-type Ca(2+) channel blocker, and NFA (30 or 100 µM, with or without 10 µM indomethacin to inhibit cyclooxygenases) or T16A(Inh)-A01 (10 µM), TMEM16A/Cl(Ca) channel inhibitors, than that of control animals. In conclusion, augmented Cl(Ca)/TMEM16A channel activity is a major contributor to the changes in electromechanical coupling of PA in this model of PH. TMEM16A-encoded channels may therefore represent a novel therapeutic target in this disease.


Assuntos
Canais de Cloreto/biossíntese , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/fisiopatologia , Animais , Anoctamina-1 , Bloqueadores dos Canais de Cálcio/farmacologia , Agonistas dos Canais de Cloreto , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/fisiologia , Inibidores de Ciclo-Oxigenase/farmacologia , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/tratamento farmacológico , Hipertrofia Ventricular Direita/induzido quimicamente , Hipertrofia Ventricular Direita/tratamento farmacológico , Indometacina/farmacologia , Masculino , Monocrotalina/toxicidade , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Nifedipino/farmacologia , Ácido Niflúmico/farmacologia , Técnicas de Patch-Clamp , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Pirimidinas/farmacologia , Ratos , Ratos Wistar , Serotonina/farmacologia , Tiazóis/farmacologia
3.
Circulation ; 122(4): 370-8, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20625113

RESUMO

BACKGROUND: Mechanisms promoting the transition from hypertensive heart disease to heart failure with preserved ejection fraction are poorly understood. When inappropriate for salt status, mineralocorticoid (deoxycorticosterone acetate) excess causes hypertrophy, fibrosis, and diastolic dysfunction. Because cardiac mineralocorticoid receptors are protected from mineralocorticoid binding by the absence of 11-beta hydroxysteroid dehydrogenase, salt-mineralocorticoid-induced inflammation is postulated to cause oxidative stress and to mediate cardiac effects. Although previous studies have focused on salt/nephrectomy in accelerating mineralocorticoid-induced cardiac effects, we hypothesized that hypertensive heart disease is associated with oxidative stress and sensitizes the heart to mineralocorticoid, accelerating hypertrophy, fibrosis, and diastolic dysfunction. METHODS AND RESULTS: Cardiac structure and function, oxidative stress, and mineralocorticoid receptor-dependent gene transcription were measured in sham-operated and transverse aortic constriction (studied 2 weeks later) mice without and with deoxycorticosterone acetate administration, all in the setting of normal-salt diet. Compared with sham mice, sham plus deoxycorticosterone acetate mice had mild hypertrophy without fibrosis or diastolic dysfunction. Transverse aortic constriction mice displayed compensated hypertensive heart disease with hypertrophy, increased oxidative stress (osteopontin and NOX4 gene expression), and normal systolic function, filling pressures, and diastolic stiffness. Compared with transverse aortic constriction mice, transverse aortic constriction plus deoxycorticosterone acetate mice had similar left ventricular systolic pressure and fractional shortening but more hypertrophy, fibrosis, and diastolic dysfunction with increased lung weights, consistent with heart failure with preserved ejection fraction. There was progressive activation of markers of oxidative stress across the groups but no evidence of classic mineralocorticoid receptor-dependent gene transcription. CONCLUSIONS: Pressure-overload hypertrophy sensitizes the heart to mineralocorticoid excess, which promotes the transition to heart failure with preserved ejection fraction independently of classic mineralocorticoid receptor-dependent gene transcription.


Assuntos
Insuficiência Cardíaca/induzido quimicamente , Mineralocorticoides/efeitos adversos , Volume Sistólico/fisiologia , Aldosterona/efeitos adversos , Animais , Fator Natriurético Atrial/genética , Capilares/patologia , Colágeno/genética , Grupo dos Citocromos b/genética , Desoxicorticosterona/farmacologia , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Inflamação/patologia , Inflamação/fisiopatologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/genética , Osteopontina/genética , Estresse Oxidativo/fisiologia , RNA/genética , RNA/isolamento & purificação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Volume Sistólico/efeitos dos fármacos , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/fisiologia
4.
Am J Physiol Cell Physiol ; 298(5): C1006-17, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164377

RESUMO

The purpose of this study was to determine whether extracellular matrix (ECM) composition through integrin receptors modulated the volume-sensitive osmolyte anion channels (VSOACs) in skeletal muscle-derived C2C12 cells. Cl(-) currents were recorded in whole cell voltage-clamped cells grown on laminin (LM), fibronectin (FN), or in the absence of a defined ECM (NM). Basal membrane currents recorded in isotonic media (300 mosmol/kg) were larger in cells grown on FN (3.8-fold at +100 mV) or LM (8.8-fold at +100 mV) when compared with NM. VSOAC currents activated by cell exposure to hypotonic solution were larger in cells grown on LM (1.72-fold at +100 mV) or FN (1.75-fold at +100 mV) compared with NM. Additionally, the kinetics of VSOAC activation was approximately 27% quicker on FN and LM. These currents were tamoxifen sensitive, displayed outward rectification, reversed at the equilibrium potential of Cl(-) and inactivated at potentials >+60 mV. Specific knockdown of beta(1)-integrin by short hairpin RNA interference strongly inhibited the VSOAC Cl(-) currents in cells plated on FN. In conclusion, ECM composition and integrins profoundly influence the biophysical properties and mechanisms of onset of VSOACs.


Assuntos
Canais de Cloreto/metabolismo , Matriz Extracelular/metabolismo , Integrinas/metabolismo , Mioblastos/fisiologia , Simportadores de Sódio-Bicarbonato/metabolismo , Acidose/sangue , Acidose/induzido quimicamente , Acidose/metabolismo , Acidose/urina , Animais , Caspase 3/metabolismo , Morte Celular , Regulação da Expressão Gênica , Ácido Glutâmico , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Ácido Clorídrico/farmacologia , Camundongos , Neurônios/metabolismo , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Simportadores de Sódio-Bicarbonato/genética
5.
Am J Physiol Cell Physiol ; 299(5): C948-59, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20686072

RESUMO

Recently, overexpression of the genes TMEM16A and TMEM16B has been shown to produce currents qualitatively similar to native Ca(2+)-activated Cl(-) currents (I(ClCa)) in vascular smooth muscle. However, there is no information about this new gene family in vascular smooth muscle, where Cl(-) channels are a major depolarizing mechanism. Qualitatively similar Cl(-) currents were evoked by a pipette solution containing 500 nM Ca(2+) in smooth muscle cells isolated from BALB/c mouse portal vein, thoracic aorta, and carotid artery. Quantitative PCR using SYBR Green chemistry and primers specific for transmembrane protein (TMEM) 16A or the closely related TMEM16B showed TMEM16A expression as follows: portal vein > thoracic aorta > carotid artery > brain. In addition, several alternatively spliced variant transcripts of TMEM16A were detected. In contrast, TMEM16B expression was very low in smooth muscle. Western blot analysis with different antibodies directed against TMEM16A revealed a number of products with a consistent band at ∼120 kDa, except portal vein, where an 80-kDa band predominated. TMEM16A protein was identified in the smooth muscle layers of 4-µm-thick slices of portal vein, thoracic aorta, and carotid artery. In isolated myocytes, fluorescence specific to a TMEM16A antibody was detected diffusely throughout the cytoplasm, as well as near the membrane. The same antibody used in Western blot analysis of lysates from vascular tissues also recognized an ∼147-kDa mouse TMEM16A-green fluorescent protein (GFP) fusion protein expressed in HEK 293 cells, which correlated to a similar band detected by a GFP antibody. Patch-clamp experiments revealed that I(ClCa) generated by transfection of TMEM16A-GFP in HEK 293 cells displayed remarkable similarities to I(ClCa) recorded in vascular myocytes, including slow kinetics, steep outward rectification, and a response similar to the pharmacological agent niflumic acid. This study shows that TMEM16A expression is robust in murine vascular smooth muscle cells, consolidating the view that this gene is a viable candidate for the native Ca(2+)-activated Cl(-) channel in this cell type.


Assuntos
Canais de Cloreto/metabolismo , Miócitos de Músculo Liso/fisiologia , Biossíntese de Proteínas , Isoformas de Proteínas/metabolismo , Processamento Alternativo , Animais , Anoctamina-1 , Linhagem Celular , Canais de Cloreto/genética , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Miócitos de Músculo Liso/citologia , Técnicas de Patch-Clamp , Isoformas de Proteínas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Distribuição Tecidual
6.
J Biol Chem ; 284(47): 32507-21, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19767392

RESUMO

The present study was undertaken to determine whether the two ubiquitously expressed Ca(2+)-independent phosphatases PP1 and PP2A regulate Ca(2+)-activated Cl(-) currents (I(Cl(Ca))) elicited by 500 nM [Ca(2+)](i) in rabbit pulmonary artery (PA) myocytes dialyzed with or without 3 mM ATP. Reverse transcription-PCR experiments revealed the expression of PP1alpha, PP1beta/delta, PP1gamma, PP2Aalpha, PP2Abeta, PP2Balpha (calcineurin (CaN) Aalpha), and PP2Bbeta (CaN Abeta) but not PP2Bgamma (CaN Agamma) in rabbit PA. Western blot and immunofluorescence experiments confirmed the presence of all three PP1 isoforms and PP2A. Intracellular dialysis with a peptide inhibitor of calcineurin (CaN-AIP); the non-selective PP1/PP2A inhibitors okadaic acid (0.5, 10, or 30 nM), calyculin A (10 nM), or cantharidin (100 nM); and the selective PP1 inhibitor NIPP-1 (100 pM) potently antagonized the recovery of I(Cl(Ca)) in cells dialyzed with no ATP, whereas the PP2A-selective antagonist fostriecin (30 or 150 nM) was ineffective. The combined application of okadaic acid (10 nM) and CaN-autoinhibitory peptide (50 microM) did not potentiate the response of I(Cl(Ca)) in 0 ATP produced by maximally inhibiting CaN or PP1/PP2A alone. Consistent with the non-additive effects of either classes of phosphatases, the PP1 inhibitor NIPP-1 (100 pM) antagonized the recovery of I(Cl(Ca)) induced by exogenous CaN Aalpha (0.5 microM). These results demonstrate that I(Cl(Ca)) in PA myocytes is regulated by CaN and PP1 and/or PP2A. Our data also suggest the existence of a functional link between these two classes of phosphatases.


Assuntos
Cálcio/química , Cloro/química , Regulação Enzimológica da Expressão Gênica , Pulmão/metabolismo , Miócitos de Músculo Liso/citologia , Monoéster Fosfórico Hidrolases/química , Artéria Pulmonar/citologia , Animais , Cantaridina/farmacologia , Inibidores Enzimáticos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Toxinas Marinhas , Ácido Okadáico/farmacologia , Oxazóis/farmacologia , Técnicas de Patch-Clamp , Coelhos
7.
Circ Res ; 99(12): 1403-10, 2006 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-17095723

RESUMO

Integrins mechanically link the cytoskeleton to the extracellular matrix in cardiac myocytes and are thereby involved in mechanotransduction. Integrins appear to be necessary for cardiac myocyte hypertrophy. To determine the effect of increased integrin ligation and signaling on adult cardiac function, a heart-specific truncated alpha(5) integrin (gain of function) was conditionally expressed in mice. Four days later, we observed an 80% reduction in amplitude of the QRS complex, profound systolic dysfunction, decreased connexin43, loss of gap junctions, and abnormal intercalated discs. Surprisingly, isolated left ventricular myocytes contracted normally and exhibited normal Ca(2+) transients. This suggested that cell/cell electrical and/or mechanical coupling was disrupted. To distinguish electrical from mechanical coupling deficits, we compared the papillary muscle force generated by electrically stimulated versus rapid cooling contractions in which intracellular Ca(2+) is released without electrical depolarization. Both were decreased in the transgenic muscle. However, electrically stimulated contractions were more significantly reduced than rapid cooling contractures. This suggests a component of cell/cell electrical uncoupling. Optical mapping revealed a loss of the normal elliptical isochronal activation pattern implying a loss of preferential conduction through gap junctions. For the first time, we have shown that integrins can regulate both mechanical and electrical coupling in the adult heart, even in the absence of primary hemodynamic alterations. Furthermore, we demonstrated that unregulated integrin activation leads to both contractile dysfunction and arrhythmias.


Assuntos
Junções Comunicantes/fisiologia , Coração/fisiologia , Integrina alfa5/genética , Integrina alfa5/metabolismo , Contração Miocárdica/fisiologia , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/fisiologia , Calreticulina/metabolismo , Cardiomiopatias/patologia , Cardiomiopatias/fisiopatologia , Comunicação Celular/fisiologia , Permeabilidade da Membrana Celular , Conexina 43/metabolismo , Morte Súbita Cardíaca , Eletrocardiografia , Fibrose , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Músculos Papilares/patologia , Músculos Papilares/fisiologia , Fenótipo , Regulação para Cima
8.
BMC Pharmacol Toxicol ; 18(1): 47, 2017 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-28623958

RESUMO

BACKGROUND: The COP9 signalosome (CSN) is a conserved protein complex composed of 8 subunits designated CSN1-CSN8. CSN3 represents the third subunit of the CSN and maintains the integrity of the complex. CSN3 binds to the striated muscle-specific ß1D integrin tail, and its subcellular localization is altered in differentiated skeletal muscle cells. However, the role of CSN3 in skeletal muscle differentiation is unknown. The main goal of this study was to identify whether CSN3 participates in myoblast differentiation and the signalling mechanisms involved using C2C12 cells as a skeletal muscle cell model. METHODS: Small-hairpin (shRNA) was used to knockdown CSN3 in C2C12 cells. Differentiation was evaluated by immunostaining and confocal microscopy. Markers of differentiation, NF-κB signaling and CSN subunits expression, were assessed by immunoblotting and/or immunostaining. Cell proliferation was analysed by cell counting, flow cytometry and a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Data were analyzed by one or two-way analysis of variance (ANOVA) followed by post-hoc testing. RESULTS: Transduction of C2C12 cells with two distinct CSN3 shRNAs led to the production of two cells lines expressing 7% of CSN3 protein (shCSN3-Low) and 43% of CSN3 protein (CSN3-Med) compared to controls. Knockdown of CSN3 was accompanied by destabilization of several CSN subunits and increased nuclear NF-κB localization. shCSN3-Med cells expressed less myogenin and formed shorter and thinner myotubes. In contrast, the shCSN3-Low cells expressed higher levels of myogenin prior and during the differentiation and remained mononucleated throughout the differentiation period. Both CSN3 knockdown cell lines failed to express sarcomeric myosin heavy chain (MHC) protein during differentiation. The fusion index was significantly higher in control cells than in shCSN3-Med cells, whereas shCSN3-Low cells showed no cell fusion. Interestingly, CSN3 knockdown cells exhibited a significantly slower growth rate relative to the control cells. Cell cycle analysis revealed that CSN3 knockdowns delayed in S phase and had increased levels of nuclear p21/Cip1 and p27/Kip1. CONCLUSIONS: This study clarifies the first step toward unrevealing the CSN3/CSN-mediated pathways that controls C2C12 differentiation and proliferation. Further in vivo characterization of CSN/CSN3 may lead to the discovery of novel therapeutic target of skeletal muscle diseases such as muscular dystrophies.


Assuntos
Complexo do Signalossomo COP9/fisiologia , Mioblastos/citologia , Animais , Complexo do Signalossomo COP9/genética , Complexo do Signalossomo COP9/metabolismo , Ciclo Celular , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Camundongos , Músculo Esquelético/citologia , Mioblastos/metabolismo , NF-kappa B/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais
9.
Circ Res ; 94(4): 525-33, 2004 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-14726475

RESUMO

Recent evidence has identified the peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) as a regulator of cardiac energy metabolism and mitochondrial biogenesis. We describe the development of a transgenic system that permits inducible, cardiac-specific overexpression of PGC-1alpha. Expression of the PGC-1alpha transgene in this system (tet-on PGC-1alpha) is cardiac-specific in the presence of doxycycline (dox) and is not leaky in the absence of dox. Overexpression of PGC-1alpha in tet-on PGC-1alpha mice during the neonatal stages leads to a dramatic increase in cardiac mitochondrial number and size coincident with upregulation of gene markers associated with mitochondrial biogenesis. In contrast, overexpression of PGC-1alpha in the hearts of adult mice leads to a modest increase in mitochondrial number, derangements of mitochondrial ultrastructure, and development of cardiomyopathy. The cardiomyopathy in adult tet-on PGC-1alpha mice is characterized by an increase in ventricular mass and chamber dilatation. Surprisingly, removal of dox and cessation of PGC-1alpha overexpression in adult mice results in complete reversal of cardiac dysfunction within 4 weeks. These results indicate that PGC-1alpha drives mitochondrial biogenesis in a developmental stage-dependent manner permissive during the neonatal period. This unique murine model should prove useful for the study of the molecular regulatory programs governing mitochondrial biogenesis and characterization of the relationship between mitochondrial dysfunction and cardiomyopathy and as a general model of inducible, reversible cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/genética , Regulação da Expressão Gênica no Desenvolvimento , Mitocôndrias Cardíacas/fisiologia , Miócitos Cardíacos/metabolismo , Transativadores/fisiologia , Trifosfato de Adenosina/biossíntese , Fatores Etários , Animais , Animais Recém-Nascidos , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Modelos Animais de Doenças , Doxiciclina/farmacologia , Metabolismo Energético , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Genes Sintéticos , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/ultraestrutura , Cadeias Pesadas de Miosina/genética , Especificidade de Órgãos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/fisiologia , Sequências Reguladoras de Ácido Nucleico/efeitos dos fármacos , Transativadores/biossíntese , Transativadores/genética , Fatores de Transcrição , Transgenes
10.
Pulm Circ ; 5(2): 244-68, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26064450

RESUMO

Increased peripheral resistance of small distal pulmonary arteries is a hallmark signature of pulmonary hypertension (PH) and is believed to be the consequence of enhanced vasoconstriction to agonists, thickening of the arterial wall due to remodeling, and increased thrombosis. The elevation in arterial tone in PH is attributable, at least in part, to smooth muscle cells of PH patients being more depolarized and displaying higher intracellular Ca(2+) levels than cells from normal subjects. It is now clear that downregulation of voltage-dependent K(+) channels (e.g., Kv1.5) and increased expression and activity of voltage-dependent (Cav1.2) and voltage-independent (e.g., canonical and vanilloid transient receptor potential [TRPC and TRPV]) Ca(2+) channels play an important role in the functional remodeling of pulmonary arteries in PH. This review focuses on an anion-permeable channel that is now considered a novel excitatory mechanism in the systemic and pulmonary circulations. It is permeable to Cl(-) and is activated by a rise in intracellular Ca(2+) concentration (Ca(2+)-activated Cl(-) channel, or CaCC). The first section outlines the biophysical and pharmacological properties of the channel and ends with a description of the molecular candidate genes postulated to encode for CaCCs, with particular emphasis on the bestrophin and the newly discovered TMEM16 and anoctamin families of genes. The second section provides a review of the various sources of Ca(2+) activating CaCCs, which include stimulation by mobilization from intracellular Ca(2+) stores and Ca(2+) entry through voltage-dependent and voltage-independent Ca(2+) channels. The third and final section summarizes recent findings that suggest a potentially important role for CaCCs and the gene TMEM16A in PH.

