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.
Exp Physiol ; 107(11): 1312-1325, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35938289

RESUMEN

NEW FINDINGS: What is the central question of this study? This study addresses whether a high-fat, high-sucrose diet causes cardiac and diaphragm muscle abnormalities in male rats and whether supplementation with the antioxidant N-acetylcysteine reverses diet-induced dysfunction. What is the main finding and its importance? N-Acetylcysteine attenuated the effects of high-fat, high-sucrose diet on markers of cardiac hypertrophy and diastolic dysfunction, but neither high-fat, high-sucrose diet nor N-acetylcysteine affected the diaphragm. These results support the use of N-acetylcysteine to attenuate cardiovascular dysfunction induced by a 'Western' diet. ABSTRACT: Individuals with overweight or obesity display respiratory and cardiovascular dysfunction, and oxidative stress is a causative factor in the general aetiology of obesity and of skeletal and cardiac muscle pathology. Thus, this preclinical study aimed to define diaphragmatic and cardiac morphological and functional alterations in response to an obesogenic diet in rats and the therapeutic potential of an antioxidant supplement, N-acetylcysteine (NAC). Young male Wistar rats consumed ad libitum a 'lean' or high-saturated fat, high-sucrose (HFHS) diet for ∼22 weeks and were randomized to control or NAC (2 mg/ml in the drinking water) for the last 8 weeks of the dietary intervention. We then evaluated diaphragmatic and cardiac morphology and function. Neither HFHS diet nor NAC supplementation affected diaphragm-specific force, peak power or morphology. Right ventricular weight normalized to estimated body surface area, left ventricular fractional shortening and posterior wall maximal shortening velocity were higher in HFHS compared with lean control animals and not restored by NAC. In HFHS rats, the elevated deceleration rate of early transmitral diastolic velocity was prevented by NAC. Our data showed that the HFHS diet did not compromise diaphragmatic muscle morphology or in vitro function, suggesting other possible contributors to breathing abnormalities in obesity (e.g., abnormalities of neuromuscular transmission). However, the HFHS diet resulted in cardiac functional and morphological changes suggestive of hypercontractility and diastolic dysfunction. Supplementation with NAC did not affect diaphragm morphology or function but attenuated some of the cardiac abnormalities in the rats receiving the HFHS diet.


Asunto(s)
Acetilcisteína , Sacarosa , Animales , Masculino , Ratas , Acetilcisteína/uso terapéutico , Antioxidantes/uso terapéutico , Dieta Alta en Grasa , Ácidos Grasos , Obesidad , Ratas Wistar , Músculos Respiratorios
2.
Gene ; 770: 145356, 2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33333219

RESUMEN

The diaphragm is the main inspiratory muscle, and the chronic phase post-myocardial infarction (MI) is characterized by diaphragm morphological, contractile, and metabolic abnormalities. However, the mechanisms of diaphragm weakness are not fully understood. In the current study, we aimed to identify the transcriptome changes associated with diaphragm abnormalities in the chronic stage MI. We ligated the left coronary artery to cause MI in rats and performed RNA-sequencing (RNA-Seq) in diaphragm samples 16 weeks post-surgery. The sham group underwent thoracotomy and pericardiotomy but no artery ligation. We identified 112 differentially expressed genes (DEGs) out of a total of 9664 genes. Myocardial infarction upregulated and downregulated 42 and 70 genes, respectively. Analysis of DEGs in the framework of skeletal muscle-specific biological networks suggest remodeling in the neuromuscular junction, extracellular matrix, sarcomere, cytoskeleton, and changes in metabolism and iron homeostasis. Overall, the data are consistent with pathological remodeling of the diaphragm and reveal potential biological targets to prevent diaphragm weakness in the chronic stage MI.


Asunto(s)
Diafragma/metabolismo , Proteínas Musculares/biosíntesis , Infarto del Miocardio/metabolismo , RNA-Seq , Transcriptoma , Animales , Diafragma/patología , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Ratas , Ratas Sprague-Dawley
3.
J Mol Cell Cardiol ; 139: 238-249, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32035137

RESUMEN

Inspiratory dysfunction occurs in patients with heart failure with reduced ejection fraction (HFrEF) in a manner that depends on disease severity and by mechanisms that are not fully understood. In the current study, we tested whether HFrEF effects on diaphragm (inspiratory muscle) depend on disease severity and examined putative mechanisms for diaphragm abnormalities via global and redox proteomics. We allocated male rats into Sham, moderate (mHFrEF), or severe HFrEF (sHFrEF) induced by myocardial infarction and examined the diaphragm muscle. Both mHFrEF and sHFrEF caused atrophy in type IIa and IIb/x fibers. Maximal and twitch specific forces (N/cm2) were decreased by 19 ± 10% and 28 ± 13%, respectively, in sHFrEF (p < .05), but not in mHFrEF. Global proteomics revealed upregulation of sarcomeric proteins and downregulation of ribosomal and glucose metabolism proteins in sHFrEF. Redox proteomics showed that sHFrEF increased reversibly oxidized cysteine in cytoskeletal and thin filament proteins and methionine in skeletal muscle α-actin (range 0.5 to 3.3-fold; p < .05). In conclusion, fiber atrophy plus contractile dysfunction caused diaphragm weakness in HFrEF. Decreased ribosomal proteins and heighted reversible oxidation of protein thiols are candidate mechanisms for atrophy or anabolic resistance as well as loss of specific force in sHFrEF.


Asunto(s)
Diafragma/metabolismo , Diafragma/fisiopatología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Proteómica , Volumen Sistólico , Actinas/metabolismo , Animales , Masculino , Metionina/metabolismo , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Miofibrillas/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Oxidación-Reducción , Ratas Sprague-Dawley
4.
Am J Physiol Lung Cell Mol Physiol ; 316(4): L679-L690, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30702345

RESUMEN

Heart failure with reduced ejection fraction (HFREF) increases neutral sphingomyelinase (NSMase) activity and mitochondrial reactive oxygen species (ROS) emission and causes diaphragm weakness. We tested whether a systemic pharmacological NSMase inhibitor or short-hairpin RNA (shRNA) targeting NSMase isoform 3 (NSMase3) would prevent diaphragm abnormalities induced by HFREF caused by myocardial infarction. In the pharmacological intervention, we used intraperitoneal injection of GW4869 or vehicle. In the genetic intervention, we injected adeno-associated virus serotype 9 (AAV9) containing shRNA targeting NSMase3 or a scrambled sequence directly into the diaphragm. We also studied acid sphingomyelinase-knockout mice. GW4869 prevented the increase in diaphragm ceramide content, weakness, and tachypnea caused by HFREF. For example, maximal specific forces (in N/cm2) were vehicle [sham 31 ± 2 and HFREF 26 ± 2 ( P < 0.05)] and GW4869 (sham 31 ± 2 and HFREF 31 ± 1). Respiratory rates were (in breaths/min) vehicle [sham 61 ± 3 and HFREF 84 ± 11 ( P < 0.05)] and GW4869 (sham 66 ± 2 and HFREF 72 ± 2). AAV9-NSMase3 shRNA prevented heightening of diaphragm mitochondrial ROS and weakness [in N/cm2, AAV9-scrambled shRNA: sham 31 ± 2 and HFREF 27 ± 2 ( P < 0.05); AAV9-NSMase3 shRNA: sham 30 ± 1 and HFREF 30 ± 1] but displayed tachypnea. Both wild-type and ASMase-knockout mice with HFREF displayed diaphragm weakness. Our study suggests that activation of NSMase3 causes diaphragm weakness in HFREF, presumably through accumulation of ceramide and elevation in mitochondrial ROS. Our data also reveal a novel inhibitory effect of GW4869 on tachypnea in HFREF likely mediated by changes in neural control of breathing.


Asunto(s)
Diafragma/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Debilidad Muscular/prevención & control , ARN Interferente Pequeño/genética , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/genética , Compuestos de Anilina/farmacología , Animales , Compuestos de Bencilideno/farmacología , Diafragma/enzimología , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/terapia , Humanos , Masculino , Ratones , Ratones Noqueados , Debilidad Muscular/genética , Debilidad Muscular/fisiopatología , Ratas , Ratas Wistar , Esfingomielina Fosfodiesterasa/deficiencia , Volumen Sistólico/genética , Volumen Sistólico/fisiología
6.
Exp Gerontol ; 103: 69-79, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29289553

RESUMEN

AIM: Inspiratory muscle (diaphragm) function declines with age, contributing to exercise intolerance and impaired airway clearance. Studies of diaphragm dysfunction in rodents have focused on moderate aging (~24months); thus, the impact of advanced age on the diaphragm and potential mechanisms of dysfunction are less clear. Therefore, we aimed to define the effects of advanced age on the mechanics, morphology, and global and redox proteome of the diaphragm. METHODS: We studied diaphragm from young (6months) and very old male mice (30months). Diaphragm function was evaluated using isolated muscle bundles. Proteome analyses followed LC-MS/MS processing of diaphragm muscle. RESULTS: Advanced aging decreased diaphragm peak power by ~35% and maximal isometric specific force by ~15%, and prolonged time to peak twitch tension by ~30% (P<0.05). These changes in contractile properties were accompanied, and might be caused by, decreases in abundance of calsequestrin, sarcoplasmic reticulum Ca2+-ATPase, sarcalumenin, and parvalbumin that were revealed by our label-free proteomics data. Advanced aging also increased passive stiffness (P<0.05), which might be a consequence of an upregulation of cytoskeletal and extracellular matrix proteins identified by proteomics. Analyses of cysteine redox state indicated that the main diaphragm abnormalities with advanced aging are in metabolic enzymes and mitochondrial proteins. CONCLUSION: Our novel findings are that the most pronounced impact of advanced aging on the diaphragm is loss of peak power and disrupted cysteine redox homeostasis in metabolic enzymes and mitochondrial proteins.


Asunto(s)
Envejecimiento/fisiología , Diafragma/fisiopatología , Mitocondrias/metabolismo , Proteoma/metabolismo , Animales , Cisteína/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Oxidación-Reducción , Condicionamiento Físico Animal , Proteómica
7.
Am J Physiol Regul Integr Comp Physiol ; 310(8): R707-10, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26864813

RESUMEN

Respiratory dysfunction is prevalent in critically ill patients and can lead to adverse clinical outcomes, including respiratory failure and increased mortality. Respiratory muscles, which normally sustain respiration through inspiratory muscle contractions, become weakened during critical illness, and recent studies suggest that respiratory muscle weakness is related to systemic inflammation. Here, we investigate the pathophysiological role of the inflammatory JAK1/3 signaling pathway in diaphragm weakness in two distinct experimental models of critical illness. In the first experiment, mice received subcutaneous injections of PBS or C26 cancer cells and were fed chow formulated with or without the JAK1/3 inhibitor R548 for 26 days. Diaphragm specific force was significantly reduced in tumor-bearing mice receiving standard chow; however, treatment with the JAK1/3 inhibitor completely prevented diaphragm weakness. Diaphragm cross-sectional area was diminished by ∼25% in tumor-bearing mice but was similar to healthy mice in tumor-bearing animals treated with R548. In the second study, mice received sham surgery or coronary artery ligation, leading to myocardial infarction (MI), and were treated with R548 or vehicle 1 h postsurgery, and once daily for 3 days. Diaphragm specific force was comparable between sham surgery/vehicle, sham surgery/R548 and MI/R548 groups, but significantly decreased in the MI/vehicle group. Markers of oxidative damage and activated caspase-3, mechanisms previously identified to reduce muscle contractility, were not elevated in diaphragm extracts. These experiments implicate JAK1/3 signaling in cancer- and MI-mediated diaphragm weakness in mice, and provide a compelling case for further investigation.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Diafragma/efectos de los fármacos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 3/antagonistas & inhibidores , Fuerza Muscular/efectos de los fármacos , Debilidad Muscular/prevención & control , Infarto del Miocardio/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Trastornos Respiratorios/prevención & control , Animales , Caquexia/enzimología , Caquexia/etiología , Caquexia/fisiopatología , Neoplasias del Colon/complicaciones , Neoplasias del Colon/enzimología , Neoplasias del Colon/fisiopatología , Diafragma/enzimología , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Janus Quinasa 1/metabolismo , Janus Quinasa 3/metabolismo , Masculino , Ratones Endogámicos C57BL , Contracción Muscular/efectos de los fármacos , Debilidad Muscular/enzimología , Debilidad Muscular/etiología , Debilidad Muscular/fisiopatología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Respiración/efectos de los fármacos , Trastornos Respiratorios/enzimología , Trastornos Respiratorios/etiología , Trastornos Respiratorios/fisiopatología , Transducción de Señal/efectos de los fármacos
8.
J Appl Physiol (1985) ; 120(7): 733-42, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26846552

RESUMEN

Diaphragm muscle weakness in chronic heart failure (CHF) is caused by elevated oxidants and exacerbates breathing abnormalities, exercise intolerance, and dyspnea. However, the specific source of oxidants that cause diaphragm weakness is unknown. We examined whether mitochondrial reactive oxygen species (ROS) cause diaphragm weakness in CHF by testing the hypothesis that CHF animals treated with a mitochondria-targeted antioxidant have normal diaphragm function. Rats underwent CHF or sham surgery. Eight weeks after surgeries, we administered a mitochondrial-targeted antioxidant (MitoTEMPO; 1 mg·kg(-1)·day(-1)) or sterile saline (Vehicle). Left ventricular dysfunction (echocardiography) pre- and posttreatment and morphological abnormalities were consistent with the presence of CHF. CHF elicited a threefold (P < 0.05) increase in diaphragm mitochondrial H2O2 emission, decreased diaphragm glutathione content by 23%, and also depressed twitch and maximal tetanic force by ∼20% in Vehicle-treated animals compared with Sham (P < 0.05 for all comparisons). Diaphragm mitochondrial H2O2 emission, glutathione content, and twitch and maximal tetanic force were normal in CHF animals receiving MitoTEMPO. Neither CHF nor MitoTEMPO altered the diaphragm protein levels of antioxidant enzymes: superoxide dismutases (CuZn-SOD or MnSOD), glutathione peroxidase, and catalase. In both Vehicle and MitoTEMPO groups, CHF elicited a ∼30% increase in cytochrome c oxidase activity, whereas there were no changes in citrate synthase activity. Our data suggest that elevated mitochondrial H2O2 emission causes diaphragm weakness in CHF. Moreover, changes in protein levels of antioxidant enzymes or mitochondrial content do not seem to mediate the increase in mitochondria H2O2 emission in CHF and protective effects of MitoTEMPO.


Asunto(s)
Diafragma/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Mitocondrias/metabolismo , Debilidad Muscular/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/metabolismo , Diafragma/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Glutatión/metabolismo , Glutatión Peroxidasa , Insuficiencia Cardíaca/metabolismo , Peróxido de Hidrógeno/metabolismo , Debilidad Muscular/metabolismo , Oxidación-Reducción , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar , Superóxidos/metabolismo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
9.
BMC Cancer ; 14: 997, 2014 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-25539728

RESUMEN

BACKGROUND: Evidence from cachectic cancer patients and animal models of cancer cachexia supports the involvement of Forkhead box O (FoxO) transcription factors in driving cancer-induced skeletal muscle wasting. However, the genome-wide gene networks and associated biological processes regulated by FoxO during cancer cachexia are unknown. We hypothesize that FoxO is a central upstream regulator of diverse gene networks in skeletal muscle during cancer that may act coordinately to promote the wasting phenotype. METHODS: To inhibit endogenous FoxO DNA-binding, we transduced limb and diaphragm muscles of mice with AAV9 containing the cDNA for a dominant negative (d.n.) FoxO protein (or GFP control). The d.n.FoxO construct consists of only the FoxO3a DNA-binding domain that is highly homologous to that of FoxO1 and FoxO4, and which outcompetes and blocks endogenous FoxO DNA binding. Mice were subsequently inoculated with Colon-26 (C26) cells and muscles harvested 26 days later. RESULTS: Blocking FoxO prevented C26-induced muscle fiber atrophy of both locomotor muscles and the diaphragm and significantly spared force deficits. This sparing of muscle size and function was associated with the differential regulation of 543 transcripts (out of 2,093) which changed in response to C26. Bioinformatics analysis of upregulated gene transcripts that required FoxO revealed enrichment of the proteasome, AP-1 and IL-6 pathways, and included several atrophy-related transcription factors, including Stat3, Fos, and Cebpb. FoxO was also necessary for the cancer-induced downregulation of several gene transcripts that were enriched for extracellular matrix and sarcomere protein-encoding genes. We validated these findings in limb muscles and the diaphragm through qRT-PCR, and further demonstrate that FoxO1 and/or FoxO3a are sufficient to increase Stat3, Fos, Cebpb, and the C/EBPß target gene, Ubr2. Analysis of the Cebpb proximal promoter revealed two bona fide FoxO binding elements, which we further establish are necessary for Cebpb promoter activation in response to IL-6, a predominant cytokine in the C26 cancer model. CONCLUSIONS: These findings provide new evidence that FoxO-dependent transcription is a central node controlling diverse gene networks in skeletal muscle during cancer cachexia, and identifies novel candidate genes and networks for further investigation as causative factors in cancer-induced wasting.


Asunto(s)
Caquexia/etiología , Neoplasias del Colon/complicaciones , Neoplasias del Colon/genética , Factores de Transcripción Forkhead/metabolismo , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Músculo Esquelético/metabolismo , Secuencia de Aminoácidos , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Vectores Genéticos/genética , Xenoinjertos , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Complejo de la Endopetidasa Proteasomal/metabolismo , Reproducibilidad de los Resultados , Alineación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transducción Genética
11.
J Cell Sci ; 127(Pt 7): 1441-53, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24463822

RESUMEN

The Forkhead box O (FoxO) transcription factors are activated, and necessary for the muscle atrophy, in several pathophysiological conditions, including muscle disuse and cancer cachexia. However, the mechanisms that lead to FoxO activation are not well defined. Recent data from our laboratory and others indicate that the activity of FoxO is repressed under basal conditions via reversible lysine acetylation, which becomes compromised during catabolic conditions. Therefore, we aimed to determine how histone deacetylase (HDAC) proteins contribute to activation of FoxO and induction of the muscle atrophy program. Through the use of various pharmacological inhibitors to block HDAC activity, we demonstrate that class I HDACs are key regulators of FoxO and the muscle-atrophy program during both nutrient deprivation and skeletal muscle disuse. Furthermore, we demonstrate, through the use of wild-type and dominant-negative HDAC1 expression plasmids, that HDAC1 is sufficient to activate FoxO and induce muscle fiber atrophy in vivo and is necessary for the atrophy of muscle fibers that is associated with muscle disuse. The ability of HDAC1 to cause muscle atrophy required its deacetylase activity and was linked to the induction of several atrophy genes by HDAC1, including atrogin-1, which required deacetylation of FoxO3a. Moreover, pharmacological inhibition of class I HDACs during muscle disuse, using MS-275, significantly attenuated both disuse muscle fiber atrophy and contractile dysfunction. Together, these data solidify the importance of class I HDACs in the muscle atrophy program and indicate that class I HDAC inhibitors are feasible countermeasures to impede muscle atrophy and weakness.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Histona Desacetilasa 1/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Acetilación , Animales , Histona Desacetilasa 1/genética , Humanos , Masculino , Ratones , Músculo Esquelético/patología , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Transducción de Señal
12.
PLoS One ; 8(4): e62687, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23626847

RESUMEN

Skeletal muscle regeneration following injury is a highly coordinated process that involves transient muscle inflammation, removal of necrotic cellular debris and subsequent replacement of damaged myofibers through secondary myogenesis. However, the molecular mechanisms which coordinate these events are only beginning to be defined. In the current study we demonstrate that Heat shock protein 70 (Hsp70) is increased following muscle injury, and is necessary for the normal sequence of events following severe injury induced by cardiotoxin, and physiological injury induced by modified muscle use. Indeed, Hsp70 ablated mice showed a significantly delayed inflammatory response to muscle injury induced by cardiotoxin, with nearly undetected levels of both neutrophil and macrophage markers 24 hours post-injury. At later time points, Hsp70 ablated mice showed sustained muscle inflammation and necrosis, calcium deposition and impaired fiber regeneration that persisted several weeks post-injury. Through rescue experiments reintroducing Hsp70 intracellular expression plasmids into muscles of Hsp70 ablated mice either prior to injury or post-injury, we confirm that Hsp70 optimally promotes muscle regeneration when expressed during both the inflammatory phase that predominates in the first four days following severe injury and the regenerative phase that predominates thereafter. Additional rescue experiments reintroducing Hsp70 protein into the extracellular microenvironment of injured muscles at the onset of injury provides further evidence that Hsp70 released from damaged muscle may drive the early inflammatory response to injury. Importantly, following induction of physiological injury through muscle reloading following a period of muscle disuse, reduced inflammation in 3-day reloaded muscles of Hsp70 ablated mice was associated with preservation of myofibers, and increased muscle force production at later time points compared to WT. Collectively our findings indicate that depending on the nature and severity of muscle injury, therapeutics which differentially target both intracellular and extracellular localized Hsp70 may optimally preserve muscle tissue and promote muscle functional recovery.


Asunto(s)
Proteínas HSP70 de Choque Térmico/genética , Inflamación/genética , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología , Regeneración/genética , Animales , Calcinosis/genética , Calcinosis/patología , Proteínas HSP70 de Choque Térmico/metabolismo , Inflamación/inmunología , Inflamación/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/inmunología , Músculo Esquelético/patología , Factores de Tiempo
13.
PLoS One ; 8(4): e62702, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23614054

RESUMEN

Pulmonary hypertension (PH) causes loss of body weight and inspiratory (diaphragm) muscle dysfunction. A model of PH induced by drug (monocrotaline, MCT) has been extensively used in mice to examine the etiology of PH. However, it is unclear if PH induced by MCT in mice reproduces the loss of body weight and diaphragm muscle dysfunction seen in patients. This is a pre-requisite for widespread use of mice to examine mechanisms of cachexia and diaphragm abnormalities in PH. Thus, we measured body and soleus muscle weight, food intake, and diaphragm contractile properties in mice after 6-8 weeks of saline (control) or MCT (600 mg/kg) injections. Body weight progressively decreased in PH mice, while food intake was similar in both groups. PH decreased (P<0.05) diaphragm maximal isometric specific force, maximal shortening velocity, and peak power. Protein carbonyls in whole-diaphragm lysates and the abundance of select myofibrillar proteins were unchanged by PH. Our findings show diaphragm isometric and isotonic contractile abnormalities in a murine model of PH induced by MCT. Overall, the murine model of PH elicited by MCT mimics loss of body weight and diaphragm muscle weakness reported in PH patients.


Asunto(s)
Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Contracción Muscular , Animales , Atrofia/complicaciones , Peso Corporal/efectos de los fármacos , Diafragma/efectos de los fármacos , Diafragma/patología , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Monocrotalina/farmacología , Monocrotalina/uso terapéutico , Contracción Muscular/efectos de los fármacos
14.
FASEB J ; 27(7): 2600-10, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23515443

RESUMEN

Cancer cachexia is characterized by a continuous loss of locomotor skeletal muscle mass, which causes profound muscle weakness. If this atrophy and weakness also occurs in diaphragm muscle, it could lead to respiratory failure, which is a major cause of death in patients with cancer. Thus, the purpose of the current study was to determine whether colon-26 (C-26) cancer cachexia causes diaphragm muscle fiber atrophy and weakness and compromises ventilation. All diaphragm muscle fiber types were significantly atrophied in C-26 mice compared to controls, and the atrophy-related genes, atrogin-1 and MuRF1, significantly increased. Maximum isometric specific force of diaphragm strips, absolute maximal calcium activated force, and maximal specific calcium-activated force of permeabilized diaphragm fibers were all significantly decreased in C-26 mice compared to controls. Further, isotonic contractile properties of the diaphragm were affected to an even greater extent than isometric function. Ventilation measurements demonstrated that C-26 mice have a significantly lower tidal volume compared to controls under basal conditions and, unlike control mice, an inability to increase breathing frequency, tidal volume, and, thus, minute ventilation in response to a respiratory challenge. These data demonstrate that C-26 cancer cachexia causes profound respiratory muscle atrophy and weakness and ventilatory dysfunction.


Asunto(s)
Caquexia/fisiopatología , Neoplasias del Colon/fisiopatología , Diafragma/fisiopatología , Atrofia Muscular/fisiopatología , Insuficiencia Respiratoria/fisiopatología , Actinas/metabolismo , Animales , Western Blotting , Caquexia/etiología , Línea Celular Tumoral , Neoplasias del Colon/complicaciones , Diafragma/metabolismo , Diafragma/patología , Expresión Génica , Inmunohistoquímica , Ratones , Proteínas Musculares/genética , Debilidad Muscular/etiología , Debilidad Muscular/fisiopatología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Atrofia Muscular/etiología , Cadenas Pesadas de Miosina/metabolismo , Insuficiencia Respiratoria/etiología , Músculos Respiratorios/metabolismo , Músculos Respiratorios/patología , Músculos Respiratorios/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas de Motivos Tripartitos , Tropomiosina/metabolismo , Troponina/metabolismo , Ubiquitina-Proteína Ligasas/genética
15.
Med Hypotheses ; 79(5): 673-7, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22939314

RESUMEN

Despite remarkable effectiveness of reperfusion and drug therapies to reduce morbidity and mortality following myocardial infarction (MI), many patients have debilitating symptoms and impaired left ventricular (LV) function highlighting the need for improved post-MI therapies. A promising concept currently under investigation is intramyocardial injection of high-water content, polymeric biomaterial gels (e.g., hydrogels) to modulate myocardial scar formation and LV adverse remodeling. We propose a degradable, bioactive hydrogel that forms a unique microstructure of continuous, parallel capillary-like channels (Capgel). We hypothesize that the innovative architecture and composition of Capgel can serve as a platform for endogenous cell recruitment and drug/cell delivery, therefore facilitating myocardial repair after MI.


Asunto(s)
Alginatos/química , Infarto del Miocardio/terapia , Factor de Células Madre/administración & dosificación , Trasplante de Células Madre , Humanos , Hidrogeles , Microscopía Electrónica de Rastreo
16.
Int J Sport Nutr Exerc Metab ; 21(2): 146-54, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21558576

RESUMEN

UNLABELLED: N-acetylcysteine (NAC) is a thiol donor with antioxidant properties that has potential use as an ergogenic aid. However, NAC is associated with adverse reactions that limit its use in humans. PURPOSE: The authors evaluated NAC efficacy as a thiol donor before handgrip exercise, measuring changes in serum cysteine and glutathione status and recording adverse reactions in adult subjects across a range of doses. METHODS: Healthy individuals ingested NAC capsules (9 ± 2 or 18 ± 4 mg/kg) or solution (0, 35, 70, or 140 mg/kg). Venous blood samples were collected and subjects answered a questionnaire about adverse reactions. RESULTS: Low doses of NAC (capsules) did not affect plasma cysteine or glutathione or cause adverse reactions. Adverse reactions to NAC solution were predominantly mild and gastrointestinal (GI). Intensity of GI reactions to 140 mg/kg NAC was significantly higher than placebo (in a.u., 0.67 ± 0.16 vs. 0.07 ± 0.04; p < .05). Plasma cysteine concentration increased with NAC dose from 9.3 ± 0.7 µM (placebo) to 65.3 ± 6.7 µM (140 mg/kg); however, there was no difference (p > .05) in plasma cysteine for 70 mg/kg vs. 140 mg/kg. Similar increases were observed for the ratio of cysteine to total cysteine, which was directly related to handgrip exercise performance. Plasma glutathione was elevated and oxidized glutathione diminished (p < .05) with NAC 140 mg/kg vs. placebo. CONCLUSION: NAC effects on plasma thiols are maximized by oral administration of 70 mg/kg, a dose that does not cause significant adverse reactions.


Asunto(s)
Acetilcisteína/administración & dosificación , Fuerza de la Mano/fisiología , Compuestos de Sulfhidrilo/sangre , Acetilcisteína/efectos adversos , Acetilcisteína/metabolismo , Adulto , Rendimiento Atlético/fisiología , Estudios Cruzados , Cisteína/sangre , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Glutatión/sangre , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
17.
BMC Musculoskelet Disord ; 12: 15, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21235761

RESUMEN

BACKGROUND: Muscle weakness is associated with a variety of chronic disorders such as emphysema (EMP) and congestive heart failure (CHF) as well as aging. Therapies to treat muscle weakness associated with chronic disease or aging are lacking. Corticotrophin releasing factor 2 receptor (CRF2R) agonists have been shown to maintain skeletal muscle mass and force production in a variety of acute conditions that lead to skeletal muscle wasting. HYPOTHESIS: We hypothesize that treating animals with a CRF2R agonist will maintain skeletal muscle mass and force production in animals with chronic disease and in aged animals. METHODS: We utilized animal models of aging, CHF and EMP to evaluate the potential of CRF2R agonist treatment to maintain skeletal muscle mass and force production in aged animals and animals with CHF and EMP. RESULTS: In aged rats, we demonstrate that treatment with a CRF2R agonist for up to 3 months results in greater extensor digitorum longus (EDL) force production, EDL mass, soleus mass and soleus force production compared to age matched untreated animals. In the hamster EMP model, we demonstrate that treatment with a CRF2R agonist for up to 5 months results in greater EDL force production in EMP hamsters when compared to vehicle treated EMP hamsters and greater EDL mass and force in normal hamsters when compared to vehicle treated normal hamsters. In the rat CHF model, we demonstrate that treatment with a CRF2R agonist for up to 3 months results in greater EDL and soleus muscle mass and force production in CHF rats and normal rats when compared to the corresponding vehicle treated animals. CONCLUSIONS: These data demonstrate that the underlying physiological conditions associated with chronic diseases such as CHF and emphysema in addition to aging do not reduce the potential of CRF2R agonists to maintain skeletal muscle mass and force production.


Asunto(s)
Envejecimiento/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Fuerza Muscular/efectos de los fármacos , Debilidad Muscular/tratamiento farmacológico , Músculo Esquelético/efectos de los fármacos , Péptidos/uso terapéutico , Receptores de Hormona Liberadora de Corticotropina/agonistas , Envejecimiento/fisiología , Animales , Enfermedad Crónica , Cricetinae , Dinamarca , Modelos Animales de Enfermedad , Femenino , Masculino , Mesocricetus , Contracción Muscular/fisiología , Fuerza Muscular/fisiología , Debilidad Muscular/etiología , Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiología , Músculo Esquelético/fisiopatología , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas Lew , Receptores de Hormona Liberadora de Corticotropina/fisiología
18.
Am J Physiol Lung Cell Mol Physiol ; 300(2): L225-31, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21097524

RESUMEN

Doxorubicin, a common chemotherapeutic agent, causes respiratory muscle weakness in both patients and rodents. Tumor necrosis factor-α (TNF), a proinflammatory cytokine that depresses diaphragm force, is elevated following doxorubicin chemotherapy. TNF-induced diaphragm weakness is mediated through TNF type 1 receptor (TNFR1). These findings lead us to hypothesize that TNF/TNFR1 signaling mediates doxorubicin-induced diaphragm muscle weakness. We tested this hypothesis by treating C57BL/6 mice with a clinical dose of doxorubicin (20 mg/kg) via intravenous injection. Three days later, we measured contractile properties of muscle fiber bundles isolated from the diaphragm. We tested the involvement of TNF/TNFR1 signaling using pharmaceutical and genetic interventions. Etanercept, a soluble TNF receptor, and TNFR1 deficiency protected against the depression in diaphragm-specific force caused by doxorubicin. Doxorubicin stimulated an increase in TNFR1 mRNA and protein (P < 0.05) in the diaphragm, along with colocalization of TNFR1 to the plasma membrane. These results suggest that doxorubicin increases diaphragm sensitivity to TNF by upregulating TNFR1, thereby causing respiratory muscle weakness.


Asunto(s)
Diafragma/efectos de los fármacos , Diafragma/fisiopatología , Doxorrubicina/efectos adversos , Debilidad Muscular/inducido químicamente , Debilidad Muscular/fisiopatología , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Antineoplásicos/efectos adversos , Secuencia de Bases , Cartilla de ADN/genética , Etanercept , Inmunoglobulina G/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Debilidad Muscular/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Factor de Necrosis Tumoral , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba/efectos de los fármacos
19.
Med Sci Sports Exerc ; 43(6): 1025-31, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20980926

RESUMEN

UNLABELLED: Reactions involving thiol biochemistry seem to play a crucial role in skeletal muscle fatigue. N-acetylcysteine amide (NACA) and L-ergothioneine (ERGO) are thiol-based antioxidants available for human use that have not been evaluated for effects on muscle fatigue. PURPOSE: To test the hypothesis that NACA and ERGO delay skeletal muscle fatigue. METHODS: We exposed mouse diaphragm fiber bundles to buffer (CTRL), NACA, ERGO, or N-acetylcysteine (NAC; positive control). Treatments were performed in vitro using 10 mM for 60 min at 37 °C. After treatment, we determined the muscle force-frequency and fatigue characteristics. RESULTS: The force-frequency relationship was shifted to the left by ERGO and to the right by NACA compared with CTRL and NAC. Maximal tetanic force was similar among groups. The total force-time integral (FTI; N · s · cm) during the fatigue trial was decreased by NACA (420 ± 35, P < 0.05), unaffected by ERGO (657 ± 53), and increased by NAC (P < 0.05) compared with CTRL (581 ± 54). The rate of contraction (dF/dtMAX) during the fatigue trial was not affected by any of the treatments tested. NAC, but not NACA or ERGO, delayed the slowing of muscle relaxation (dF/dtMIN) during fatigue. CONCLUSIONS: In summary, NACA and ERGO did not delay skeletal muscle fatigue in vitro. We conclude that these antioxidants are unlikely to improve human exercise performance.


Asunto(s)
Acetilcisteína/análogos & derivados , Antioxidantes/farmacología , Diafragma/efectos de los fármacos , Ergotioneína/farmacología , Fatiga Muscular/efectos de los fármacos , Acetilcisteína/farmacología , Animales , Ratones , Ratones Endogámicos C57BL , Relajación Muscular/efectos de los fármacos
20.
Am J Physiol Cell Physiol ; 299(3): C552-60, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20519448

RESUMEN

Sphingomyelinase (SMase) hydrolyzes membrane sphingomyelin into ceramide, which increases oxidants in nonmuscle cells. Serum SMase activity is elevated in sepsis and heart failure, conditions where muscle oxidants are increased, maximal muscle force is diminished, and fatigue is accelerated. We tested the hypotheses that exogenous SMase and accumulation of ceramide in muscle increases oxidants in muscle cells, depresses specific force of unfatigued muscle, and accelerates the fatigue process. We also anticipated that the antioxidant N-acetylcysteine (NAC) would prevent SMase effects on muscle function. We studied the responses of C2C12 myotubes and mouse diaphragm to SMase treatment in vitro. We observed that SMase caused a 2.8-fold increase in total ceramide levels in myotubes. Exogenous ceramide and SMase elevated oxidant activity in C2C12 myotubes by 15-35% (P < 0.05) and in diaphragm muscle fiber bundles by 58-120% (P < 0.05). The SMase-induced increase in diaphragm oxidant activity was prevented by NAC. Exogenous ceramide depressed diaphragm force by 55% (P < 0.05), while SMase depressed maximal force by 30% (P < 0.05) and accelerated fatigue--effects opposed by treatment with NAC. In conclusion, our findings suggest that SMase stimulates a ceramide-oxidant signaling pathway that results in muscle weakness and fatigue.


Asunto(s)
Fatiga Muscular/fisiología , Músculo Esquelético/fisiología , Oxidantes/fisiología , Esfingomielina Fosfodiesterasa/fisiología , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Proteínas Bacterianas/farmacología , Línea Celular , Ceramidas/metabolismo , Citosol/metabolismo , Diafragma/fisiología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Transducción de Señal , Esfingomielina Fosfodiesterasa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA