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










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(2)2023 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-36675170

RESUMEN

Binge Drinking (BD) corresponds to episodes of ingestion of large amounts of ethanol in a short time, typically ≤2 h. BD occurs across all populations, but young and sports-related people are especially vulnerable. However, the short- and long-term effects of episodic BD on skeletal muscle function have been poorly explored. Young rats were randomized into two groups: control and episodic Binge-Like ethanol protocol (BEP) (ethanol 3 g/kg IP, 4 episodes of 2-days ON-2-days OFF paradigm). Muscle function was evaluated two weeks after the last BEP episode. We found that rats exposed to BEP presented decreased muscle strength and increased fatigability, compared with control animals. Furthermore, we observed that skeletal muscle from rats exposed to BEP presented muscle atrophy, evidenced by reduced fiber size and increased expression of atrophic genes. We also observed that BEP induced fibrotic and inflammation markers, accompanied by mislocalization of nNOSµ and high levels of protein nitration. Our findings suggest that episodic binge-like ethanol exposure alters contractile capacity and increases fatigue by mechanisms involving atrophy, fibrosis, and inflammation, which remain for at least two weeks after ethanol clearance. These pathological features are common to several neuromuscular diseases and might affect muscle performance and health in the long term.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas , Etanol , Ratas , Animales , Etanol/efectos adversos , Etanol/metabolismo , Músculo Esquelético/metabolismo , Inflamación/metabolismo , Atrofia Muscular/metabolismo , Fuerza Muscular , Fibrosis , Consumo Excesivo de Bebidas Alcohólicas/metabolismo
2.
Early Interv Psychiatry ; 17(4): 378-384, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35716056

RESUMEN

AIM: To evaluate the impact of long-acting injectable antipsychotics (LAIs) on the risk of hospitalization and the length of hospitalization in the setting of an early intervention program for patients with recent-onset psychosis. METHODS: Observational, retrospective study conducted under routine clinical practice conditions. We included all patients admitted from July 2015 to April 2020 to the Early Intervention Program in Psychosis. We analysed the incidence of hospitalization and hospitalization days before and after treatment with LAIs and calculated the incidence rate ratio (IRR). We also compared the outcomes of patients treated with LAIs with those of the patients maintained on oral antipsychotics using a binomial negative regression analysis. RESULTS: A total of 170 patients were included in the program. Of them, 34 (20%) received LAIs (aripiprazole [n = 22], and paliperidone/risperidone [n = 12]). There was an 89% reduction in the incidence of hospitalizations after treatment with LAIs (IRR 0.11, 95%CI 0.05-0.21; p < .0001). The IRR for LAIs vs. oral antipsychotics was 0.87 (95%CI, 0.24-3.18; p = .829). The presence of a substance use disorder significantly increased the rate of hospitalizations by 123% (IRR 2.23, 95%CI 1.31-3.78). Analyses of hospitalization days showed similar results. CONCLUSIONS: Our results suggest that LAIs are useful for the management of patients with recent-onset psychosis who fail treatment with oral antipsychotics. Whether LAIs are superior to oral antipsychotics as first-line treatment of patients with early psychosis and/or could play a special role in managing patients with early psychosis and comorbid substance use disorders should be further evaluated.


Asunto(s)
Antipsicóticos , Trastornos Psicóticos , Esquizofrenia , Humanos , Antipsicóticos/efectos adversos , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/epidemiología , Estudios Retrospectivos , Intervención Médica Temprana , Preparaciones de Acción Retardada/uso terapéutico , Administración Oral , Trastornos Psicóticos/tratamiento farmacológico , Trastornos Psicóticos/epidemiología
3.
Am J Drug Alcohol Abuse ; 48(2): 133-147, 2022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35389308

RESUMEN

Background: Chronic alcohol misuse is associated with alcoholic myopathy, characterized by skeletal muscle weakness and atrophy. Moreover, there is evidence that sports-related people seem to exhibit a greater prevalence of problematic alcohol consumption, especially binge drinking (BD), which might not cause alcoholic myopathy but can negatively impact muscle function and amateur and professional athletic performance.Objective: To review the literature concerning the effects of alcohol consumption on skeletal muscle function and structure that can affect muscle performance.Methodology: We examined the currently available literature (PubMed, Google Scholars) to develop a narrative review summarizing the knowledge about the effects of alcohol on skeletal muscle function and exercise performance, obtained from studies in human beings and animal models for problematic alcohol consumption.Results: Exercise- and sport-based studies indicate that alcohol consumption can negatively affect muscle recovery after vigorous exercise, especially in men, while women seem less affected. Clinical studies and pre-clinical laboratory research have led to the knowledge of some of the mechanisms involved in alcohol-related muscle dysfunction, including an imbalance between anabolic and catabolic pathways, reduced regeneration, increased inflammation and fibrosis, and deficiencies in energetic balance and mitochondrial function. These pathological features can appear not only under chronic alcohol misuse but also in other alcohol consumption patterns.Conclusions: Most laboratory-based studies use chronic or acute alcohol exposure, while episodic BD, the most common drinking pattern in amateur and professional athletes, is underrepresented. Nevertheless, alcohol consumption negatively affects skeletal muscle health through different mechanisms, which collectively might contribute to reduced sports performance.


Asunto(s)
Alcoholismo , Rendimiento Atlético , Enfermedades Musculares , Consumo de Bebidas Alcohólicas/metabolismo , Alcoholismo/epidemiología , Animales , Rendimiento Atlético/fisiología , Etanol/farmacología , Femenino , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología
4.
Int J Mol Sci ; 22(10)2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-34063397

RESUMEN

The Cellular Communication Network (CCN) family of matricellular proteins comprises six proteins that share conserved structural features and play numerous biological roles. These proteins can interact with several receptors or soluble proteins, regulating cell signaling pathways in various tissues under physiological and pathological conditions. In the skeletal muscle of mammals, most of the six CCN family members are expressed during embryonic development or in adulthood. Their roles during the adult stage are related to the regulation of muscle mass and regeneration, maintaining vascularization, and the modulation of skeletal muscle fibrosis. This work reviews the CCNs proteins' role in skeletal muscle physiology and disease, focusing on skeletal muscle fibrosis and its regulation by Connective Tissue Growth factor (CCN2/CTGF). Furthermore, we review evidence on the modulation of fibrosis and CCN2/CTGF by the renin-angiotensin system and the kallikrein-kinin system of vasoactive peptides.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Músculo Esquelético/fisiología , Péptidos/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Cininas/metabolismo , Familia de Multigenes , Proteínas Musculares/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Regeneración , Sistema Renina-Angiotensina
5.
J Cell Commun Signal ; 14(2): 147-158, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32088838

RESUMEN

Hypoxia refers to the decrease in oxygen tension in the tissues, and the central effector of the hypoxic response is the transcription factor Hypoxia-Inducible Factor α (HIF1-α). Transient hypoxia in acute events, such as exercising or regeneration after damage, play an important role in skeletal muscle physiology and homeostasis. However, sustained activation of hypoxic signaling is a feature of skeletal muscle injury and disease, which can be a consequence of chronic damage but can also increase the severity of the pathology and worsen its outcome. Here, we review evidence that supports the idea that hypoxia and HIF-1α can contribute to the establishment of fibrosis in skeletal muscle through its crosstalk with other profibrotic factors, such as Transforming growth factor ß (TGF-ß), the induction of profibrotic cytokines expression, as is the case of Connective Tissue Growth Factor (CTGF/CCN2), or being the target of the Renin-angiotensin system (RAS).

6.
J Cell Commun Signal ; 12(1): 413-421, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28887614

RESUMEN

Connective tissue growth factor (CTGF/CCN2) has strong inflammatory and profibrotic activities. Its expression is enhanced in skeletal muscular dystrophies such as Duchenne muscular dystrophy (DMD), a myopathy characterized by exacerbated inflammation and fibrosis. In dystrophic tissue, necrotic-regenerative foci, myofibroblasts, newly-regenerated muscle fibers and necrosis all occur simultaneously. To determine if CCN2 is involved in the appearance of the foci, we studied their presence and characteristics in mdx mice (DMD mouse model) compared to mdx mice hemizygous for CCN2 (mdx-Ccn2+/-). We used laser capture microdissection followed by gene expression and immunofluorescence analyses to investigate fibrotic, inflammation and regeneration markers in damaged and non-damaged areas in mdx and mdx-Ccn2+/- skeletal muscle. Mdx mice foci express elevated mRNAs levels of transforming growth factor type beta, collagen, fibronectin, the myofribroblast marker α-SMA, and the myogenic transcription factor myogenin. Mdx foci also show elevated levels of MCP-1 and CD-68 positive cells, indicating that CCN2 could be inducing an inflammatory response. We found a significant reduction in the number of foci in mdx-Ccn2+/- mice muscle. Fibrotic and inflammatory markers were also decreased in these foci. We did not observe any difference in Pax7 mRNA levels, a marker for satellite cells, in mdx mice compared to mdx-Ccn2+/- mice. Thus, CCN2 appears to be involved in the fibrotic response as well as in the inflammatory response in the dystrophic skeletal muscle.

7.
J Cell Commun Signal ; 12(3): 589-601, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29250740

RESUMEN

The Kallikrein Kinin System (KKS) is a vasoactive peptide system with known functions in the maintenance of tissue homeostasis, renal function and blood pressure. The main effector peptide of KKS is Bradykinin (BK). This ligand has two receptors: a constitutive B2 receptor (B2R), which has been suggested to have anti-fibrotic effects in renal and cardiac models of fibrosis; and the inducible B1 receptor (B1R), whose expression is induced by damage and inflammation. Inflammation and fibrosis are hallmarks of Duchenne muscular dystrophy (DMD), therefore we hypothesized that the KKS may play a role in this disease. To evaluate this hypothesis we used the mdx mouse a model for DMD. We blocked the endogenous activity of the KKS by treating mdx mice with B2R antagonist (HOE-140) or B1R antagonist (DesArgLeu8BK (DALBK)) for four weeks. Both antagonists increased damage, fibrosis, TGF-ß and Smad-dependent signaling, CTGF/CCN-2 levels as well as the number of CD68 positive inflammatory cells. B2R blockade also reduced isolated muscle contraction force. These results indicate that the endogenous KKS has a protective role in the dystrophic muscle. The KKS may be a new target for future therapies to reduce inflammation and fibrosis in dystrophic muscle.

9.
Methods Mol Biol ; 1489: 513-521, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27734402

RESUMEN

CCN2 or connective tissue growth factor (CTGF) is a matricellular protein that regulates several cellular processes. In skeletal muscle, CTGF is a key modulator of fibrogenesis, is increased in pathological conditions such as muscular dystrophies, and plays a major role in the pathology outcome. Overexpression of CTGF in skeletal muscle of wild-type mice results in muscle damage, fibrosis, and reduction of strength. In contrast, a decrease in CTGF in dystrophic mice increases strength and reduces damage and fibrosis. Thus, CTGF is a relevant target to study in skeletal muscle pathology and its possible modulation by different treatments or potential new drugs to develop new strategies for the treatment of muscular dystrophies. We summarize the techniques used to detect CTGF in the skeletal muscle of dystrophic mdx mice.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/patología , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Dis Model Mech ; 9(4): 441-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26851244

RESUMEN

Immobilization is a form of disuse characterized by a loss of strength and muscle mass. Among the main features are decreased IGF-1/Akt signalling and increased ubiquitin-proteasome pathway signalling, which induce greater myosin heavy chain degradation. Activation of the classical renin-angiotensin system (RAS) causes deleterious effects in skeletal muscle, including muscle wasting. In contrast, angiotensin-(1-7) [Ang-(1-7)], a peptide of the non-classical RAS, produces beneficial effects in skeletal muscle. However, the role of Ang-(1-7) in skeletal muscle disuse atrophy and independent of classical RAS activation has not been evaluated. Therefore, we assessed the functions of Ang-(1-7) and the Mas receptor in disuse muscle atrophyin vivousing unilateral cast immobilization of the hind limb in male, 12-week-old wild-type (WT) and Mas-knockout (Mas KO) mice for 1 and 14 days. Additionally, we evaluated the participation of IGF-1/IGFR-1/Akt signalling and ubiquitin-proteasome pathway expression on the effects of Ang-(1-7) immobilization-induced muscle atrophy. Our results found that Ang-(1-7) prevented decreased muscle strength and reduced myofiber diameter, myosin heavy chain levels, and the induction of atrogin-1 and MuRF-1 expressions, all of which normally occur during immobilization. Analyses indicated that Ang-(1-7) increases IGF-1/IGFR-1/Akt pathway signalling through IGFR-1 and Akt phosphorylation, and the concomitant activation of two downstream targets of Akt, p70S6K and FoxO3. These anti-atrophic effects of Ang-(1-7) were not observed in Mas KO mice, indicating crucial participation of the Mas receptor. This report is the first to propose anti-atrophic effects of Ang-(1-7) via the Mas receptor and the participation of the IGF-1/IGFR-1/Akt/p70S6K/FoxO3 mechanism in disuse skeletal muscle atrophy.


Asunto(s)
Angiotensina I/uso terapéutico , Atrofia Muscular/tratamiento farmacológico , Trastornos Musculares Atróficos/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Angiotensina I/farmacología , Animales , Factor I del Crecimiento Similar a la Insulina/metabolismo , Contracción Isométrica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Proteínas Musculares/metabolismo , Fuerza Muscular/efectos de los fármacos , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Atrofia Muscular/fisiopatología , Trastornos Musculares Atróficos/metabolismo , Trastornos Musculares Atróficos/patología , Trastornos Musculares Atróficos/fisiopatología , Cadenas Pesadas de Miosina/metabolismo , Fragmentos de Péptidos/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor IGF Tipo 1/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
11.
Biofactors ; 41(2): 111-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25809912

RESUMEN

Duchenne muscular dystrophy is a genetic disorder characterized by myofiber degeneration, muscle weakness, and increased fibrosis. Transforming growth factor type-ß (TGF-ß), a central mediator of fibrosis, is upregulated in fibrotic diseases. Angiotensin-(1-7) [Ang-(1-7)] is a peptide with actions that oppose those of angiotensin-II (Ang II). Ang-(1-7) effects are mediated by the Mas receptor. Treatment with Ang-(1-7) produce positive effects in the mdx mouse, normalizing skeletal muscle architecture, decreasing local fibrosis, and fibroblasts, and improving muscle function. Mdx mice deficient for the Mas receptor showed the opposite effects. To identify the cell type(s) responsible for Mas receptor expression, and to characterize whether profibrotic effectors had any effect on its expression, we determined the effect of profibrotic agents on Mas expression. TGF-ß, but not connective tissue growth factor or Ang-II, reduced the expression of Mas receptor in fibroblasts isolated from skeletal muscle cells and fibroblasts from two established cell lines. In contrast, no effects were observed in myoblasts and differentiated myotubes. This inhibition was mediated by the Smad-dependent (canonical) and the PI3K and MEK1/2 (noncanonical) TGF-ß signaling pathways. When both canonical and noncanonical inhibitors of the TGF-ß-dependent pathways were added together, the inhibitory effect of TGF-ß on Mas expression was lost. The decrease in Mas receptor induced by TGF-ß in fibroblasts reduced the Ang-(1-7) mediated stimulation of phosphorylation of AKT pathway proteins. These results suggest that reduction of Mas receptor in fibroblasts, by TGF-ß, could increase the fibrotic phenotype observed in dystrophic skeletal muscle decreasing the beneficial effect of Ang-(1-7).


Asunto(s)
Fibroblastos/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Mioblastos/efectos de los fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/farmacología , Angiotensina I/farmacología , Angiotensina II/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Regulación de la Expresión Génica , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Mioblastos/metabolismo , Mioblastos/patología , Especificidad de Órganos , Fragmentos de Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
12.
PLoS One ; 9(4): e93449, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24695436

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common inherited neuromuscular disease and is characterized by absence of the cytoskeletal protein dystrophin, muscle wasting, and fibrosis. We previously demonstrated that systemic infusion or oral administration of angiotensin-(1-7) (Ang-(1-7)), a peptide with opposing effects to angiotensin II, normalized skeletal muscle architecture, decreased local fibrosis, and improved muscle function in mdx mice, a dystrophic model for DMD. In this study, we investigated the presence, activity, and localization of ACE2, the enzyme responsible for Ang-(1-7) production, in wild type (wt) and mdx skeletal muscle and in a model of induced chronic damage in wt mice. All dystrophic muscles studied showed higher ACE2 activity than wt muscle. Immunolocalization studies indicated that ACE2 was localized mainly at the sarcolemma and, to a lesser extent, associated with interstitial cells. Similar results were observed in the model of chronic damage in the tibialis anterior (TA) muscle. Furthermore, we evaluated the effect of ACE2 overexpression in mdx TA muscle using an adenovirus containing human ACE2 sequence and showed that expression of ACE2 reduced the fibrosis associated with TA dystrophic muscles. Moreover, we observed fewer inflammatory cells infiltrating the mdx muscle. Finally, mdx gastrocnemius muscles from mice infused with Ang-(1-7), which decreases fibrosis, contain less ACE2 associated with the muscle. This is the first evidence supporting ACE2 as an important therapeutic target to improve the dystrophic skeletal muscle phenotype.


Asunto(s)
Fibrosis/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Proteínas del Citoesqueleto/metabolismo , Fibrosis/patología , Humanos , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/patología , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/patología , Fragmentos de Péptidos/metabolismo
13.
Hum Mol Genet ; 23(5): 1237-49, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24163134

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common inherited neuromuscular disease, and is characterized by the lack of dystrophin, muscle wasting, increased transforming growth factor (TGF)-ß Smad-dependent signalling and fibrosis. Acting via the Mas receptor, angiotensin-1-7 [Ang-(1-7)], is part of the renin-angiotensin system, with the opposite effect to that of angiotensin II. We hypothesized that the Ang-(1-7)/Mas receptor axis might protect chronically damaged tissues as in skeletal muscle of the DMD mouse model mdx. Infusion or oral administration of Ang-(1-7) in mdx mice normalized skeletal muscle architecture, decreased local fibrosis and improved muscle function in vitro and in vivo. These positive effects were mediated by the inhibition of TGF-ß Smad signalling, which in turn led to reduction of the pro-fibrotic microRNA miR-21 concomitant with a reduction in the number of TCF4 expressing fibroblasts. Mdx mice infused with Mas antagonist (A-779) and mdx deficient for the Mas receptor showed highly deteriorated muscular architecture, increased fibrosis and TGF-ß signalling with diminished muscle strength. These results suggest that this novel compound Ang-(1-7) might be used to improve quality of life and delay death in individuals with DMD and this drug should be investigated in further pre-clinical trials.


Asunto(s)
Angiotensina I/farmacología , Fuerza Muscular/efectos de los fármacos , Distrofia Muscular de Duchenne/metabolismo , Fragmentos de Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Angiotensina I/administración & dosificación , Animales , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Humanos , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Fragmentos de Péptidos/administración & dosificación , Receptores de Superficie Celular/antagonistas & inhibidores
14.
Biofactors ; 39(4): 467-75, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23460581

RESUMEN

Excessive deposition of extracellular matrix (ECM) proteins, a condition known as fibrosis, is a hallmark of Duchenne muscular dystrophy. Among the factors that trigger muscle fibrosis are transforming growth factor beta (TGF-ß) and angiotensin II (Ang-II). Ang-II belongs to the renin-angiotensin system, and its biological effects are exerted by Ang-II receptors type 1 and type 2 (AT-1 and AT-2, respectively). This study aims to determine the effect of TGF-ß1 on the expression of AT-1 and AT-2 receptor in skeletal muscle. C2 C12 myoblasts exposed to TGF-ß1 showed a dose-dependent increase in AT-2 expression but with no effect on AT-1 levels. Injection of TGF-ß1 in the skeletal muscle of mice increased the levels of AT-2 and ECM protein but unchanged AT-1 levels. We also detected higher expression levels of AT-2 receptor in dystrophic skeletal muscle of mdx mice than in normal mice. The induction of AT-2 was mediated by the canonical TGF-ß pathway because under the inhibitory conditions of the kinase activity of TGFß receptor I or the knockdown of Smad2/3 levels, TGF-ß-induced AT-2 receptor increase was strongly inhibited. Furthermore, we demonstrated that p38MAPK activity in response to TGF-ß is also required for AT-2 increase as evaluated by a p38MAPK inhibitor. Our results show that the levels of AT-2 but not AT-1 receptor are modulated by the pro-fibrotic factor TGF-ß1 in myoblasts and mouse skeletal muscle. This finding suggests that AT-2 might be involved in the physiopathology of fibrosis in dystrophic skeletal muscle.


Asunto(s)
Receptor de Angiotensina Tipo 2/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Mioblastos/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/genética , Sistema Renina-Angiotensina , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología
15.
Int J Biochem Cell Biol ; 44(11): 1993-2002, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22964022

RESUMEN

Fibrotic disorders are typically characterised by excessive connective tissue and extracellular matrix (ECM) deposition that preclude the normal healing of different tissues. Several skeletal muscle dystrophies are characterised by extensive fibrosis. Among the factors involved in skeletal muscle fibrosis is angiotensin II (Ang-II), a key protein of the renin-angiotensin system (RAS). We previously demonstrated that myoblasts responded to Ang-II by increasing the ECM protein levels mediated by AT-1 receptors, implicating an Ang-II-induced reactive oxygen species (ROS) by a NAD(P)H oxidase-dependent mechanism. In this paper, we show that in myoblasts, Ang-II induced the increase of transforming growth factor beta 1 (TGF-ß1) and connective tissue growth factor (CTGF) expression through its AT-1 receptor. This effect is dependent of the NAD(P)H oxidase (NOX)-induced ROS, as indicated by a decrease of the expression of both pro-fibrotic factors when the ROS production was inhibited via the NOX inhibitor apocynin. The increase in pro-fibrotic factors levels was paralleled by enhanced p38MAPK and ERK1/2 phosphorylation in response to Ang-II. However, only the p38MAPK activity was critical for the Ang-II-induced fibrotic effects, as indicated by the decrease in the Ang-II-induced TGF-ß1 and CTGF expression and fibronectin levels by SB-203580, an inhibitor of the p38MAPK, but not by U0126, an inhibitor of ERK1/2 phosphorylation. Furthermore, we showed that the Ang-II-dependent p38MAPK activation, but not the ERK1/2 phosphorylation, was necessary for the NOX-derived ROS. In addition, we demonstrated that TGF-ß1 expression was required for the Ang-II-induced pro-fibrotic effects evaluated by using SB-431542, an inhibitor of TGF-ßRI kinase activity, and by knocking down TGF-ß1 levels by shRNA technique. These results strongly suggest that the fibrotic response to Ang-II is mediated by the AT-1 receptor and requires the p38MAPK phosphorylation, NOX-induced ROS, and TGF-ß1 expression increase mediated by Ang-II in skeletal muscle cells.


Asunto(s)
Angiotensina II/farmacología , Células Musculares/enzimología , Células Musculares/patología , Músculo Esquelético/patología , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibronectinas/metabolismo , Fibrosis , Ratones , Modelos Biológicos , Células Musculares/efectos de los fármacos , NADPH Oxidasas/metabolismo , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/genética
16.
J Biol Chem ; 287(9): 6773-87, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22203668

RESUMEN

Decorin is a small proteoglycan, composed of 12 leucine-rich repeats (LRRs) that modulates the activity of transforming growth factor type ß (TGF-ß) and other growth factors, and thereby influences proliferation and differentiation in a wide array of physiological and pathological processes, such as fibrosis, in several tissues and organs. Previously we described two novel modulators of the TGF-ß-dependent signaling pathway: LDL receptor-related protein (LRP-1) and decorin. Here we have determined the regions in decorin that are responsible for interaction with LRP-1 and are involved in TGF-ß-dependent binding and signaling. Specifically, we used decorin deletion mutants, as well as peptides derived from internal LRR regions, to determine the LRRs responsible for these decorin functions. Our results indicate that LRR6 and LRR5 participate in the interaction with LRP-1 and TGF-ß as well as in its dependent signaling. Furthermore, the internal region (LRR6i), composed of 11 amino acids, is responsible for decorin binding to LRP-1 and subsequent TGF-ß-dependent signaling. Furthermore, using an in vivo approach, we also demonstrate that the LRR6 region of decorin can inhibit TGF-ß mediated action in response to skeletal muscle injury.


Asunto(s)
Decorina/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Músculo Esquelético , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Decorina/química , Decorina/genética , Endocitosis/fisiología , Fibrosis , Gliburida , Humanos , Leucina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
17.
Biochem Biophys Res Commun ; 410(3): 665-70, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21693104

RESUMEN

Fibrotic disorders are typified by excessive connective tissue and extracellular matrix (ECM) deposition that precludes normal healing processes in different tissues. Angiotensin-II (Ang-II) is involved in the fibrotic response. Several muscular dystrophies are characterized by extensive fibrosis. However, the exact role of Ang-II in skeletal muscle fibrosis is unknown. Here we show that myoblasts responded to Ang-II by increasing protein levels of connective tissue growth factor (CTGF/CCN2), collagen-III and fibronectin. These Ang-II-induced pro-fibrotic effects were mediated by AT-1 receptors. Remarkably, Ang-II induced reactive oxygen species (ROS) via a NAD(P)H oxidase-dependent mechanism, as shown by inhibition of ROS production via the NAD(P)H oxidase inhibitors diphenylene iodonium (DPI) and apocynin. This increase in ROS is critical for Ang-II-induced fibrotic effects, as indicated by the decrease in Ang-II-induced CTGF and fibronectin levels by DPI and apocynin. We also show that Ang-II-induced ROS production and fibrosis require PKC activity as indicated by the generic PKC inhibitor chelerythrine. These results strongly suggest that the fibrotic response induced by Ang-II is mediated by AT-1 receptor and requires NAD(P)H-induced ROS in skeletal muscle cells.


Asunto(s)
Angiotensina II/fisiología , Músculo Esquelético/patología , Distrofias Musculares/metabolismo , Mioblastos/patología , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Angiotensina II/farmacología , Animales , Células Cultivadas , Fibrosis , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Distrofias Musculares/patología , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo
18.
J Endocrinol ; 206(1): 75-83, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20453075

RESUMEN

Despite the importance of adipocyte formation for adipose tissue physiology, current knowledge about the mechanisms that regulate the recruitment of progenitor cells to undergo adipogenic differentiation is limited. A role for locally generated angiotensin II emerged from studies with human and murine cells. Preadipose cells from different human fat depots show reduced response to adipogenic stimuli when exposed to angiotensin II. This investigation sought to gain an insight into the intracellular mechanisms involved in the anti-adipogenic response of human preadipose cells from omental fat to angiotensin II. Its effect was evaluated on cells stimulated to adipogenic differentiation in vitro, by assessment of glycerol-3-phosphate dehydrogenase activity and expression of early markers of adipogenesis. Extracellular signal-regulated kinase(1,2) (ERK(1,2)) pathway activation was inferred from the phosphorylated to total ERK(1,2) ratio determined by western blot. Exposure to angiotensin II throughout the 10-day differentiation period resulted in a reduced adipogenic response. A similar anti-adipogenic effect was observed when this hormone was present during the first 48 h of induction to differentiation. Angiotensin II treatment had no consequences on CCAAT/enhancer-binding protein beta and peroxisome proliferator-activated receptor gamma (PPARG) induction, but increased the phosphorylated form of the key adipogenic regulator PPARG. Upon angiotensin II exposure, a raise of phosphorylated ERK(1,2) was determined, which was more prominent 8-20 h after induction of adipogenesis (when controls reached negligible values). Chemical inhibition of ERK(1,2) phosphorylation prevented angiotensin II-dependent reduction in adipogenesis. These results support the participation of the mitogen-activated protein kinase/ERK(1,2) pathway in the anti-adipogenic effect of angiotensin II on preadipose cells from human omental adipose tissue.


Asunto(s)
Adipocitos/citología , Adipogénesis/efectos de los fármacos , Angiotensina II/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , PPAR gamma/metabolismo , Angiotensina II/farmacología , Western Blotting , Células Cultivadas , Medios de Cultivo , Activación Enzimática/efectos de los fármacos , Glicerolfosfato Deshidrogenasa/metabolismo , Humanos , Epiplón , Fosforilación/efectos de los fármacos
19.
J Sex Med ; 7(10): 3414-23, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20456629

RESUMEN

INTRODUCTION: Sexual dysfunction occurs frequently in patients with schizophrenia. AIM: To assess the sexual function of institutionalized patients with schizophrenia and compare it to both that of noninstitutionalized schizophrenic patients as well as patients without any mental illness. METHODS: A cross-sectional comparative analysis between three groups: Group A: 75 patients with schizophrenia and/or schizoaffective disorder according to the International Classification of Diseases, 10th Edition (ICD-10) criteria, admitted into public psychiatric residential facilities; Group B: 41 patients with the same diagnosis who live in the community; Group C: 152 patients with no mental illness attended to by a Primary Care Medical Center. MAIN OUTCOME MEASURES: The Mini-International Neuropsychiatric Interview, Positive and Negative Syndrome Scale for Schizophrenia and Changes in Sexual Function Questionnaire were used to complete the study. RESULTS: When institutionalized patients were compared to the control group, the impairment of sexual function was identified in all areas except Arousal in women and the percentage of dysfunction in Desire-Interest for both sexes. The results for noninstitutionalized schizophrenic males indicated a deficit in Pleasure and Orgasm and in Pleasure and Desire-Frequency for females, when compared with the control group. The overall subscale indicated that 71.2% of males and 57.1% of females in the institutionalized group experienced sexual dysfunction compared with 10% of males and 50% of females in the noninstitutionalized group, and only 12.7% of women in the control group. CONCLUSIONS: Most areas of sexual function are impaired among patients with schizophrenia, especially among institutionalized patients. However, their percentage of sexual dysfunction associated with sexual thoughts or fantasies was similar to that of patients without mental illness.


Asunto(s)
Esquizofrenia/complicaciones , Disfunciones Sexuales Psicológicas/complicaciones , Adolescente , Adulto , Estudios de Casos y Controles , Estudios Transversales , Femenino , Humanos , Pacientes Internos/psicología , Masculino , Estado Civil , Persona de Mediana Edad , Escalas de Valoración Psiquiátrica , Psicología , Trastornos Psicóticos/complicaciones , Trastornos Psicóticos/psicología , Psicología del Esquizofrénico , Disfunciones Sexuales Psicológicas/psicología , Adulto Joven
20.
Obesity (Silver Spring) ; 15(7): 1643-6, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17636081

RESUMEN

OBJECTIVE: The ability to form new adipose cells is important to adipose tissue physiology; however, the mechanisms controlling the recruitment of adipocyte progenitors are poorly understood. A role for locally generated angiotensin II in this process is currently proposed. Given that visceral adipose tissue reportedly expresses higher levels of angiotensinogen compared with other depots and the strong association of augmented visceral fat mass with the adverse consequences of obesity, we studied the role of angiotensin II in regulating adipogenic differentiation in omental fat of obese and non-obese humans. RESEARCH METHODS AND PROCEDURES: The angiotensin II effect on adipose cell formation was evaluated in human omental adipocyte progenitor cells that were stimulated to adipogenic differentiation in vitro. The adipogenic response was measured by the activity of the differentiation marker glycerol-3-phosphate dehydrogenase. RESULTS: Angiotensin II reduced the adipogenic response of adipocyte progenitor cells, and the extent of the decrease correlated directly with the subjects' BMI (p=0.01, R2=0.30). A 56.3+/-3.4% and 44.5+/-2.7% reduction of adipogenesis was found in obese and non-obese donors' cells, respectively (p<0.01). The effect of angiotensin II was reversed by type 1 angiotensin receptor antagonist losartan. DISCUSSION: A greater anti-adipogenic response to angiotensin II in omental adipose progenitor cells from obese subjects opens a venue to understand the deregulation of visceral fat tissue cellularity that has been associated with severe functional abnormalities of the obese condition.


Asunto(s)
Adipocitos/fisiología , Tejido Adiposo/fisiopatología , Angiotensina II/farmacología , Obesidad/fisiopatología , Adipocitos/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Composición Corporal , Diferenciación Celular/efectos de los fármacos , Ingestión de Energía , Metabolismo Energético , Humanos , Epiplón , Células del Estroma/efectos de los fármacos , Células del Estroma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...