11.
PLoS One ; 4(9): e7172, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19779629

RESUMO

While considerable evidence supports the causal relationship between increases in c-Myc (Myc) and cardiomyopathy as a part of a "fetal re-expression" pattern, the functional role of Myc in mechanisms of cardiomyopathy remains unclear. To address this, we developed a bitransgenic mouse that inducibly expresses Myc under the control of the cardiomyocyte-specific MHC promoter. In adult mice the induction of Myc expression in cardiomyocytes in the heart led to the development of severe hypertrophic cardiomyopathy followed by ventricular dysfunction and ultimately death from congestive heart failure. Mechanistically, following Myc activation, cell cycle markers and other indices of DNA replication were significantly increased suggesting that cell cycle-related events might be a primary mechanism of cardiac dysfunction. Furthermore, pathological alterations at the cellular level included alterations in mitochondrial function with dysregulation of mitochondrial biogenesis and defects in electron transport chain complexes I and III. These data are consistent with the known role of Myc in several different pathways including cell cycle activation, mitochondrial proliferation, and apoptosis, and indicate that Myc activation in cardiomyocytes is an important regulator of downstream pathological sequelae. Moreover, our findings indicate that the induction of Myc in cardiomyocytes is sufficient to cause cardiomyopathy and heart failure, and that sustained induction of Myc, leading to cell cycle re-entry in adult cardiomyocytes, represents a maladaptive response for the mature heart.


Assuntos
Cardiomiopatia Hipertrófica/metabolismo , Insuficiência Cardíaca/metabolismo , Mitocôndrias/patologia , Animais , Apoptose , Ciclo Celular , Proliferação de Células , Transporte de Elétrons , Feminino , Hipertrofia , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Modelos Biológicos , Miócitos Cardíacos/citologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/metabolismo
12.
Cell Commun Adhes ; 15(3): 247-60, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18979294

RESUMO

Yeast two-hybrid analysis (Fields and Song, 1989, Nature, 340:245-246) was used to screen a human heart library to isolate proteins interacting with the adult muscle-specific beta1D integrin but not with beta1A integrin. In addition to previously identified interactions (RACK 1(Liliental and Chang, 1998, Journal of Biological Chemistry, 273:2379-2383) and alpha-actinin (Otey et al., 1990, Journal of Cell Biology, 111:721-729), the authors isolated several novel candidates. These include subunit 3 (CSN3/Sgn3) of the COP9 signalosome complex, cyclins D1, D2, and D3, RanBPM, and a recently identified protein COG8/DOR1. These protein interactions were specific for beta1D integrin, as no binding to beta1A integrin cytoplasmic domain was measurable by two-hybrid analysis. This paper presents the initial characterization of the interaction of CSN3 with beta1D integrin, the localization of CSN3 and the other COP9 signalosome subunits in embryonic and adult cardiac myocytes and their response to muscle cell differentiation.


Assuntos
Núcleo Celular/metabolismo , Matriz Extracelular/metabolismo , Cadeias beta de Integrinas/metabolismo , Integrina beta1/metabolismo , Complexos Multiproteicos/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/fisiologia , Peptídeo Hidrolases/metabolismo , Proteínas Quinases/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Ligação Competitiva , Complexo do Signalossomo COP9 , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Humanos , Camundongos , Microscopia de Fluorescência , Modelos Biológicos , Músculo Esquelético/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Miocárdio/química , Proteínas Nucleares/metabolismo , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína/fisiologia , Proteínas Proto-Oncogênicas , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
13.
Am J Physiol Heart Circ Physiol ; 289(2): H777-84, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15778276

RESUMO

Atrial (ANP) and brain (BNP) natriuretic peptides are hormones of myocardial cell origin. These hormones bind to the natriuretic peptide A receptor (NPRA) throughout the body, stimulating cGMP production and playing a key role in blood pressure control. Because NPRA receptors are present on cardiomyocytes, we hypothesized that natriuretic peptides may have direct autocrine or paracrine effects on cardiomyocytes or adjacent cardiac cells. Because both natriuretic peptides and NPRA gene expression are upregulated in states of pressure overload, we speculated that the effects of the natriuretic peptides on cardiac structure and function would be most apparent after pressure overload. To attenuate cardiomyocyte NPRA activity, transgenic mice with cardiac specific expression of a dominant-negative (DN-NPRA) mutation (HCAT D 893A) in the NPRA receptor were created. Cardiac structure and function were assessed (avertin anesthesia) in the absence and presence of pressure overload produced by suprarenal aortic banding. In the absence of pressure overload, basal and BNP-stimulated guanylyl cyclase activity assessed in cardiac membrane fractions was reduced. However, systolic blood pressure, myocardial cGMP, log plasma ANP levels, and ventricular structure and function were similar in wild-type (WT-NPRA) and DN-NPRA mice. In the presence of pressure overload, myocardial cGMP levels were reduced, and ventricular hypertrophy, fibrosis, filling pressures, and mortality were increased in DN-NPRA compared with WT-NPRA mice. In addition to their hormonal effects, endogenous natriuretic peptides exert physiologically relevant autocrine and paracrine effects via cardiomyocyte NPRA receptors to modulate cardiac hypertrophy and fibrosis in response to pressure overload.


Assuntos
Guanilato Ciclase/metabolismo , Hipertensão/mortalidade , Hipertensão/fisiopatologia , Miocárdio/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Remodelação Ventricular , Animais , Aorta , Fator Natriurético Atrial/sangue , Pressão Sanguínea , GMP Cíclico/metabolismo , Ecocardiografia , Expressão Gênica , Genes Dominantes , Guanilato Ciclase/genética , Ligadura , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Mutação , Receptores do Fator Natriurético Atrial/genética , Transgenes
14.
J Electrocardiol ; 38(4 Suppl): 40-4, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16226072

RESUMO

BACKGROUND: Epicardial potentials reveal the strong effects of fiber anisotropy, rotation, imbrication, and coupling on propagation in the intact heart. From the patterns of the surface potentials, we can obtain information about the local fiber orientation, anisotropy, the transmural fiber rotation, and which direction the wave front is traveling through the wall. In this study, lessons learned from epicardial potential mapping of large hearts were applied to studies conducted in genetically altered mouse hearts. METHODS: An inducible model of the overexpression of a gain-of-function alpha5 integrin (cytoplasmic domain truncation) was created in mouse. After 3 days of administration of doxycycline, the animals exhibited an altered electrical phenotype of markedly reduced amplitude of the QRS complex on the surface electrocardiogram. Epicardial potentials were recorded from Langendorff-perfused mouse hearts with alpha5 integrin gain-of-function mutations and from wild-type (WT) control hearts. A cylindrical electrode array consisting of 184 sites with 1-mm uniform interelectrode spacing was placed around the heart, and unipolar electrograms were recorded during atrial and ventricular stimulation at different basic cycle lengths. RESULTS: The total ventricular activation time for the transgenic animals was greater than that of the WT hearts for atrial and ventricular pacing locations. The isopotential maps from the mutated hearts showed a loss of anisotropy, as revealed by the more rounded and less elliptically shaped wave fronts seen immediately after epicardial point stimulation when compared with WT hearts. The weaker potential maxima in the mutated hearts did not exhibit the normal expansion and rotation associated with an advancing wave front in a normal heart, suggesting abnormalities in myocyte coupling in these hearts. Isopotential maps provided additional information about fiber architecture from the electric field that was not obtained from optical recordings alone. These findings provided a phenotypic characterization and specific insights into the mechanisms of the electrical abnormalities associated with altered integrin signaling in cardiac myocytes.


Assuntos
Sistema de Condução Cardíaco/fisiopatologia , Pericárdio/fisiopatologia , Potenciais de Ação , Animais , Mapeamento Potencial de Superfície Corporal , Estimulação Cardíaca Artificial , Modelos Animais de Doenças , Estimulação Elétrica , Técnicas Eletrofisiológicas Cardíacas , Feminino , Ventrículos do Coração/fisiopatologia , Integrinas/metabolismo , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Fenótipo , Transdução de Sinais
15.
J Card Fail ; 8(4): 262-72, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12397575

RESUMO

BACKGROUND: Integrins are heterodimeric receptors that couple the extracellular matrix to intracellular signaling pathways and the cyoskeleton. Integrins are strain transducers and candidates for modulators or effectors of cardiac hypertrophy. METHODS: To begin to probe this function, we have transgenically expressed a chimeric protein that alters integrin function in cardiomyocytes. The transgene (Tac-beta(1D)) consists of the biologically inert extracellular and transmembrane domain of the interleukin-2 receptor alpha subunit (Tac) fused to the cytoplasmic tail of the human beta(1D) integrin driven by the cardiac alpha-myosin heavy chain promoter. Transgene expression results in a severe, usually fatal, perinatal cardiac phenotype, characterized by initial electrocardiographic abnormalities followed by extensive myocyte loss, macrophage infiltration, and replacement fibrosis. RESULTS: Expression of Tac-beta(1D) resulted in displacement of endogenous beta(1D) integrin from Z-lines and T-tubules, decreased expression of endogenous beta(1D), and disrupted the fibronectin pericellular matrix. These results are consistent with an essential role for beta(1) integrins in maintenance of cardiomyocyte viability and interaction with extracellular matrix. CONCLUSION: The appearance of conduction abnormalities before morphologic changes suggests that integrins are important in the development or maintenance of the conducting system of the heart.


Assuntos
Genes Letais/genética , Integrina beta1/biossíntese , Integrina beta1/genética , Miócitos Cardíacos/metabolismo , Fenótipo , Animais , Animais Recém-Nascidos/genética , Modelos Animais de Doenças , Eletrocardiografia , Matriz Extracelular/genética , Matriz Extracelular/ultraestrutura , Feminino , Fibronectinas/biossíntese , Fibronectinas/genética , Regulação da Expressão Gênica/genética , Átrios do Coração/anormalidades , Átrios do Coração/citologia , Átrios do Coração/ultraestrutura , Sistema de Condução Cardíaco/anormalidades , Sistema de Condução Cardíaco/citologia , Sistema de Condução Cardíaco/fisiopatologia , Ventrículos do Coração/anormalidades , Ventrículos do Coração/citologia , Ventrículos do Coração/ultraestrutura , Masculino , Camundongos/genética , Camundongos Transgênicos/genética , Modelos Cardiovasculares , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Índice de Gravidade de Doença
16.
Am J Physiol Heart Circ Physiol ; 287(5): H2164-72, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15256372

RESUMO

Delayed cardiac relaxation in failing hearts has been attributed to reduced activity and/or expression of sarco(endo)plasmic reticulum Ca2+-ATPase 2a (SERCA2a). Although constitutive overexpression of SERCA2a has proven effective in preventing cardiac dysfunction, it is unclear whether increasing SERCA2a expression in hearts with preexisting hypertrophy will be therapeutic. To test this hypothesis, we generated a binary transgenic (BTG) system that allows tetracycline-inducible, cardiac-specific SERCA2a expression. In this system (tet-on SERCA2a), a FLAG-tagged SERCA2a transgene is expressed in the presence of doxycycline (Dox) but not in the absence of Dox (2.3-fold more mRNA, 45% more SERCA2a protein). Calcium transients measured in isolated cardiac myocytes from nonbanded Dox-treated BTG mice showed an accelerated calcium decline and an increased systolic Ca2+ peak. Sarcoplasmic reticulum (SR) calcium loading was increased by 45% in BTG mice. In the presence of pressure overload (aortic banding), echocardiographic analysis revealed that expression of SERCA2a-FLAG caused an improvement in fractional shortening. SERCA2a-FLAG expression alleviated the resultant cardiac dysfunction. This was illustrated by an increase in the rate of decline of the calcium transient. Cell shortening and SR calcium loading were also improved in cardiac myocytes isolated from banded BTG mice after SERCA2a overexpression. In conclusion, we generated a novel transgenic mouse that conditionally overexpresses SERCA2a. This model is suitable for both long- and short-term studies of the effects of controlled SERCA2a expression on cardiac function. In addition, inducible overexpression of SERCA2a improved cardiac function and calcium handling in mice with established contractile dysfunction.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Cálcio/metabolismo , Cardiomegalia/fisiopatologia , Doxiciclina/farmacologia , Coração/fisiopatologia , Hipertensão/complicações , Animais , Proteínas de Ligação ao Cálcio/metabolismo , ATPases Transportadoras de Cálcio/genética , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Expressão Gênica , Camundongos , Camundongos Transgênicos , Contração Miocárdica , Miocárdio/metabolismo , Ratos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA