Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cardiovasc Diabetol ; 22(1): 294, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37891673

RESUMEN

BACKGROUND: The PI3K/AKT pathway transduces the majority of the metabolic actions of insulin. In addition to cytosolic targets, insulin-stimulated phospho-AKT also translocates to mitochondria in the myocardium. Mouse models of diabetes exhibit impaired mitochondrial AKT signaling but the implications of this on cardiac structure and function is unknown. We hypothesized that loss of mitochondrial AKT signaling is a critical step in cardiomyopathy and reduces cardiac oxidative phosphorylation. METHODS: To focus our investigation on the pathophysiological consequences of this mitochondrial signaling pathway, we generated transgenic mouse models of cardiac-specific, mitochondria-targeting, dominant negative AKT1 (CAMDAKT) and constitutively active AKT1 expression (CAMCAKT). Myocardial structure and function were examined using echocardiography, histology, and biochemical assays. We further investigated the underlying effects of mitochondrial AKT1 on mitochondrial structure and function, its interaction with ATP synthase, and explored in vivo metabolism beyond the heart. RESULTS: Upon induction of dominant negative mitochondrial AKT1, CAMDAKT mice developed cardiac fibrosis accompanied by left ventricular hypertrophy and dysfunction. Cardiac mitochondrial oxidative phosphorylation efficiency and ATP content were reduced, mitochondrial cristae structure was lost, and ATP synthase structure was compromised. Conversely, CAMCAKT mice were protected against development of diabetic cardiomyopathy when challenged with a high calorie diet. Activation of mitochondrial AKT1 protected cardiac function and increased fatty acid uptake in myocardium. In addition, total energy expenditure was increased in CAMCAKT mice, accompanied by reduced adiposity and reduced development of fatty liver. CONCLUSION: CAMDAKT mice modeled the effects of impaired mitochondrial signaling which occurs in the diabetic myocardium. Disruption of this pathway is a key step in the development of cardiomyopathy. Activation of mitochondrial AKT1 in CAMCAKT had a protective role against diabetic cardiomyopathy as well as improved metabolism beyond the heart.


Asunto(s)
Diabetes Mellitus , Cardiomiopatías Diabéticas , Proteínas Proto-Oncogénicas c-akt , Animales , Ratones , Adenosina Trifosfato/metabolismo , Diabetes Mellitus/metabolismo , Cardiomiopatías Diabéticas/diagnóstico por imagen , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Metabolismo Energético , Insulina/farmacología , Ratones Transgénicos , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Sci Rep ; 13(1): 18239, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880325

RESUMEN

Electronic cigarettes (e-cigarettes) are a prevalent alternative to conventional nicotine cigarettes among smokers and people who have never smoked. Increased concentrations of serum free fatty acids (FFAs) are crucial in generating lipotoxicity. We studied the effects of acipimox, an antilipolytic drug, on e-cigarette-induced cardiac dysfunction. C57BL/6J wild-type mice on high fat diet were treated with saline, e-cigarette with 2.4% nicotine [e-cigarette (2.4%)], and e-cigarette (2.4%) plus acipimox for 12 weeks. Fractional shortening and ejection fraction were diminished in mice exposed to e-cigarettes (2.4%) compared with saline and acipimox-treated mice. Mice exposed to e-cigarette (2.4%) had increased circulating levels of inflammatory cytokines and FFAs, which were diminished by acipimox. Gene Set Enrichment Analysis revealed that e-cigarette (2.4%)-treated mice had gene expression changes in the G2/M DNA damage checkpoint pathway that was normalized by acipimox. Accordingly, we showed that acipimox suppressed the nuclear localization of phospho-p53 induced by e-cigarette (2.4%). Additionally, e-cigarette (2.4%) increased the apurinic/apyrimidinic sites, a marker of oxidative DNA damage which was normalized by acipimox. Mice exposed to e-cigarette (2.4%) had increased cardiac Heme oxygenase 1 protein levels and 4-hydroxynonenal (4-HNE). These markers of oxidative stress were decreased by acipimox. Therefore, inhibiting lipolysis with acipimox normalizes the physiological changes induced by e-cigarettes and the associated increase in inflammatory cytokines, oxidative stress, and DNA damage.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Humanos , Ratones , Animales , Nicotina , Lipólisis , Ratones Endogámicos C57BL , Fenotipo , Citocinas
3.
Front Cardiovasc Med ; 9: 879726, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35463745

RESUMEN

Electronic cigarettes or e-cigarettes are the most frequently used tobacco product among adolescents. Despite the widespread use of e-cigarettes and the known detrimental cardiac consequences of nicotine, the effects of e-cigarettes on the cardiovascular system are not well-known. Several in vitro and in vivo studies delineating the mechanisms of the impact of e-cigarettes on the cardiovascular system have been published. These include mechanisms associated with nicotine or other components of the aerosol or thermal degradation products of e-cigarettes. The increased hyperlipidemia, sympathetic dominance, endothelial dysfunction, DNA damage, and macrophage activation are prominent effects of e-cigarettes. Additionally, oxidative stress and inflammation are unifying mechanisms at many levels of the cardiovascular impairment induced by e-cigarette exposure. This review outlines the contribution of e-cigarettes in the development of cardiovascular diseases and their molecular underpinnings.

4.
PLoS One ; 15(10): e0239671, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33002059

RESUMEN

In spite of the widespread use of electronic cigarettes, also known as e-cigarettes, and the proposed adverse cardiac effects of nicotine, the detrimental effects of e-cigarettes on the heart are not well known. This study examines the detrimental effects of e-cigarettes with nicotine at doses that yield circulating nicotine and cotinine in the ranges similar to the levels found in habitual smokers, and a high fat diet (HFD) on cardiac structure and function in a commonly used model of diet-induced obesity (DIO). C57BL/6J mice on an HFD were exposed to e-cigarette in the presence (2.4% nicotine) or absence (0% nicotine) of nicotine and saline aerosol for 12 weeks. Echocardiographic data demonstrated a decrease in left ventricular (LV) fractional shortening, LV ejection fraction, and velocity of circumferential fiber shortening (VCF) in mice treated with e-cigarette (2.4% nicotine) compared to e-cigarette (0% nicotine) or saline exposed mice. Cardiomyocytes (CMs) of mice treated with e-cigarette (2.4% nicotine) exhibited LV abnormalities, including lipid accumulation (ventricular steatosis), myofibrillar derangement and destruction, and mitochondrial hypertrophy, as revealed by transmission electron microscopy. The detrimental effects of e-cigarettes (2.4% nicotine) on cardiac structure and function was accompanied by increased oxidative stress, plasma free fatty acid levels, CM apoptosis, and inactivation of AMP-activated protein kinase and activation of its downstream target, acetyl-CoA-carboxylase. Our results indicate profound adverse effects of e-cigarettes (2.4% nicotine) on the heart in obese mice and raise questions about the safety of the nicotine e-cigarettes use.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Sistemas Electrónicos de Liberación de Nicotina , Corazón/efectos de los fármacos , Ratones Obesos , Miocardio/patología , Fumar/efectos adversos , Animales , Cotinina/sangre , Ecocardiografía , Ácidos Grasos no Esterificados/sangre , Corazón/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Miocardio/ultraestructura , Nicotina/efectos adversos , Nicotina/sangre , Estrés Oxidativo/efectos de los fármacos , Disfunción Ventricular Izquierda/inducido químicamente
5.
Am J Physiol Heart Circ Physiol ; 317(2): H445-H459, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31172811

RESUMEN

Electronic cigarettes (e-cigarettes), also known as electronic nicotine delivery systems, are a popular alternative to conventional nicotine cigarettes, both among smokers and those who have never smoked. In spite of the widespread use of e-cigarettes and the proposed detrimental cardiac and atherosclerotic effects of nicotine, the effects of e-cigarettes on these systems are not known. In this study, we investigated the cardiovascular and cardiac effects of e-cigarettes with and without nicotine in apolipoprotein-E knockout (ApoE-/-) mice. We developed an e-cigarette exposure model that delivers nicotine in a manner similar to that of human e-cigarettes users. Using commercially available e-cigarettes, bluCig PLUS, ApoE-/- mice were exposed to saline, e-cigarette without nicotine [e-cigarette (0%)], and e-cigarette with 2.4% nicotine [e-cigarette (2.4%)] aerosol for 12 wk. Echocardiographic data show that mice treated with e-cigarette (2.4%) had decreased left ventricular fractional shortening and ejection fraction compared with e-cigarette (0%) and saline. Ventricular transcriptomic analysis revealed changes in genes associated with metabolism, circadian rhythm, and inflammation in e-cigarette (2.4%)-treated ApoE-/- mice. Transmission electron microscopy revealed that cardiomyocytes of mice treated with e-cigarette (2.4%) exhibited ultrastructural abnormalities indicative of cardiomyopathy. Additionally, we observed increased oxidative stress and mitochondrial DNA mutations in mice treated with e-cigarette (2.4%). ApoE-/- mice on e-cigarette (2.4%) had also increased atherosclerotic lesions compared with saline aerosol-treated mice. These results demonstrate adverse effects of e-cigarettes on cardiac function in mice.NEW & NOTEWORTHY The present study is the first to show that mice exposed to nicotine electronic cigarettes (e-cigarettes) have decreased cardiac fractional shortening and ejection fraction in comparison with controls. RNA-seq analysis reveals a proinflammatory phenotype induced by e-cigarettes with nicotine. We also found increased atherosclerosis in the aortic root of mice treated with e-cigarettes with nicotine. Our results show that e-cigarettes with nicotine lead to detrimental effects on the heart that should serve as a warning to e-cigarette users and agencies that regulate them.


Asunto(s)
Aterosclerosis/etiología , Sistemas Electrónicos de Liberación de Nicotina , Nicotina/toxicidad , Agonistas Nicotínicos/toxicidad , Volumen Sistólico , Vapeo/efectos adversos , Disfunción Ventricular Izquierda/etiología , Función Ventricular Izquierda , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Modelos Animales de Enfermedad , Exposición por Inhalación/efectos adversos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Estrés Oxidativo , Placa Aterosclerótica , Especies Reactivas de Oxígeno/metabolismo , Transcriptoma , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
6.
J Am Heart Assoc ; 4(12)2015 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-26702077

RESUMEN

BACKGROUND: Duchenne muscular dystrophy is a fatal cardiac and skeletal muscle disease resulting from mutations in the dystrophin gene. We have previously demonstrated that a dystrophin-associated protein, sarcospan (SSPN), ameliorated Duchenne muscular dystrophy skeletal muscle degeneration by activating compensatory pathways that regulate muscle cell adhesion (laminin-binding) to the extracellular matrix. Conversely, loss of SSPN destabilized skeletal muscle adhesion, hampered muscle regeneration, and reduced force properties. Given the importance of SSPN to skeletal muscle, we investigated the consequences of SSPN ablation in cardiac muscle and determined whether overexpression of SSPN into mdx mice ameliorates cardiac disease symptoms associated with Duchenne muscular dystrophy cardiomyopathy. METHODS AND RESULTS: SSPN-null mice exhibited cardiac enlargement, exacerbated cardiomyocyte hypertrophy, and increased fibrosis in response to ß-adrenergic challenge (isoproterenol; 0.8 mg/day per 2 weeks). Biochemical analysis of SSPN-null cardiac muscle revealed reduced sarcolemma localization of many proteins with a known role in cardiomyopathy pathogenesis: dystrophin, the sarcoglycans (α-, δ-, and γ-subunits), and ß1D integrin. Transgenic overexpression of SSPN in Duchenne muscular dystrophy mice (mdx(TG)) improved cardiomyofiber cell adhesion, sarcolemma integrity, cardiac functional parameters, as well as increased expression of compensatory transmembrane proteins that mediate attachment to the extracellular matrix. CONCLUSIONS: SSPN regulates sarcolemmal expression of laminin-binding complexes that are critical to cardiac muscle function and protects against transient and chronic injury, including inherited cardiomyopathy.


Asunto(s)
Cardiomiopatías/etiología , Proteínas Portadoras/fisiología , Corazón/efectos de los fármacos , Isoproterenol/farmacología , Proteínas de la Membrana/fisiología , Distrofia Muscular de Duchenne/complicaciones , Proteínas de Neoplasias/fisiología , Animales , Cardiomiopatías/patología , Forma MB de la Creatina-Quinasa/sangre , Ecocardiografía , Técnica del Anticuerpo Fluorescente , Corazón/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distrofia Muscular de Duchenne/patología , Miocardio/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcolema/fisiología
7.
J Pathol ; 237(4): 482-94, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26213100

RESUMEN

The low molecular weight protein tyrosine phosphatase (LMPTP), encoded by the ACP1 gene, is a ubiquitously expressed phosphatase whose in vivo function in the heart and in cardiac diseases remains unknown. To investigate the in vivo role of LMPTP in cardiac function, we generated mice with genetic inactivation of the Acp1 locus and studied their response to long-term pressure overload. Acp1(-/-) mice develop normally and ageing mice do not show pathology in major tissues under basal conditions. However, Acp1(-/-) mice are strikingly resistant to pressure overload hypertrophy and heart failure. Lmptp expression is high in the embryonic mouse heart, decreased in the postnatal stage, and increased in the adult mouse failing heart. We also show that LMPTP expression increases in end-stage heart failure in humans. Consistent with their protected phenotype, Acp1(-/-) mice subjected to pressure overload hypertrophy have attenuated fibrosis and decreased expression of fibrotic genes. Transcriptional profiling and analysis of molecular signalling show that the resistance of Acp1(-/-) mice to pathological cardiac stress correlates with marginal re-expression of fetal cardiac genes, increased insulin receptor beta phosphorylation, as well as PKA and ephrin receptor expression, and inactivation of the CaMKIIδ pathway. Our data show that ablation of Lmptp inhibits pathological cardiac remodelling and suggest that inhibition of LMPTP may be of therapeutic relevance for the treatment of human heart failure.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Cardiomiopatía de Takotsubo/metabolismo , Animales , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Ratas
8.
Sci Rep ; 5: 9042, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25761764

RESUMEN

Cardiac dysfunction is a prominent cause of mortality in myotonic dystrophy I (DM1), a disease where expanded CUG repeats bind and disable the muscleblind-like family of splice regulators. Deletion of muscleblind-like 1 (Mbnl1(ΔE2/ΔE2)) in 129 sv mice results in QRS, QTc widening, bundle block and STc narrowing at 2-4 months of age. With time, cardiac function deteriorates further and at 6 months, decreased R wave amplitudes, sinus node dysfunction, cardiac hypertrophy, interstitial fibrosis, multi-focal myocardial fiber death and calcification manifest. Sudden death, where no end point illness is overt, is observed at a median age of 6.5 and 4.8 months in ~67% and ~86% of male and female Mbnl1(ΔE2/ΔE2) mice, respectively. Mbnl1 depletion results in the persistence of embryonic splice isoforms in a network of cardiac RNAs, some of which have been previously implicated in DM1, regulating sodium and calcium currents, Scn5a, Junctin, Junctate, Atp2a1, Atp11a, Cacna1s, Ryr2, intra and inter cellular transport, Clta, Stx2, Tjp1, cell survival, Capn3, Sirt2, Csda, sarcomere and cytoskeleton organization and function, Trim55, Mapt, Pdlim3, Pdlim5, Sorbs1, Sorbs2, Fhod1, Spag9 and structural components of the sarcomere, Myom1, Tnnt2, Zasp. Thus this study supports a key role for Mbnl1 loss in the initiation of DM1 cardiac disease.


Asunto(s)
Empalme Alternativo , Eliminación de Gen , Distrofia Miotónica/genética , Isoformas de ARN , Proteínas de Unión al ARN/genética , Animales , Arritmia Sinusal , Calcinosis , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Fibrosis , Expresión Génica , Orden Génico , Marcación de Gen , Sitios Genéticos , Longevidad/genética , Masculino , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Distrofia Miotónica/fisiopatología , Fenotipo
9.
J Mol Cell Cardiol ; 53(6): 790-800, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22999861

RESUMEN

White mature adipocytes give rise to multipotent cells, so-called de-differentiated fat (DFAT) cells, when losing their fat in culture. The objective of this study was to examine the ability of DFAT cells to give rise to endothelial cells (ECs) in vitro and vivo. We demonstrate that mouse and human DFAT cells, derived from adipose tissue and lipospirate, respectively, initially lack expression of CD34, CD31, CD146, CD45 and pericyte markers, distinguishing them from progenitor cells previously identified in adipose stroma. The DFAT cells spontaneously differentiate into vascular ECs in vitro, as determined by real-time PCR, fluorescence activated cell sorting, immunostaining, and formation of tube structures. Treatment with bone morphogenetic protein (BMP)4 and BMP9, important in regulating angiogenesis, significantly enhances the EC differentiation. Furthermore, adipocyte-derived cells from Green Fluorescent Protein-transgenic mice were detected in the vasculature of infarcted myocardium up to 6 weeks after ligation of the left anterior descending artery in mice. We conclude that adipocyte-derived multipotent cells are able to spontaneously give rise to ECs, a process that is promoted by BMPs and may be important in cardiovascular regeneration and in physiological and pathological changes in fat and other tissues.


Asunto(s)
Adipocitos Blancos/citología , Desdiferenciación Celular/fisiología , Diferenciación Celular/fisiología , Células Madre Multipotentes/citología , Adipocitos Blancos/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 4/farmacología , Desdiferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Expresión Génica , Factores de Diferenciación de Crecimiento/farmacología , Humanos , Masculino , Ratones , Ratones Transgénicos , Células Madre Multipotentes/efectos de los fármacos , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Trasplante de Células Madre
10.
J Physiol ; 590(23): 6213-26, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22988135

RESUMEN

The circadian system co-ordinates the temporal patterning of behaviour and many underlying biological processes. In some cases, the regulated outputs of the circadian system, such as activity, may be able to feed back to alter core clock processes. In our studies, we used four wheel-access conditions (no access; free access; early night; and late night) to manipulate the duration and timing of activity while under the influence of a light-dark cycle. In wild-type mice, scheduled wheel access was able to increase ambulatory activity, inducing a level of exercise driven at various phases of the light-dark cycle. Scheduled exercise also manipulated the magnitude and phasing of the circadian-regulated outputs of heart rate and body temperature. At a molecular level, the phasing and amplitude of PER2::LUCIFERASE (PER2::LUC) expression rhythms in the SCN and peripheral tissues of Per2::Luc knockin mice were altered by scheduled exercise. We then tested whether scheduled wheel access could improve deficits observed in vasointestinal polypeptide-deficient mice under the influence of a light-dark cycle. We found that scheduled wheel access during the late night improved many of the behavioural, physiological and molecular deficits previously described in vasointestinal polypeptide-deficient mice. Our results raise the possibility that scheduled exercise could be used as a tool to modulate daily rhythms and, when applied, may counteract some of the negative impacts of ageing and disease on the circadian system.


Asunto(s)
Ritmo Circadiano/fisiología , Condicionamiento Físico Animal/fisiología , Péptido Intestinal Vasoactivo/fisiología , Animales , Conducta Animal/fisiología , Temperatura Corporal/fisiología , Expresión Génica , Frecuencia Cardíaca/fisiología , Ratones , Ratones Transgénicos , Proteínas Circadianas Period/genética
11.
Am J Physiol Heart Circ Physiol ; 300(1): H241-50, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20952671

RESUMEN

The circadian system, driven by the suprachiasmatic nucleus (SCN), regulates properties of cardiovascular function. The dysfunction of this timing system can result in cardiac pathology. The neuropeptide vasoactive intestinal peptide (VIP) is crucial for circadian rhythms in a number of biological processes including SCN electrical activity and wheel running behavior. Anatomic evidence indicates that SCN neurons expressing VIP are well positioned to drive circadian regulation of cardiac function through interactions with the autonomic centers. In this study, we tested the hypothesis that loss of VIP would result in circadian deficits in heart rate (HR) and clock gene expression in cardiac tissue. We implanted radiotelemetry devices into VIP-deficient mice and wild-type (WT) controls and continuously recorded HR, body temperature, and cage activity in freely moving mice. Under light-dark conditions, VIP-deficient mice displayed weak rhythms in HR, body temperature, and cage activity, with onsets that were advanced in phase compared with WT mice. Similarly, clock gene expression in cardiac tissue was rhythmic but phase advanced in mutant mice. In constant darkness, the normal circadian rhythms in HR were lost in VIP-deficient mice; however, most mutant mice continued to exhibit circadian rhythms of body temperature with shortened free-running period. The loss of VIP altered, but did not abolish, autonomic regulation of HR. Analysis of the echocardiograms did not find any evidence for a loss of cardiac function in VIP-deficient mice, and the size of the hearts did not differ between genotypes. These results demonstrate that VIP is an important regulator of physiological circadian rhythmicity in the heart.


Asunto(s)
Ritmo Circadiano/fisiología , Frecuencia Cardíaca/fisiología , Actividad Motora/fisiología , Miocardio/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Factores de Transcripción ARNTL/metabolismo , Análisis de Varianza , Animales , Temperatura Corporal/fisiología , Ecocardiografía , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Proteínas Circadianas Period/metabolismo , Receptores de Péptido Intestinal Vasoactivo/genética , Receptores de Péptido Intestinal Vasoactivo/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telemetría , Péptido Intestinal Vasoactivo/genética
12.
PLoS One ; 5(5): e10763, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20505827

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is the most common, lethal disease of childhood. One of 3500 new-born males suffers from this universally-lethal disease. Other than the use of corticosteroids, little is available to affect the relentless progress of the disease, leading many families to use dietary supplements in hopes of reducing the progression or severity of muscle wasting. Arginine is commonly used as a dietary supplement and its use has been reported to have beneficial effects following short-term administration to mdx mice, a genetic model of DMD. However, the long-term effects of arginine supplementation are unknown. This lack of knowledge about the long-term effects of increased arginine metabolism is important because elevated arginine metabolism can increase tissue fibrosis, and increased fibrosis of skeletal muscles and the heart is an important and potentially life-threatening feature of DMD. METHODOLOGY: We use both genetic and nutritional manipulations to test whether changes in arginase metabolism promote fibrosis and increase pathology in mdx mice. Our findings show that fibrotic lesions in mdx muscle are enriched with arginase-2-expressing macrophages and that muscle macrophages stimulated with cytokines that activate the M2 phenotype show elevated arginase activity and expression. We generated a line of arginase-2-null mutant mdx mice and found that the mutation reduced fibrosis in muscles of 18-month-old mdx mice, and reduced kyphosis that is attributable to muscle fibrosis. We also observed that dietary supplementation with arginine for 17-months increased mdx muscle fibrosis. In contrast, arginine-2 mutation did not reduce cardiac fibrosis or affect cardiac function assessed by echocardiography, although 17-months of dietary supplementation with arginine increased cardiac fibrosis. Long-term arginine treatments did not decrease matrix metalloproteinase-2 or -9 or increase the expression of utrophin, which have been reported as beneficial effects of short-term treatments. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that arginine metabolism by arginase promotes fibrosis of muscle in muscular dystrophy and contributes to kyphosis. Our findings also show that long-term, dietary supplementation with arginine exacerbates fibrosis of dystrophic heart and muscles. Thus, commonly-practiced dietary supplementation with arginine by DMD patients has potential risk for increasing pathology when performed for long periods, despite reports of benefits acquired with short-term supplementation.


Asunto(s)
Arginina/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/patología , Miocardio/metabolismo , Miocardio/patología , Animales , Arginasa/metabolismo , Arginina/administración & dosificación , Arginina/farmacología , Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/patología , Citocinas/metabolismo , Distrofina/deficiencia , Distrofina/metabolismo , Fibrosis , Eliminación de Gen , Inflamación/complicaciones , Inflamación/enzimología , Inflamación/patología , Cifosis/complicaciones , Cifosis/enzimología , Cifosis/patología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/enzimología , Distrofia Muscular Animal/complicaciones , Distrofia Muscular Animal/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Transporte de Proteínas/efectos de los fármacos , Células Th2/efectos de los fármacos
13.
Neural Dev ; 5: 6, 2010 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-20184720

RESUMEN

BACKGROUND: All-trans retinoic acid (atRA) is required for nervous system development, including the developing hindbrain region. Neuron navigator 2 (Nav2) was first identified as an atRA-responsive gene in human neuroblastoma cells (retinoic acid-induced in neuroblastoma 1, Rainb1), and is required for atRA-mediated neurite outgrowth. In this paper, we explore the importance of Nav2 in nervous system development and function in vivo. RESULTS: Nav2 hypomorphic homozygous mutants show decreased survival starting at birth. Nav2 mutant embryos show an overall reduction in nerve fiber density, as well as specific defects in cranial nerves IX (glossopharyngeal) and X (vagus). Nav2 hypomorphic mutant adult mice also display a blunted baroreceptor response compared to wild-type controls. CONCLUSIONS: Nav2 functions in mammalian nervous system development, and is required for normal cranial nerve development and blood pressure regulation in the adult.


Asunto(s)
Presión Sanguínea/fisiología , Nervios Craneales/crecimiento & desarrollo , Desarrollo Embrionario/genética , Proteínas del Tejido Nervioso/metabolismo , Presorreceptores/metabolismo , Animales , Presión Sanguínea/genética , Nervios Craneales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Nervio Glosofaríngeo/crecimiento & desarrollo , Nervio Glosofaríngeo/metabolismo , Homocigoto , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neuronas/fisiología , Presorreceptores/crecimiento & desarrollo , Rombencéfalo/crecimiento & desarrollo , Rombencéfalo/metabolismo , Nervio Vago/crecimiento & desarrollo , Nervio Vago/metabolismo
14.
J Mol Cell Cardiol ; 47(4): 552-60, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19683723

RESUMEN

Na,K-ATPase is composed of two essential alpha- and beta-subunits, both of which have multiple isoforms. Evidence indicates that the Na,K-ATPase enzymatic activity as well as its alpha(1), alpha(3) and beta(1) isoforms are reduced in the failing human heart. The catalytic alpha-subunit is the receptor for cardiac glycosides such as digitalis, used for the treatment of congestive heart failure. The role of the Na,K-ATPase beta(1)-subunit (Na,K-beta(1)) in cardiac function is not known. We used Cre/loxP technology to inactivate the Na,K-beta(1) gene exclusively in the ventricular cardiomyocytes. Animals with homozygous Na,K-beta(1) gene excision were born at the expected Mendelian ratio, grew into adulthood, and appeared to be healthy until 10 months of age. At 13-14 months, these mice had 13% higher heart/body weight ratios, and reduced contractility as revealed by echocardiography compared to their wild-type (WT) littermates. Pressure overload by transverse aortic constriction (TAC) in younger mice, resulted in compensated hypertrophy in WT mice, but decompensation in the Na,K-beta(1) KO mice. The young KO survivors of TAC exhibited decreased contractile function and mimicked the effects of the Na,K-beta(1) KO in older mice. Further, we show that intact hearts of Na,K-beta(1) KO anesthetized mice as well as isolated cardiomyocytes were insensitive to ouabain-induced positive inotropy. This insensitivity was associated with a reduction in NCX1, one of the proteins involved in regulating cardiac contractility. In conclusion, our results demonstrate that Na,K-beta(1) plays an essential role in regulating cardiac contractility and that its loss is associated with significant pathophysiology of the heart.


Asunto(s)
Eliminación de Gen , Contracción Miocárdica/efectos de los fármacos , Miocardio/enzimología , Ouabaína/farmacología , Subunidades de Proteína/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Cardiomegalia/enzimología , Cardiomegalia/fisiopatología , Separación Celular , Pruebas de Función Cardíaca , Immunoblotting , Ratones , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Especificidad de Órganos/efectos de los fármacos , Presión , Intercambiador de Sodio-Calcio/metabolismo
15.
J Surg Res ; 153(2): 217-23, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18694573

RESUMEN

BACKGROUND: Adipose tissue consists of mature adipocytes and a mononuclear cell fraction termed adipose tissue-derived cells (ADCs). Within these heterogeneous ADCs exists a mesenchymal stem cell-like cell population, termed adipose tissue-derived stem cells. An important clinical advantage of adipose tissue-derived stem cells over other mesenchymal stem cell populations is the fact that they can be isolated in real time in sufficient quantity, such that ex vivo expansion is not necessary to obtain clinically relevant numbers for various therapeutic applications. MATERIALS AND METHODS: The aim of this investigation was to evaluate the therapeutic potential of freshly isolated ADCs in treating rats acutely following myocardial infarction. Rats underwent 45 min of left anterior descending artery occlusion followed by reperfusion. Fifteen minutes post-myocardial infarction, saline or 5 x 10(6) ADCs from green fluorescent protein-expressing transgenic rats were injected into the chamber of the left ventricle. Left ventricular function and morphometry was followed with 2-D echocardiography for 12 wk, at which point hearts were harvested for histological analysis. RESULTS: Twelve weeks following cell therapy, left ventricular end-diastolic dimension was less dilated while the ejection fraction and cardiac output of ADC-treated rats were significantly improved compared to control rats (P < 0.01). Despite this benefit, absolute engraftment rates were low. This paradox may be partially explained by ADC-induced increases in both capillary and arteriole densities. CONCLUSIONS: These data confirm the therapeutic benefit of freshly isolated ADCs delivered post-MI and suggest a novel beneficial mechanism for ADCs through a potent proangiogenic effect.


Asunto(s)
Tejido Adiposo/citología , Trasplante de Células Madre Mesenquimatosas , Infarto del Miocardio/terapia , Neovascularización Fisiológica , Remodelación Ventricular , Animales , Arteriolas/crecimiento & desarrollo , Capilares/crecimiento & desarrollo , Vasos Coronarios/crecimiento & desarrollo , Masculino , Ratas , Ratas Endogámicas Lew , Función Ventricular Izquierda
16.
Circ Res ; 102(10): 1222-9, 2008 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-18420946

RESUMEN

Normally, cell cycle progression is tightly coupled to the accumulation of cell mass; however, the mechanisms whereby proliferation and cell growth are linked are poorly understood. We have identified cyclin (Cyc)D2, a G(1) cyclin implicated in mediating S phase entry, as a potential regulator of hypertrophic growth in adult post mitotic myocardium. To examine the role of CycD2 and its downstream targets, we subjected CycD2-null mice to mechanical stress. Hypertrophic growth in response to transverse aortic constriction was attenuated in CycD2-null compared with wild-type mice. Blocking the increase in CycD2 in response to hypertrophic agonists prevented phosphorylation of CycD2-target Rb (retinoblastoma gene product) in vitro, and mice deficient for Rb had potentiated hypertrophic growth. Hypertrophic growth requires new protein synthesis and transcription of tRNA genes by RNA polymerase (pol) III, which increases with hypertrophic signals. This load-induced increase in RNA pol III activity is augmented in Rb-deficient hearts. Rb binds and represses Brf-1 and TATA box binding protein (TBP), subunits of RNA pol III-specific transcription factor B, in adult myocardium under basal conditions. However, this association is disrupted in response to transverse aortic constriction. RNA pol III activity is unchanged in CycD2(-/-) myocardium after transverse aortic constriction, and there is no dissociation of TBP from Rb. These investigations identify an essential role for the CycD2-Rb pathway as a governor of cardiac myocyte enlargement in response to biomechanical stress and, more fundamentally, as a regulator of the load-induced activation of RNA pol III.


Asunto(s)
Cardiomegalia/metabolismo , Ciclinas/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , ARN Polimerasa III/metabolismo , Proteína de Retinoblastoma/metabolismo , Factores de Edad , Animales , Cardiomegalia/patología , Tamaño de la Célula , Células Cultivadas , Ciclina D2 , Ciclinas/genética , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F3/metabolismo , Factor de Transcripción E2F4/metabolismo , Factor de Transcripción E2F5/metabolismo , Ratones , Ratones Mutantes , Miocardio/citología , Miocitos Cardíacos/citología , Fosforilación , Ratas , Proteína de Retinoblastoma/genética , Transducción de Señal/fisiología , Estrés Mecánico
17.
J Surg Res ; 148(2): 164-71, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18067924

RESUMEN

The mechanism(s) underlying the beneficial effects of adult mesenchymal stem cells (MSCs) after myocardial infarction (MI) is poorly understood. One possible explanation is the ability of MSCs to secrete cytokines, which modulate cardiomyocyte survival and function. MSCs express at least two cytoprotective cytokines, hepatocyte growth factor (HGF) and stromal cell-derived factor-1 alpha (CXCL12). The aim of our study was to compare the effects of these two cytokines administered acutely post-MI. We subjected adult male Lewis rats to myocardial ischemia/reperfusion injury. Immediately upon reperfusion, polymers saturated with HGF or CXCL12 were placed onto the infarcted anterior wall and the rats were allowed to recover. Echocardiographic analysis at 4 wk post-MI to assess left ventricular (LV) function revealed that LV ejection fraction was increased in the HGF treated group compared with the phosphate-buffered saline (PBS) control group. Likewise, LV end diastolic dimension was reduced in the HGF treated group compared with the PBS control group. Similarly, invasive hemodynamics at 12 wk showed improved contractility and relaxation in the HGF treated group compared with the PBS control group. In contrast, no significant effect on LV function was seen in the CXCL12 treated group. To determine the potential mechanism for this effect, infarct size (IFS) at 72 h was determined. IFS was decreased 4.2-fold in the HGF treated group compared with the PBS control group. Thus, HGF acutely post-MI using polymer delivery reduces IFS, leading to beneficial effects on post-MI LV remodeling.


Asunto(s)
Quimiocina CXCL12/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Células Cultivadas , Quimiocina CXCL12/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Factor de Crecimiento de Hepatocito/uso terapéutico , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Isquemia Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Ratas , Ratas Endogámicas Lew , Volumen Sistólico/efectos de los fármacos , Volumen Sistólico/fisiología , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/fisiología
18.
Cell ; 130(4): 691-703, 2007 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-17719546

RESUMEN

Vascular endothelial growth factor (VEGF) is essential for developmental and pathological angiogenesis. Here we show that in the absence of any pathological insult, autocrine VEGF is required for the homeostasis of blood vessels in the adult. Genetic deletion of vegf specifically in the endothelial lineage leads to progressive endothelial degeneration and sudden death in 55% of mutant mice by 25 weeks of age. The phenotype is manifested without detectable changes in the total levels of VEGF mRNA or protein, indicating that paracrine VEGF could not compensate for the absence of endothelial VEGF. Furthermore, wild-type, but not VEGF null, endothelial cells showed phosphorylation of VEGFR2 in the absence of exogenous VEGF. Activation of the receptor in wild-type cells was suppressed by small molecule antagonists but not by extracellular blockade of VEGF. These results reveal a cell-autonomous VEGF signaling pathway that holds significance for vascular homeostasis but is dispensable for the angiogenic cascade.


Asunto(s)
Comunicación Autocrina , Endotelio Vascular/metabolismo , Homeostasis , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Vasos Sanguíneos/citología , Hipoxia de la Célula , Supervivencia Celular , Células Cultivadas , Cobalto/toxicidad , Cruzamientos Genéticos , Ecocardiografía , Endotelio Vascular/citología , Ratones , Ratones Mutantes , Ratones Transgénicos , Modelos Biológicos , Fosforilación , ARN Mensajero/metabolismo , Telemetría , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
19.
EMBO J ; 25(16): 3869-79, 2006 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-16902412

RESUMEN

c-Myc (Myc) is highly expressed in developing embryos where it regulates body size by controlling proliferation but not cell size. However, Myc is also induced in many postmitotic tissues, including adult myocardium, in response to stress where the predominant form of growth is an increase in cell size (hypertrophy) and not number. The function of Myc induction in this setting is unproven. Therefore, to explore Myc's role in hypertrophic growth, we created mice where Myc can be inducibly inactivated, specifically in adult myocardium. Myc-deficient hearts demonstrated attenuated stress-induced hypertrophic growth, secondary to a reduction in cell growth of individual myocytes. To explore the dependence of Myc-induced cell growth on CycD2, we created bigenic mice where Myc can be selectively activated in CycD2-null adult myocardium. Myc-dependent hypertrophic growth and cell cycle reentry is blocked in CycD2-deficient hearts. However, in contrast to Myc-induced DNA synthesis, hypertrophic growth is independent of CycD2-induced Cdk2 activity. These data suggest that Myc is required for a normal hypertrophic response and that its growth-promoting effects are also mediated through a CycD2-dependent pathway.


Asunto(s)
Cardiomegalia/patología , Ciclinas/fisiología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Animales , Apoptosis , Cardiomegalia/metabolismo , Ciclo Celular , Aumento de la Célula , Proliferación Celular , Células Cultivadas , Ciclina D2 , Ciclinas/genética , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética
20.
Mol Genet Metab ; 86(1-2): 233-43, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15979918

RESUMEN

Mucopolysaccharidosis I (MPS I, alpha-l-iduronidase deficiency disease) is a heritable lysosomal storage disorder involving multiple organs, including the heart. Malfunction of the heart is also a major manifestation in the mouse model of MPS I, progressing in severity from 6 to 10 months (of a 1 year life span). In comparisons of MPS I with wild-type mice, the heart was found enlarged, with thickened septal and posterior walls, primarily because of infiltration of the muscle by storage-laden cells. Heart valves were enlarged and misshapen, and contained large numbers of highly vacuolated interstitial cells. The thickened aortic wall contained vacuolated smooth muscle cells and interrupted elastic fibers. Hemodynamic measurements and echocardiography revealed reduced left ventricular function as well as mitral and aortic regurgitation. But despite these abnormalities, free-roaming MPS I mice implanted with radio telemetry devices showed surprisingly normal heart rate and blood pressure, though their electrocardiograms were abnormal. An incidental finding of the telemetry studies was a disturbed circadian rhythm in the MPS I mice. Restoration of enzyme activity in the heart of one mouse, by transplantation of retrovirally modified bone marrow, resulted in normalization of left ventricular function as well as loss of storage vacuoles in myocytes and endothelial cells, though not in valvular interstitial cells. This study demonstrates the usefulness of the mouse model for in-depth studies of the cardiovascular component of MPS I.


Asunto(s)
Modelos Animales de Enfermedad , Corazón/fisiopatología , Mucopolisacaridosis I/fisiopatología , Animales , Trasplante de Médula Ósea , Ecocardiografía , Electrocardiografía , Terapia Genética , Vectores Genéticos , Glicosaminoglicanos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mucopolisacaridosis I/metabolismo , Mucopolisacaridosis I/terapia , Retroviridae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA