Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 327(4): H1112-H1118, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39269449

RESUMEN

Reduced muscle contractility and mitochondrial bioenergetics are the hallmarks of systolic heart failure. There is currently no therapy targeting both. Here, we show that gene delivery of Perm1 via adeno-associated virus (AAV) simultaneously enhances cardiac contractility and mitochondrial biogenesis in C57BL6 mice. Moreover, we found that PERM1 interacts with troponin C (TnC), a key contractile protein in striated muscle, and that AAV-Perm1 led to the upregulation of TnC. This study suggests that gene delivery of Perm1 may be a novel therapeutic approach to treat systolic heart failure by simultaneously restoring cardiac contractility and mitochondrial bioenergetics.NEW & NOTEWORTHY Perm1 gene delivered with AAV9 enhances cardiac contractility in mice, and it is concomitant with the increase of mitochondrial bioenergetics and upregulation of TnC. This is the first study showing that PERM1, previously known as a striated muscle-specific mitochondrial regulator, also positively regulates cardiac contractility.


Asunto(s)
Dependovirus , Ratones Endogámicos C57BL , Mitocondrias Cardíacas , Contracción Miocárdica , Animales , Dependovirus/genética , Mitocondrias Cardíacas/metabolismo , Terapia Genética/métodos , Técnicas de Transferencia de Gen , Ratones , Masculino , Vectores Genéticos , Metabolismo Energético , Miocitos Cardíacos/metabolismo , Insuficiencia Cardíaca Sistólica/fisiopatología , Insuficiencia Cardíaca Sistólica/genética , Insuficiencia Cardíaca Sistólica/metabolismo , Insuficiencia Cardíaca Sistólica/terapia
2.
Cardiovasc Res ; 118(1): 53-64, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33620071

RESUMEN

It is well established that the vasculature plays a crucial role in maintaining oxygen and nutrients supply to the heart. Increasing evidence further suggests that the microcirculation has additional roles in supporting a healthy microenvironment. Heart failure is well known to be associated with changes and functional impairment of the microvasculature. The specific ablation of protective signals in endothelial cells in experimental models is sufficient to induce heart failure. Therefore, restoring a healthy endothelium and microcirculation may be a valuable therapeutic strategy to treat heart failure. This review article will summarize the current understanding of the vascular contribution to heart failure with reduced or preserved ejection fraction. Novel therapeutic approaches including next generation pro-angiogenic therapies and non-coding RNA therapeutics, as well as the targeting of metabolites or metabolic signalling, vascular inflammation and senescence will be discussed.


Asunto(s)
Inductores de la Angiogénesis/uso terapéutico , Vasos Coronarios/efectos de los fármacos , Terapia Genética , Insuficiencia Cardíaca Diastólica/terapia , Insuficiencia Cardíaca Sistólica/terapia , Microvasos/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Vacunas/uso terapéutico , Inductores de la Angiogénesis/efectos adversos , Animales , Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , Terapia Genética/efectos adversos , Insuficiencia Cardíaca Diastólica/genética , Insuficiencia Cardíaca Diastólica/metabolismo , Insuficiencia Cardíaca Diastólica/fisiopatología , Insuficiencia Cardíaca Sistólica/genética , Insuficiencia Cardíaca Sistólica/metabolismo , Insuficiencia Cardíaca Sistólica/fisiopatología , Humanos , Microcirculación/efectos de los fármacos , Microvasos/metabolismo , Microvasos/fisiopatología , ARN no Traducido/genética , ARN no Traducido/metabolismo , Recuperación de la Función , Vacunas/efectos adversos , Función Ventricular Izquierda/efectos de los fármacos
3.
Gene ; 726: 144136, 2020 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-31629817

RESUMEN

Chronic systolic heart failure (CSHF) was a complex syndrome. Recently, vagus nerve stimulation (VNS), a novel treatment method, has emerged for the treatment of CSHF. therefore the aim of this study was to explore the possible mechanism of VNS treatment alleviating CSHF in rats. Firstly, we found after VNS treatment for 72 h, the level of B-type natriuretic peptide in VNS group was lower than that in CSHF group. In addition, VNS treatment induced the elevated left ventricular ejection fraction level, reduced left ventricular end diastolic volume and left ventricular end systolic volume level in VNS group, suggesting a mitigation of CSHF by VNS. Then we found the level of miR-183-3p in CSHF group was much lower than that in VNS group by High-throughput sequencing. The further results indicated that Bcl-2 interacting protein 3 like (BNIP3L) was identified as the target gene of miR-183-3p, and the expression of BNIP3L was notably reduced in rats of VNS group compared with CSHF group. Moreover, the down-regulated expression of miR-183-3p increased BNIP3L-mediated autophagy in rats of CSHF group compared with VNS group. Further mechanism findings demonstrated that up-regulation of miR-183-3p reduced the expression of BNIP3L, while down-regulation of miR-183-3p facilitated the expression of BNIP3L in H9c2 cells. miR-183-3p could also regulate autophagy by targeting BNIP3L in vitro, which was manifested by overexpression of miR-183-3p to inhibit BNIP3L-mediated autophagy. Our data demonstrated that VNS treatment benefited CSHF via the up-regulation of miRNA-183-3p, which reduced the BNIP3L-mediated autophagy, providing a new therapeutic direction for CSHF.


Asunto(s)
Autofagia/genética , Insuficiencia Cardíaca Sistólica/genética , Proteínas de la Membrana/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas/genética , Regulación hacia Arriba/genética , Animales , Regulación hacia Abajo/genética , Masculino , Ratas , Ratas Wistar , Volumen Sistólico/genética , Activación Transcripcional/genética , Estimulación del Nervio Vago/métodos , Función Ventricular Izquierda/genética
4.
Sci Rep ; 7: 39335, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28045017

RESUMEN

Adhesive intercellular connections at cardiomyocyte intercalated disks (IDs) support contractile force and maintain structural integrity of the heart muscle. Disturbances of the proteins at IDs deteriorate cardiac function and morphology. An adaptor protein afadin, one of the components of adherens junctions, is expressed ubiquitously including IDs. At present, the precise role of afadin in cardiac physiology or disease is unknown. To explore this, we generated conditional knockout (cKO) mice with cardiomyocyte-targeted deletion of afadin. Afadin cKO mice were born according to the expected Mendelian ratio and have no detectable changes in cardiac phenotype. On the other hand, chronic pressure overload induced by transverse aortic constriction (TAC) caused systolic dysfunction, enhanced fibrogenesis and apoptosis in afadin cKO mice. Afadin deletion increased macrophage infiltration and monocyte chemoattractant protein-1 expression, and suppressed transforming growth factor (TGF) ß receptor signaling early after TAC procedure. Afadin also associated with TGFß receptor I at IDs. Pharmacological antagonist of TGFß receptor I (SB431542) augmented mononuclear infiltration and fibrosis in the hearts of TAC-operated control mice. In conclusion, afadin is a critical molecule for cardiac protection against chronic pressure overload. The beneficial effects are likely to be a result from modulation of TGFß receptor signaling pathways by afadin.


Asunto(s)
Aorta/fisiología , Insuficiencia Cardíaca Sistólica/genética , Uniones Intercelulares/metabolismo , Macrófagos/inmunología , Proteínas de Microfilamentos/metabolismo , Miocardio/patología , Miocitos Cardíacos/fisiología , Animales , Apoptosis/genética , Movimiento Celular , Quimiocina CCL2/metabolismo , Constricción Patológica , Fibrosis , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Contracción Miocárdica , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
5.
BMC Endocr Disord ; 15: 36, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26215257

RESUMEN

BACKGROUND: The pathogenesis of diabetic cardiomyopathy (DCM) involves the enhanced activation of peroxisome proliferator activating receptor (PPAR) transcription factors, including the most prominent isoform in the heart, PPARα. In cancer cells and adipocytes, post-translational modification of PPARs have been identified, including ligand-dependent degradation of PPARs by specific ubiquitin ligases. However, the regulation of PPARs in cardiomyocytes and heart have not previously been identified. We recently identified that muscle ring finger-1 (MuRF1) and MuRF2 differentially inhibit PPAR activities by mono-ubiquitination, leading to the hypothesis that MuRF3 may regulate PPAR activity in vivo to regulate DCM. METHODS: MuRF3-/- mice were challenged with 26 weeks 60% high fat diet to induce insulin resistance and DCM. Conscious echocardiography, blood glucose, tissue triglyceride, glycogen levels, immunoblot analysis of intracellular signaling, heart and skeletal muscle morphometrics, and PPARα, PPARß, and PPARγ1 activities were assayed. RESULTS: MuRF3-/- mice exhibited a premature systolic heart failure by 6 weeks high fat diet (vs. 12 weeks in MuRF3+/+). MuRF3-/- mice weighed significantly less than sibling-matched wildtype mice after 26 weeks HFD. These differences may be largely due to resistance to fat accumulation, as MRI analysis revealed MuRF3-/- mice had significantly less fat mass, but not lean body mass. In vitro ubiquitination assays identified MuRF3 mono-ubiquitinated PPARα and PPARγ1, but not PPARß. CONCLUSIONS: These findings suggest that MuRF3 helps stabilize cardiac PPARα and PPARγ1 in vivo to support resistance to the development of DCM. MuRF3 also plays an unexpected role in regulating fat storage despite being found only in striated muscle.


Asunto(s)
Cardiomiopatías Diabéticas/genética , Dieta Alta en Grasa/efectos adversos , Insuficiencia Cardíaca Sistólica/genética , Proteínas Musculares/genética , Miocitos Cardíacos/metabolismo , Tejido Adiposo , Animales , Composición Corporal , Peso Corporal , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Insuficiencia Cardíaca Sistólica/etiología , Insuficiencia Cardíaca Sistólica/metabolismo , Técnicas In Vitro , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , PPAR alfa/metabolismo , PPAR gamma/metabolismo , PPAR-beta/metabolismo , Ubiquitinación
6.
PLoS One ; 10(5): e0128105, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26023924

RESUMEN

GATA4 and GATA6 are central cardiac transcriptional regulators. The postnatal, stage-specific function of the cardiac transcription factors GATA4 and GATA6 have not been evaluated. In part, this is because current Cre-loxP approaches to cardiac gene inactivation require time consuming and costly breeding of Cre-expressing and "floxed" mouse lines, often with limited control of the extent or timing of gene inactivation. We investigated the stage-specific functions of GATA4 and GATA6 in the postnatal heart by using adeno-associated virus serotype 9 to control the timing and extent of gene inactivation by Cre. Systemic delivery of recombinant, adeno-associated virus 9 (AAV9) expressing Cre from the cardiac specific Tnnt2 promoter was well tolerated and selectively and efficiently recombined floxed target genes in cardiomyocytes. AAV9:Tnnt2-Cre efficiently inactivated Gata4 and Gata6. Neonatal Gata4/6 inactivation caused severe, rapidly lethal systolic heart failure. In contrast, Gata4/6 inactivation in adult heart caused only mild systolic dysfunction but severe diastolic dysfunction. Reducing the dose of AAV9:Tnnt2-Cre generated mosaics in which scattered cardiomyocytes lacked Gata4/6. This mosaic knockout revealed that Gata4/6 are required cell autonomously for physiological cardiomyocyte growth. Our results define novel roles of GATA4 and GATA6 in the neonatal and adult heart. Furthermore, our data demonstrate that evaluation of gene function hinges on controlling the timing and extent of gene inactivation. AAV9:Tnnt2-Cre is a powerful tool for controlling these parameters.


Asunto(s)
Dependovirus/genética , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/metabolismo , Corazón/crecimiento & desarrollo , Integrasas/genética , Miocitos Cardíacos/fisiología , Animales , Animales Recién Nacidos , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Vectores Genéticos , Corazón/fisiología , Insuficiencia Cardíaca Sistólica/genética , Integrasas/administración & dosificación , Ratones Transgénicos
7.
Kardiologiia ; 55(9): 25-30, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-26922818

RESUMEN

AIM: to elucidate association between polymorphic markers of interleukin-6 (Il-6) and tumor necrosis factor (TNF) genes and unfavorable outcomes in patients with chronic heart failure (CHF). MATERIAL AND METHODS: We determined levels of TNF and Il-6 and genotypes of polymorphic markers G(-238)A of TNF gene (rs361525) and G(--174)C of IL-6 gene (rs1800795) in 151 patients (mean age 64.5 years) hospitalized because of decompensation of systolic CHF (left ventricular ejection fraction ≤ 40%) after stabilization of their state. Unfavorable outcomes were registered during follow-up for 2 years. RESULTS: Mean levels of NT-proBNP, Il-6, and TNF were 2481.1 ± 199.86 fmol/ml, 21.8 7.46 rg/ml, and 10.07 ± 0.65 rg/ml, respectively. 138 (94.4%), 13 (8.6%) and 0 patients were carriers of genotypes GG, AG, and AA of polymorphic marker G(-238)A of TNF gene, respectively; 54 (35.8%), 69 (45.7%), and 28 (18.5%) patients carried genotypes GG, GC, and CC of polymorphic marker G(-174)C gene IL-6, respectively. There was no association between Il-6, TNF levels and carriage of either of genotypes as well as unfavorable clinical course of CHF. Mean survival time before repetitive episode of CHF decompensation (including lethal one) was significantly shorter among carriers of A allele compared with carriers of G allele of polymorphic marker G(-238)A of TNF gene (243 ± 97.7 and 947 ± 78 days, respectively, p = 0.018). Mean time before all cause death was also shorter in carriers of A compared with carriers of G allele (289 ± 122.9 and 1039 ± 73.3 days, respectively, p = 0.03). The studied polymorphism of IL-6 gene had no prognostic value. CONCLUSION: We obtained data on association between carriage of A allele of polymorphic marker G(-238)A of TNF gene and unfavorable prognosis in patients with CHF and inpraired left ventricular systolic function.


Asunto(s)
ADN/genética , Insuficiencia Cardíaca Sistólica/genética , Polimorfismo Genético , Factor de Necrosis Tumoral alfa/genética , Alelos , Femenino , Insuficiencia Cardíaca Sistólica/metabolismo , Insuficiencia Cardíaca Sistólica/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo , Federación de Rusia/epidemiología , Tasa de Supervivencia/tendencias , Factor de Necrosis Tumoral alfa/metabolismo
8.
PLoS One ; 8(7): e68154, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874529

RESUMEN

Matrix metalloproteinase-2 (MMP-2) is increasingly recognized as a major contributor to progressive cardiac injury within the setting of ischemia-reperfusion injury and ischemic ventricular remodeling. A common feature of these conditions is an increase in oxidative stress, a process that engages multiple pro-inflammatory and innate immunity cascades. We recently reported on the identification and characterization of an intracellular isoform of MMP-2 generated by oxidative stress-mediated activation of an alternative promoter located within the first intron of the MMP-2 gene. Transcription from this site generates an N-terminal truncated 65 kDa isoform of MMP-2 (NTT-MMP-2) that lacks the secretory sequence and the inhibitory prodomain region. The NTT-MMP-2 isoform is intracellular, enzymatically active and localizes in part to mitochondria. Expression of the NTT-MMP-2 isoform triggers Nuclear Factor of Activated T-cell (NFAT) and NF-κB signaling with the expression of a highly defined innate immunity transcriptome, including Interleukin-6, MCP-1, IRF-7 and pro-apoptotic transcripts. To determine the functional significance of the NTT-MMP-2 isoform in vivo we generated cardiac-specific NTT-MMP-2 transgenic mice. These mice developed progressive cardiomyocyte and ventricular hypertrophy associated with systolic heart failure. Further, there was evidence for cardiomyocyte apoptosis and myocardial infiltration with mononuclear cells. The NTT-MMP-2 transgenic hearts also demonstrated more severe injury following ex vivo ischemia-reperfusion injury. We conclude that a novel intracellular MMP-2 isoform induced by oxidant stress directly contributes, in the absence of superimposed injury, to cardiomyocyte hypertrophy. inflammation, systolic heart failure and enhanced susceptibility to ischemia-reperfusion injury.


Asunto(s)
Insuficiencia Cardíaca Sistólica/enzimología , Inflamación/enzimología , Metaloproteinasa 2 de la Matriz/metabolismo , Miocitos Cardíacos/patología , Animales , Insuficiencia Cardíaca Sistólica/genética , Inflamación/genética , Factor 7 Regulador del Interferón/metabolismo , Interleucina-6/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Ratones , Ratones Transgénicos , Isoformas de Proteínas/metabolismo
9.
Biomed Res Int ; 2013: 283856, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23710440

RESUMEN

Matrix metalloproteinases (MMPs) are involved in the remodeling of extracellular matrix in various tissues. Their functioning could be related to the formation of complexes, containing MMP9, MMP2, tissue inhibitor of metalloproteinases type 1 (TIMP1), and neutrophil gelatinase-associated lipocalin (NGAL). Such complexes have not been investigated in either myocardial or skeletal muscles. We examined 20 male pigs with heart failure (HF), and 5 sham-operated animals. There were no differences in the mRNA expression of MMP9, MMP2, TIMP1, and NGAL between diseased and healthy animals, in either left ventricle (LV) myocardium or skeletal muscles. In LV from both diseased and healthy animals, in nonreducing and nondenaturing conditions, we demonstrated the presence of high molecular weight (HMW) complexes (130, 170, and 220 kDa) containing MMP9, TIMP1, and NGAL (also MMP2 in 220 kDa complex) without proteolytic activity, and a proteolytically active 115 kDa MMP9 form together with 72 and 68 kDa bands (proMMP2 and MMP2). Proteolytically active bands were also spontaneously released from HMW complexes. In skeletal muscles from both diseased and healthy animals, in nonreducing and nondenaturing conditions, we found no HMW complexes, and proteolytic activity was associated with the presence of 72 and 68 kDa bands (proMMP2 and MMP2).


Asunto(s)
Insuficiencia Cardíaca Sistólica/genética , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Proteínas de Fase Aguda/biosíntesis , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Insuficiencia Cardíaca Sistólica/complicaciones , Insuficiencia Cardíaca Sistólica/metabolismo , Insuficiencia Cardíaca Sistólica/patología , Humanos , Lipocalina 2 , Lipocalinas/biosíntesis , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Músculo Esquelético/patología , Miocardio/patología , Proteínas Proto-Oncogénicas/biosíntesis , Porcinos/genética , Porcinos/metabolismo , Taquicardia/complicaciones , Taquicardia/metabolismo , Taquicardia/patología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
10.
Arq. bras. cardiol ; Arq. bras. cardiol;98(5): 384-389, maio 2012. tab
Artículo en Portugués | LILACS | ID: lil-643645

RESUMEN

FUNDAMENTO: O sistema nervoso simpático apresenta grande importância na patogênese da fibrilação atrial na insuficiência cardíaca sistólica. A identificação de polimorfismos no gene ADBR1 do receptor beta1-adrenérgico representa um importante passo no conhecimento dessa patogênese. OBJETIVO: Este estudo analisou a associação entre os dois polimorfismos funcionais do gene ADBR1 do receptor beta1-adrenérgico, Ser49Gly e Arg389Gly, e a presença da fibrilação atrial em pacientes com insuficiência cardíaca sistólica. MÉTODOS: Estudo caso-controle com 144 pacientes portadores de insuficiência cardíaca sistólica, dos quais 24 com fibrilação atrial (casos) e 120 sem fibrilação atrial (controles). O DNA genômico foi extraído de leucócitos do sangue periférico e os genótipos dos polimorfismos Ser49Gly e Arg389Gly foram identificados em todos os indivíduos por PCR/RFLP (polymerase chain reaction / restriction fragment length polymorphism). RESULTADOS: A média etária foi 59 ± 13 anos, 70% dos pacientes eram do sexo masculino, 42% apresentavam causa isquêmica e 74% apresentavam hipertensão arterial sistêmica. Os genótipos Ser49Ser e Arg389Arg apresentaram associação significativa com fibrilação atrial (p = 0,005 e p = 0,01; respectivamente). Por meio de regressão logística, ambos ajustados para o tamanho do átrio esquerdo e idade, mantiveram associação significativa (Arg389Arg - odds ratios: 2,78; intervalo de confiança de 95% = 1,02 - 7,56 e Ser49Ser - odds ratios: 8,02; intervalo de confiança de 95% = 1,02 - 63,82). CONCLUSÃO: Ambos os genótipos associaram-se com fibrilação atrial nos pacientes estudados, porém apenas o polimorfismo Ser49Gly apresentava-se em equilíbrio de Hardy-Weinberg.


BACKGROUND: The sympathetic nervous system is of great importance in the pathogenesis of atrial fibrillation in systolic heart failure. The identification of polymorphisms in the beta1-adrenergic receptor gene (ADBR1) represents an important step in understanding this pathogenesis. OBJECTIVE: This study assessed the association between the two functional polymorphisms of the beta1-adrenergic receptor gene (ADBR1), Ser49Gly and Arg389Gly, and the presence of atrial fibrillation in patients with systolic heart failure. METHODS: Case-control study with 144 patients with systolic heart failure, including 24 with atrial fibrillation (cases) and 120 without atrial fibrillation (controls). Genomic DNA was extracted from peripheral blood leukocytes and the genotypes of Ser49Gly and Arg389Gly polymorphisms were identified in all individuals by PCR/RFLP (polymerase chain reaction / restriction fragment length polymorphism). RESULTS: Mean age was 59 ± 13 years, 70% of patients were males, 42% had ischemic causes and 74% had hypertension. Genotypes Ser49Ser and Arg389Arg were significantly associated with atrial fibrillation (p = 0.005 and p = 0.01, respectively). After logistic regression, both adjusted for left atrial size and age, the significant association persisted (Arg389Arg - odds ratios: 2.78, 95% confidence interval = 1.02 to 7.56 and Ser49Ser - odds ratios: 8.02, 95% confidence interval = 1.02 to 63.82). CONCLUSION: Both genotypes were associated with atrial fibrillation in patients; however, only Ser49Gly polymorphism was is in Hardy-Weinberg equilibrium.


Asunto(s)
Anciano , Femenino , Humanos , Persona de Mediana Edad , Fibrilación Atrial/genética , Insuficiencia Cardíaca Sistólica/genética , Polimorfismo Genético/genética , Receptores Adrenérgicos beta 1/genética , Factores de Edad , Estudios de Casos y Controles , Intervalos de Confianza , Genotipo , Modelos Logísticos , Reacción en Cadena de la Polimerasa
11.
Am J Physiol Heart Circ Physiol ; 301(1): H253-60, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21536850

RESUMEN

The Cre-loxP system is a useful tool to study the physiological effects of gene knockout in the heart. One limitation with using this system in the heart is the toxic effect of chronic expression of the Cre recombinase. To circumvent this limitation, a widely used inducible cardiac-specific model, Myh6-MerCreMer (Cre), using tamoxifen (TAM) to activate Cre has been developed. The current study examined cardiac function in Cre-positive C57B/J6 mice exposed to one, three, or five daily doses of a 40 mg/kg TAM to induce Cre activity specifically in the heart. Echocardiography demonstrated no statistically significant differences in systolic function (SF) at baseline as assessed by fractional shortening. In mice exposed to five injections, a significant fall in all determinants of SF was observed 6 days after TAM was initiated. However, SF returned to baseline levels 10 days after TAM initiation although the hearts exhibited significant hypertrophy. Heart weight-to-tibia length ratios were 73 ± 3, 78.5 ± 6, and 87.6 ± 9 mg/cm for one, three, and five TAM injections, respectively. TAM had no effect on cardiac function or hypertrophy in Cre-negative mice. Cre-positive mice receiving five TAM injections had significant reductions in cardiac mitochondrial ATP and significant reductions in the expression of proteins important for the regulation of cardiac oxidative phosphorylation including peroxisome proliferator-activated receptor-γ coactivator-1α and pyruvate dehydrogenase kinase-4. Thus inducible cardiac-specific activation of Cre recombinase caused a transient decline in SF that was dependent on the number of TAM doses and associated with significant hypertrophy and alterations in mitochondrial ATP and important proteins involved in the regulation of cardiac oxidative phosphorylation.


Asunto(s)
Antagonistas de Estrógenos/farmacología , Insuficiencia Cardíaca Sistólica/genética , Insuficiencia Cardíaca Sistólica/fisiopatología , Integrasas/genética , Tamoxifeno/farmacología , Adenosina Trifosfato/metabolismo , Animales , Western Blotting , Citoesqueleto/metabolismo , Ecocardiografía , Metabolismo Energético/fisiología , Activación Enzimática/genética , Activación Enzimática/fisiología , Femenino , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Masculino , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocardio/citología , Miocardio/metabolismo , Cadenas Pesadas de Miosina/genética , Fosforilación Oxidativa/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Sístole/fisiología , Factores de Transcripción/metabolismo , Tubulina (Proteína)/metabolismo , Función Ventricular Izquierda/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA