Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Nutr ; 43(7): 1800-1808, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38861892

RESUMEN

BACKGROUND & AIMS: Our study aims to determine whether myostatin (MSTN) is associated with muscle mass and strength in individuals with cancer or obesity, as well as with cancer cachexia (CC) or sarcopenic obesity (SO). METHODS: The ACTICA study included individuals with CC (n = 70) or without CC (NC, n = 73). The MYDIASECRET study included individuals with obesity evaluated before (T0) and 3 months (T3) after bariatric surgery (n = 62). Body composition was assessed using bioelectrical impedance analysis (BIA). Skeletal muscle mass (SMM) and appendicular SMM (ASMM) were calculated from Janssen's and Sergi's equations, respectively, and expressed as indexes (SMMI and ASMMI). Handgrip strength (HGS) was assessed using a Jamar hand-held dynamometer. MSTN plasma levels were measured using ELISA. Spearman's coefficient was used to correlate MSTN with muscle mass and strength. Receiver operating characteristic (ROC) curve analysis was performed to identify an optimal MSTN cutoff level for the prediction of CC or SO. RESULTS: In the ACTICA study, muscle mass and strength were lower in CC individuals than in NC individuals (SMMI: 8.0 kg/m2vs 9.0 kg/m2, p = 0.004; ASMMI: 6.2 kg/m2vs 7.2 kg/m2, p < 0.001; HGS: 28 kg vs 38 kg, p < 0.001). MSTN was also lower in CC individuals than in NC individuals (1434 pg/mL vs 2149 pg/mL, p < 0.001). Muscle mass and strength were positively correlated with MSTN (SMMI: R = 0.500, p < 0.001; ASMMI: R = 0.479, p < 0.001; HGS: R = 0.495, p < 0.001). ROC curve analysis showed a MSTN cutoff level of 1548 pg/mL (AUC 0.684, sensitivity 57%, specificity 75%, p < 0.001) for the prediction of CC. In the MYDIASECRET study, muscle mass and strength were reduced at T3 (SMMI: -8%, p < 0.001; ASMMI: -12%, p < 0.001; HGS: -6%, p = 0.005). MSTN was also reduced at T3 (1773 pg/mL vs 2582 pg/mL, p < 0.001). Muscle mass and strength were positively correlated with MSTN at T0 and T3 (SMMI-T0: R = 0.388, p = 0.002; SMMI-T3: R = 0.435, p < 0.001; HGS-T0: R = 0.337, p = 0.007; HGS-T3: R = 0.313, p = 0.013). ROC curve analysis showed a MSTN cutoff level of 4225 pg/mL (AUC 0.835, sensitivity 98%, specificity 100%, p = 0.014) for the prediction of SO at T3. CONCLUSIONS: MSTN is positively correlated with muscle mass and strength in individuals with cancer or obesity, suggesting its potential use as a biomarker of muscle mass and strength. The ROC curve analysis suggests the potential use of MSTN as a screening tool for CC and SO.


Asunto(s)
Biomarcadores , Caquexia , Fuerza de la Mano , Músculo Esquelético , Miostatina , Neoplasias , Obesidad , Sarcopenia , Humanos , Miostatina/sangre , Masculino , Femenino , Neoplasias/sangre , Neoplasias/complicaciones , Neoplasias/fisiopatología , Músculo Esquelético/fisiopatología , Persona de Mediana Edad , Obesidad/sangre , Obesidad/fisiopatología , Obesidad/complicaciones , Caquexia/sangre , Caquexia/etiología , Caquexia/fisiopatología , Biomarcadores/sangre , Sarcopenia/sangre , Sarcopenia/etiología , Sarcopenia/fisiopatología , Fuerza de la Mano/fisiología , Composición Corporal , Anciano , Fuerza Muscular/fisiología , Adulto , Impedancia Eléctrica
2.
J Cachexia Sarcopenia Muscle ; 15(3): 919-933, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38572511

RESUMEN

BACKGROUND: Cancer cachexia is a life-threatening, inflammation-driven wasting syndrome that remains untreatable. Adiponectin, the most abundant adipokine, plays an important role in several metabolic processes as well as in inflammation modulation. Our aim was to test whether administration of AdipoRon (AR), a synthetic agonist of the adiponectin receptors, prevents the development of cancer cachexia and its related muscle atrophy. METHODS: The effect of AR on cancer cachexia was investigated in two distinct murine models of colorectal cancer. First, 7-week-old CD2F1 male mice were subcutaneously injected with colon-26 carcinoma cells (C26) or vehicle (CT). Six days after injection, mice were treated for 5 days with AdipoRon (50 mg/kg/day; C26 + AR) or the corresponding vehicle (CT and C26). Additionally, a genetic model, the ApcMin/+ mouse, that develops spontaneously numerous intestinal polyps, was used. Eight-week-old male ApcMin/+ mice were treated with AdipoRon (50 mg/kg/day; Apc + AR) or the corresponding vehicle (Apc) over a period of 12 weeks, with C57BL/6J wild-type mice used as controls. In both models, several parameters were assessed in vivo: body weight, grip strength and serum parameters, as well as ex vivo: molecular changes in muscle, fat and liver. RESULTS: The protective effect of AR on cachexia development was observed in both cachectic C26 and ApcMin/+ mice. In these mice, AR administration led to a significant alleviation of body weight loss and muscle wasting, together with rescued muscle strength (P < 0.05 for all). In both models, AR had a strong anti-inflammatory effect, reflected by lower systemic interleukin-6 levels (-55% vs. C26, P < 0.001 and -80% vs. Apc mice, P < 0.05), reduced muscular inflammation as indicated by lower levels of Socs3, phospho-STAT3 and Serpina3n, an acute phase reactant (P < 0.05 for all). In addition, AR blunted circulating levels of corticosterone (-46% vs. C26 mice, P < 0.001 and -60% vs. Apc mice, P < 0.05), the predominant murine glucocorticoid known to induce muscle atrophy. Accordingly, key glucocorticoid-responsive factors implicated in atrophy programmes were-or tended to be-significantly blunted in skeletal muscle by AR. Finally, AR protected against lipid metabolism alterations observed in ApcMin/+ mice, as it mitigated the increase in circulating triglyceride levels (-38%, P < 0.05) by attenuating hepatic triglyceride synthesis and fatty acid uptake by the liver. CONCLUSIONS: Altogether, these results show that AdipoRon rescued the cachectic phenotype by alleviating body weight loss and muscle atrophy, along with restraining inflammation and hypercorticism in preclinical murine models. Therefore, AdipoRon could represent an innovative therapeutic strategy to counteract cancer cachexia.


Asunto(s)
Caquexia , Inflamación , Receptores de Adiponectina , Animales , Caquexia/etiología , Caquexia/tratamiento farmacológico , Caquexia/metabolismo , Ratones , Receptores de Adiponectina/agonistas , Receptores de Adiponectina/metabolismo , Masculino , Inflamación/tratamiento farmacológico , Modelos Animales de Enfermedad , Línea Celular Tumoral , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Piperidinas
3.
Eur J Endocrinol ; 189(3): 409-421, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37638789

RESUMEN

IMPORTANCE AND OBJECTIVE: The identification of myokines susceptible to improve glucose homeostasis following bariatric surgery could lead to new therapeutic approaches for type 2 diabetes. METHODS: Changes in the homeostasis model assessment (HOMA) test were assessed in patients before and 3 months after bariatric surgery. Changes in myokines expression and circulating levels were assessed using real-time quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). Myokines known to regulate glucose homeostasis were identified using literature (targeted study) and putative myokines using RNA-sequencing (untargeted study). A linear regression analysis adjusted for age and sex was used to search for associations between changes in the HOMA test and changes in myokines. RESULTS: In the targeted study, brain-derived neurotrophic factor (BDNF) expression was upregulated (+30%, P = .006) while BDNF circulating levels were decreased (-12%, P = .001). Upregulated BDNF expression was associated with decreased HOMA of insulin resistance (HOMA-IR) (adjusted estimate [95% confidence interval {CI}]: -0.51 [-0.88 to -0.13], P = .010). Decreased BDNF serum levels were associated with decreased HOMA of beta-cell function (HOMA-B) (adjusted estimate [95% CI] = 0.002 [0.00002-0.0031], P = .046). In the untargeted study, upregulated putative myokines included XYLT1 (+64%, P < .001), LGR5 (+57, P< .001), and SPINK5 (+46%, P < .001). Upregulated LGR5 was associated with decreased HOMA-IR (adjusted estimate [95% CI] = -0.50 [-0.86 to -0.13], P = .009). Upregulated XYLT1 and SPINK5 were associated with increased HOMA of insulin sensitivity (HOMA-S) (respectively, adjusted estimate [95% CI] = 109.1 [28.5-189.8], P = .009 and 16.5 [0.87-32.19], P = .039). CONCLUSIONS: Improved glucose homeostasis following bariatric surgery is associated with changes in myokines expression and circulating levels. In particular, upregulation of BDNF, XYLT1, SPINK5, and LGR5 is associated with improved insulin sensitivity. These results suggest that these myokines could contribute to improved glucose homeostasis following bariatric surgery. STUDY REGISTRATION: NCT03341793 on ClinicalTrials.gov (https://clinicaltrials.gov/).


Asunto(s)
Cirugía Bariátrica , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Humanos , Factor Neurotrófico Derivado del Encéfalo , Diabetes Mellitus Tipo 2/cirugía , Glucosa
4.
Nature ; 617(7962): 827-834, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37165186

RESUMEN

Skeletal muscle atrophy is a hallmark of the cachexia syndrome that is associated with poor survival and reduced quality of life in patients with cancer1. Muscle atrophy involves excessive protein catabolism and loss of muscle mass and strength2. An effective therapy against muscle wasting is currently lacking because mechanisms driving the atrophy process remain incompletely understood. Our gene expression analysis in muscle tissues indicated upregulation of ectodysplasin A2 receptor (EDA2R) in tumour-bearing mice and patients with cachectic cancer. Here we show that activation of EDA2R signalling promotes skeletal muscle atrophy. Stimulation of primary myotubes with the EDA2R ligand EDA-A2 triggered pronounced cellular atrophy by induction of the expression of muscle atrophy-related genes Atrogin1 and MuRF1. EDA-A2-driven myotube atrophy involved activation of the non-canonical NFĸB pathway and was dependent on NFκB-inducing kinase (NIK) activity. Whereas EDA-A2 overexpression promoted muscle wasting in mice, deletion of either EDA2R or muscle NIK protected tumour-bearing mice from loss of muscle mass and function. Tumour-induced oncostatin M (OSM) upregulated muscle EDA2R expression, and muscle-specific oncostatin M receptor (OSMR)-knockout mice were resistant to tumour-induced muscle wasting. Our results demonstrate that EDA2R-NIK signalling mediates cancer-associated muscle atrophy in an OSM-OSMR-dependent manner. Thus, therapeutic targeting of these pathways may be beneficial in prevention of muscle loss.


Asunto(s)
Caquexia , Atrofia Muscular , Neoplasias , Transducción de Señal , Receptor Xedar , Animales , Ratones , Caquexia/complicaciones , Caquexia/etiología , Caquexia/metabolismo , Caquexia/patología , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Atrofia Muscular/prevención & control , Neoplasias/complicaciones , Neoplasias/metabolismo , Neoplasias/patología , Receptor Xedar/metabolismo , Humanos , Ligandos , Receptores de Oncostatina M/metabolismo , Oncostatina M/metabolismo , Quinasa de Factor Nuclear kappa B
5.
Cells ; 11(7)2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-35406681

RESUMEN

Activin A (ActA) is considered to play a major role in cancer-induced cachexia (CC). Indeed, circulating ActA levels are elevated and predict survival in patients with CC. However, the mechanisms by which ActA mediates CC development and in particular skeletal muscle (SM) atrophy in humans are not yet fully understood. In this work, we aimed to investigate the effects of ActA on human SM and in mouse models of CC. We used a model of human muscle cells in culture to explore how ActA acts towards human SM. In this model, recombinant ActA induced myotube atrophy associated with the decline of MyHC-ß/slow, the main myosin isoform in human muscle cells studied. Moreover, ActA inhibited the expression and activity of MEF2C, the transcription factor regulating MYH7, the gene which codes for MyHC-ß/slow. This decrease in MEF2C was involved in the decline of MyHC-ß/slow expression, since inhibition of MEF2C by a siRNA leads to the decrease in MyHC-ß/slow expression. The relevance of this ActA/MEF2C pathway in vivo was supported by the parallel decline of MEF2C expression and SM mass, which are both blunted by ActA inhibition, in animal models of CC. In this work, we showed that ActA is a potent negative regulator of SM mass by inhibiting MyHC-ß/slow synthesis through downregulation of MEF2C. This observation highlights a novel interaction between ActA signaling and MEF2C transcriptional activity which contributes to SM atrophy in CC models.


Asunto(s)
Activinas , Factores de Transcripción MEF2 , Atrofia Muscular , Enfermedades Musculares , Animales , Caquexia/metabolismo , Humanos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Desarrollo de Músculos/genética
6.
Front Physiol ; 12: 677746, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220542

RESUMEN

Skeletal muscle, the most abundant tissue in the body, plays vital roles in locomotion and metabolism. Understanding the cellular processes that govern regulation of muscle mass and function represents an essential step in the development of therapeutic strategies for muscular disorders. Myostatin, a member of the TGF-ß family, has been identified as a negative regulator of muscle development. Indeed, its inhibition induces an extensive skeletal muscle hypertrophy requiring the activation of Smad 1/5/8 and the Insulin/IGF-I signaling pathway, but whether other molecular mechanisms are involved in this process remains to be determined. Using transcriptomic data from various Myostatin inhibition models, we identified Pak1 as a potential mediator of Myostatin action on skeletal muscle mass. Our results show that muscle PAK1 levels are systematically increased in response to Myostatin inhibition, parallel to skeletal muscle mass, regardless of the Myostatin inhibition model. Using Pak1 knockout mice, we investigated the role of Pak1 in the skeletal muscle hypertrophy induced by different approaches of Myostatin inhibition. Our findings show that Pak1 deletion does not impede the skeletal muscle hypertrophy magnitude in response to Myostatin inhibition. Therefore, Pak1 is permissive for the skeletal muscle mass increase caused by Myostatin inhibition.

7.
Cancers (Basel) ; 12(11)2020 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-33142864

RESUMEN

Loss of skeletal muscle mass in cancer cachexia is recognized as a predictor of mortality. This study aimed to characterize the changes in the muscle secretome associated with cancer cachexia to gain a better understanding of the mechanisms involved and to identify secreted proteins which may reflect this wasting process. The changes in the muscle proteome of the C26 model were investigated by label-free proteomic analysis followed by a bioinformatic analysis in order to identify potentially secreted proteins. Multiple reaction monitoring and Western blotting were used to verify the presence of candidate proteins in the circulation. Our results revealed a marked increased muscular production of several acute phase reactants (APR: Haptoglobin, Serine protease inhibitor A3N, Complement C3, Serum amyloid A-1 protein) which are released in the circulation during C26 cancer cachexia. This was confirmed in other models of cancer cachexia as well as in cancer patients. Glucocorticoids and proinflammatory cytokines are responsible for an increased production of APR by muscle cells. Finally, their muscular expressions are strongly positively correlated with body weight loss as well as the muscular induction of atrogens. Our study demonstrates therefore a marked increased production of APR by the muscle in cancer cachexia.

8.
J Cachexia Sarcopenia Muscle ; 9(5): 929-946, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29989354

RESUMEN

BACKGROUND: Glucocorticoids (GC) play a major role in muscle atrophy. As skeletal muscle is a secretory organ, characterization of the muscle secretome elicited by muscle atrophy should allow to better understand the cellular mechanisms and to identify circulating biomarkers of this condition. Our project aimed to identify the changes in the muscle secretome associated with GC-induced muscle atrophy and susceptible to translate into circulation. METHODS: We have identified the GC-induced changes in the secretome of C2 C12 muscle cells by proteomic analysis, and then, we have determined how these changes translate into the circulation of mice or human subjects exposed to high concentrations of GC. RESULTS: This approach led us to identify Serpina3n as one of the most markedly secreted protein in response to GC. Our original in vitro results were confirmed in vivo by an increased expression of Serpina3n in skeletal muscle (3.9-fold; P < 0.01) and in the serum (two-fold; P < 0.01) of mice treated with GC. We also observed increased levels of the human orthologue Serpina3 in the serum of Cushing's syndrome patients compared with healthy controls matched for age and sex (n = 9/group, 2.5-fold; P < 0.01). An increase of Serpina3n was also demonstrated in muscle atrophy models mediated by GC such as cancer cachexia (four-fold; P < 0.01), sepsis (12.5-fold; P < 0.001), or diabetes (two-fold; P < 0.01). In contrast, levels of Serpina3n both in skeletal muscle and in the circulation were reduced in several models of muscle hypertrophy induced by myostatin inhibition (P < 0.01). Furthermore, a cluster of data suggests that the regulation of muscle Serpina3n involves mTOR, an essential determinant of the muscle cell size. CONCLUSIONS: Taken together, these data suggest that Serpina3n may represent a circulating biomarker of muscle atrophy associated to GC and, broadly, a reflection of dynamic changes in muscle mass.


Asunto(s)
Glucocorticoides/efectos adversos , Atrofia Muscular/etiología , Atrofia Muscular/metabolismo , Serpinas/metabolismo , Animales , Estudios de Casos y Controles , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Liquida , Síndrome de Cushing/complicaciones , Dexametasona/efectos adversos , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Masculino , Ratones , Atrofia Muscular/patología , Mioblastos , Proteoma , Proteómica/métodos , Serpinas/sangre , Espectrometría de Masas en Tándem
9.
J Proteome Res ; 16(10): 3477-3490, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28810121

RESUMEN

Skeletal muscle, the most abundant body tissue, plays vital roles in locomotion and metabolism. Myostatin is a negative regulator of skeletal muscle mass. In addition to increasing muscle mass, Myostatin inhibition impacts muscle contractility and energy metabolism. To decipher the mechanisms of action of the Myostatin inhibitors, we used proteomic and transcriptomic approaches to investigate the changes induced in skeletal muscles of transgenic mice overexpressing Follistatin, a physiological Myostatin inhibitor. Our proteomic workflow included a fractionation step to identify weakly expressed proteins and a comparison of fast versus slow muscles. Functional annotation of altered proteins supports the phenotypic changes induced by Myostatin inhibition, including modifications in energy metabolism, fiber type, insulin and calcium signaling, as well as membrane repair and regeneration. Less than 10% of the differentially expressed proteins were found to be also regulated at the mRNA level but the Biological Process annotation, and the KEGG pathways analysis of transcriptomic results shows a great concordance with the proteomic data. Thus this study describes the most extensive omics analysis of muscle overexpressing Follistatin, providing molecular-level insights to explain the observed muscle phenotypic changes.


Asunto(s)
Hipertrofia/genética , Enfermedades Musculares/genética , Miostatina/genética , Proteómica , Transcriptoma/genética , Animales , Modelos Animales de Enfermedad , Folistatina/farmacología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Hipertrofia/inducido químicamente , Hipertrofia/patología , Ratones , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/patología , Miostatina/antagonistas & inhibidores , Regeneración/genética
10.
J Cachexia Sarcopenia Muscle ; 8(5): 768-777, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28712119

RESUMEN

BACKGROUND: Several experimental evidences pinpoint the possible role of Activin A (ActA) as a driver of cancer cachexia. Supporting this hypothesis, we showed recently that human cancer cachexia is associated with high ActA levels. Moreover, ActA levels were correlated with body weight loss and skeletal muscle density, two prognostic factors in cancer patients. Our goal was therefore to investigate the value of ActA to predict survival in cancer patients. METHODS: Patients with colorectal or lung cancer were prospectively enrolled at the time of diagnosis or relapse between January 2012 and March 2014. At baseline, patients had clinical, nutritional, and functional assessment. Body composition and skeletal muscle density were measured by CT scan, and plasma ActA concentrations were determined. Overall survival (OS) was analysed since inclusion to 24 months later. RESULTS: Survival data were available for 149 patients out of 152. Patients with high ActA (≥408 pg/mL) had lower OS than those with low levels, regardless the type of cancer (OS in colorectal cancer, 50% vs. 79%, P < 0.05; and in lung cancer, 27% vs. 67%, P = 0.001). The multivariable analysis confirmed the prognostic value of ActA independently of tumour stage or inflammatory markers, particularly in lung cancer. Low muscularity was also an independent prognostic factor. CONCLUSIONS: Our study demonstrates that high ActA level is an independent prognosis factor of survival in cancer patients. More than a basic marker of the severity of the neoplastic disease or of the inflammatory process, ActA seems to influence survival by contributing to the development of cachexia and loss of skeletal muscle mass.


Asunto(s)
Activinas/sangre , Biomarcadores de Tumor , Neoplasias/sangre , Neoplasias/mortalidad , Tejido Adiposo/patología , Adulto , Anciano , Anciano de 80 o más Años , Composición Corporal , Caquexia/sangre , Caquexia/etiología , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Estadificación de Neoplasias , Neoplasias/complicaciones , Estado Nutricional , Tamaño de los Órganos , Pronóstico , Adulto Joven
11.
Am J Physiol Endocrinol Metab ; 309(6): E557-67, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26219865

RESUMEN

Follistatin, a physiological inhibitor of myostatin, induces a dramatic increase in skeletal muscle mass, requiring the type 1 IGF-I receptor/Akt/mTOR pathway. The aim of the present study was to investigate the role of IGF-I and insulin, two ligands of the IGF-I receptor, in the follistatin hypertrophic action on skeletal muscle. In a first step, we showed that follistatin increases muscle mass while being associated with a downregulation of muscle IGF-I expression. In addition, follistatin retained its full hypertrophic effect toward muscle in hypophysectomized animals despite very low concentrations of circulating and muscle IGF-I. Furthermore, follistatin did not increase muscle sensitivity to IGF-I in stimulating phosphorylation of Akt but, surprisingly, decreased it once hypertrophy was present. Taken together, these observations indicate that increased muscle IGF-I production or sensitivity does not contribute to the muscle hypertrophy caused by follistatin. Unlike low IGF-I, low insulin, as obtained by streptozotocin injection, attenuated the hypertrophic action of follistatin on skeletal muscle. Moreover, the full anabolic response to follistatin was restored in this condition by insulin but also by IGF-I infusion. Therefore, follistatin-induced muscle hypertrophy requires the activation of the insulin/IGF-I pathway by either insulin or IGF-I. When insulin or IGF-I alone is missing, follistatin retains its full anabolic effect, but when both are deficient, as in streptozotocin-treated animals, follistatin fails to stimulate muscle growth.


Asunto(s)
Folistatina/genética , Factor I del Crecimiento Similar a la Insulina/farmacología , Insulina/metabolismo , Músculo Esquelético/efectos de los fármacos , Miostatina/genética , Receptor IGF Tipo 1/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo , Folistatina/efectos de los fármacos , Folistatina/metabolismo , Hipertrofia/metabolismo , Hipofisectomía , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miostatina/efectos de los fármacos , Miostatina/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
12.
J Clin Endocrinol Metab ; 100(5): 2030-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25751105

RESUMEN

CONTEXT: Cachexia is a multifactorial syndrome, characterized by the loss of skeletal muscle mass and not fully reversible by nutritional support. Recent animal observations suggest that production of Activin A (ActA) and Myostatin (Mstn) by some tumors might contribute to cancer cachexia. OBJECTIVE: Our goal was to investigate the role of ActA and Mstn in the development of the human cancer cachexia. DESIGN/SETTING: The ACTICA study is a cross-sectional study, which prospectively enrolled patients from a tertiary-care center between January 2012 and March 2014. Subjects/Outcome Measures: One hundred fifty two patients with colorectal or lung cancer had clinical, nutritional and functional assessment. Body composition was measured by CT-scan, anthropometry, and bioimpedance. Plasma concentrations of ActA, Mstn, and Follistatin were determined. RESULTS: Cachexia was associated with reduced lean and fat mass (p < .01 and p < .001), reduced physical function, lower quality of life, and increased symptoms (QLQC30; p < .001). Anorexia (SNAQ score < 14) was more common in cachectic patients (CC) than in noncachectic patients (CNC) (p < .001). ActA concentrations in CC patients were higher than in CNC patients (+40%; p < .001) and were correlated positively with weight loss (R = 0.323; p < .001) and negatively with the SNAQ score (R = -0.225; p < .01). In contrast, Mstn concentrations were decreased in CC patients compared to CNC patients (-35%; p < .001). CONCLUSIONS: These results demonstrate an association between circulating concentrations of ActA and the presence of the anorexia/cachexia syndrome in cancer patients. Given the known muscle atrophic effects of ActA, our study suggests that increased circulating concentrations of ActA may contribute to the development of cachexia in cancer patients.


Asunto(s)
Activinas/sangre , Caquexia/etiología , Neoplasias Colorrectales/sangre , Neoplasias Pulmonares/sangre , Miostatina/sangre , Adulto , Anciano , Anciano de 80 o más Años , Composición Corporal , Caquexia/sangre , Neoplasias Colorrectales/complicaciones , Estudios Transversales , Femenino , Humanos , Neoplasias Pulmonares/complicaciones , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Calidad de Vida
13.
Peptides ; 33(2): 351-3, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22244812

RESUMEN

Urotensin II (UII) and urocortin (UCN) are potent contributors to the physiopathology of heart failure. Our study investigated the effects of UII and UCN on the expression of myostatin (Mstn) in primary culture of adult cardiomyocytes. Adult rat cardiomyocytes were stimulated for 48 h with UII and UCN. Cell size and protein content were determined. Mstn gene expression was determined by real time quantitative polymerase chain reaction. Treatment with UII and UCN stimulates hypertrophy of adult cardiomyocytes. This effect was associated with a twofold increase of Mstn gene expression. We have established for the first time that the two hypertrophic peptides UII and UCN stimulate the expression of Mstn.


Asunto(s)
Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miostatina/metabolismo , Urocortinas/fisiología , Urotensinas/fisiología , Animales , Células Cultivadas , Expresión Génica , Regulación de la Expresión Génica , Masculino , Miocardio/citología , Miostatina/genética , Ratas , Ratas Wistar
14.
Heart Vessels ; 27(2): 202-7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21505854

RESUMEN

Urocortin-1 (UCN), a member of the corticotropin-releasing factor, is a cardioprotective peptide, and is also involved in cardiac hypertrophy. The involvement of GSK-3ß, a pivotal kinase in cardiac hypertrophy, in response to UCN is not yet documented. Cardiomyocytes from adult rats were stimulated for 48 h with UCN. Cell size, protein, and DNA contents were determined. Phosphorylated and total forms GSK-3ß and the total amount of ß-catenin were quantified by Western immunoblots. The effects of astressin, a UCN competitive receptor antagonist, were also evaluated. UCN increased cell size and the protein-to-DNA ratio, in accordance with a hypertrophic response. This effect was associated with increased phosphorylation of GSK-3ß and marked accumulation of ß-catenin, a downstream element to GSK-3ß. All these effects were prevented by astressin and LY294002, an inhibitor of the phosphatidyl-inositol-3-kinase. UCN-induced cardiomyocytes hypertrophy is associated with regulation of GSK-3ß, a pivotal kinase involved in cardiac hypertrophy, in a PI3K-dependent manner. Furthermore, the pharmacological blockade of UCN receptors was able to prevent UCN-induced hypertrophy, which leads to inhibition of the Akt/GSK-3ß pathway.


Asunto(s)
Cardiomegalia/enzimología , Tamaño de la Célula , Glucógeno Sintasa Quinasa 3/metabolismo , Miocitos Cardíacos/enzimología , Transducción de Señal , Urocortinas/metabolismo , Animales , Western Blotting , Cardiomegalia/patología , Cardiomegalia/prevención & control , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Hormona Liberadora de Corticotropina/farmacología , Glucógeno Sintasa Quinasa 3 beta , Masculino , Morfolinas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fragmentos de Péptidos/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo , Transducción de Señal/efectos de los fármacos , beta Catenina/metabolismo
15.
Am J Physiol Endocrinol Metab ; 297(1): E157-64, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19435857

RESUMEN

Follistatin (FS) inhibits several members of the TGF-beta superfamily, including myostatin (Mstn), a negative regulator of muscle growth. Mstn inhibition by FS represents a potential therapeutic approach of muscle atrophy. The aim of our study was to investigate the mechanisms of the FS-induced muscle hypertrophy. To test the role of satellite cells in the FS effect, we used irradiation to destroy their proliferative capacity. FS overexpression increased the muscle weight by about 37% in control animals, but the increase reached only 20% in irradiated muscle, supporting the role of cell proliferation in the FS-induced hypertrophy. Surprisingly, the muscle hypertrophy caused by FS reached the same magnitude in Mstn-KO as in WT mice, suggesting that Mstn might not be the only ligand of FS involved in the regulation of muscle mass. To assess the role of activin (Act), another FS ligand, in the FS-induced hypertrophy, we electroporated FSI-I, a FS mutant that does not bind Act with high affinity. Whereas FS electroporation increased muscle weight by 32%, the muscle weight gain induced by FSI-I reached only 14%. Furthermore, in Mstn-KO mice, FSI-I overexpression failed to induce hypertrophy, in contrast to FS. Therefore, these results suggest that Act inhibition may contribute to FS-induced hypertrophy. Finally, the role of Act as a regulator of muscle mass was supported by the observation that ActA overexpression induced muscle weight loss (-15%). In conclusion, our results show that satellite cell proliferation and both Mstn and Act inhibition are involved in the FS-induced muscle hypertrophy.


Asunto(s)
Activinas/antagonistas & inhibidores , Proliferación Celular , Folistatina/fisiología , Atrofia Muscular/genética , Miostatina/antagonistas & inhibidores , Células Satélite del Músculo Esquelético/fisiología , Activinas/genética , Activinas/metabolismo , Animales , Folistatina/genética , Hipertrofia/genética , Hipertrofia/metabolismo , Masculino , Ratones , Ratones Noqueados , Atrofia Muscular/metabolismo , Miostatina/genética , Miostatina/metabolismo , Ratas , Ratas Transgénicas , Ratas Wistar , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
16.
Am J Physiol Endocrinol Metab ; 292(1): E145-50, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16926385

RESUMEN

Myofibrillar protein loss occurring in catabolic situations is considered to be mediated by the release of proinflammatory cytokines and associated with a decrease in circulating and muscle levels of insulin-like growth factor I (IGF-I). In this paper, we investigated whether the C(2)C(12) myotube atrophy caused in vitro by TNF-alpha/IFN-gamma cytokines might be reversed by exogenous IGF-I. Our results showed that, despite the presence of TNF-alpha/IFN-gamma, IGF-I retained its full ability to induce the phosphorylation of Akt, Foxo3a, and GSK-3beta (respectively, 16-fold, 9-fold, and 2-fold) together with a decrease in atrogin-1 mRNA (-39%, P < 0.001). Although this ubiquitin ligase has been reported to accelerate the degradation of MyoD, a myogenic transcription factor driving the transcription of myosin heavy chain (MHC), IGF-I failed to blunt the reduction of MyoD and MHC caused by TNF-alpha/IFN-gamma. Moreover, IGF-I only very slightly attenuated the myotube atrophy induced by TNF-alpha/IFN-gamma (TNF-alpha/IFN-gamma 15.48 mum alone vs. TNF-alpha/IFN-gamma/IGF-I 16.97 mum, P < 0.001). In conclusion, our data show that IGF-I does not reverse the myotube atrophy induced by TNF-alpha/IFN-gamma despite the phosphorylation of Foxo and GSK-3beta and the downregulation of atrogin-1 mRNA. Our study suggests therefore that factors other than IGF-I decrease are responsible for the muscle atrophy caused by proinflammatory cytokines.


Asunto(s)
Citocinas/efectos adversos , Factores de Transcripción Forkhead/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Mediadores de Inflamación/efectos adversos , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas Musculares/antagonistas & inhibidores , Atrofia Muscular/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Ligasas SKP Cullina F-box/antagonistas & inhibidores , Animales , Células Cultivadas , Proteína Forkhead Box O3 , Glucógeno Sintasa Quinasa 3 beta , Interferón gamma/farmacología , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Proteínas Musculares/metabolismo , Atrofia Muscular/etiología , Proteína MioD/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
17.
Endocrinology ; 145(11): 4806-12, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15284206

RESUMEN

In catabolic conditions, atrogin-1/MAFbx, a muscle-specific ubiquitin-ligase required for muscle atrophy, is increased, and concentrations of IGF-I, a growth factor known to have antiproteolytic action, are reduced. To define the relationship between the decline in IGF-I and the induction of atrogin-1/MAFbx, we studied the effect of IGF-I replacement on atrogin-1/MAFbx mRNA in rats fasted for 51 h and in rats made diabetic with streptozotocin (STZ). Fasting produced a 5.8-fold increase in atrogin-1/MAFbx (P < 0.001). This was attenuated to a 2.5-fold increase by injections of IGF-I (P < 0.05 vs. fasting). Animals with STZ-induced diabetes experienced a 15.1-fold increase in atrogin-1/MAFbx (P < 0.001). Normalization of their circulating IGF-I concentrations by IGF-I infusion blunted the induction of atrogin-1/MAFbx to 6.3-fold (P < 0.05 vs. STZ diabetes without IGF-I). To further delineate the regulation of atrogin-1/MAFbx by IGF-I, we studied a model of cultured muscle cells. We observed that IGF-I produced a time- and dose-dependent reduction of atrogin-1/MAFbx mRNA, with a 50% effective dose of 5 nm IGF-I, a physiological concentration. The degradation rate of atrogin-1/MAFbx mRNA was not affected by IGF-I, suggesting that the reduction of atrogin-1/MAFbx mRNA by IGF-I is a transcriptional effect. Exposure of muscle cells in culture to dexamethasone increased atrogin-1/MAFbx mRNA with a 50% effective dose of 10 nm, a pharmacological concentration. In the presence of dexamethasone, IGF-I at physiological concentrations retained its full inhibitory effect on atrogin-1/MAFbx mRNA. We conclude that IGF-I inhibits atrogin-1/MAFbx expression and speculate that this effect might contribute to the antiproteolytic action of IGF-I in muscle.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas Musculares/genética , Proteínas Ligasas SKP Cullina F-box/genética , Animales , Células Cultivadas , Dexametasona/farmacología , Ayuno/fisiología , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Glucocorticoides/farmacología , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , ARN Mensajero/análisis , Ratas , Ratas Wistar , Proteínas Ligasas SKP Cullina F-box/metabolismo
18.
Am J Physiol Heart Circ Physiol ; 285(6): H2482-91, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14613913

RESUMEN

We investigated the time course of the expression of cardiac and renal endothelin systems in tachycardia-induced heart failure in dogs. Eleven beagles underwent rapid pacing at a progressively increased rate over a period of 5 wk, with a weekly clinical examination, echocardiography, measurement of circulating and urinary endothelin-1 (ET-1), and myocardial and renal tissue biopsies. Real-time quantitative PCR was used for determinations of tissue prepro-ET-1 (ppET-1), ET-1-converting enzyme (ECE-1), and ETA and ETB receptor mRNA. Cardiac and renal tissue ET-1 contents were evaluated by immunostaining and measured by radioimmunoassay at autopsy. Rapid pacing caused a progressive increase in end-systolic and end-diastolic ventricular volumes (P < 0.05) from week 2 together with a decrease in ejection fraction and in mean velocity of circumferential shortening (P < 0.05) from week 1. These changes were tightly correlated to myocardial ppET-1 and renal ETA receptor mRNA and less so to myocardial ECE-1 mRNA, and they occurred before any increase in plasma and urinary ET-1 (P < 0.05 from week 4) and clinical signs of heart failure. Renal ppET-1 did not change. Both cardiac and renal ET-1 peptide contents were increased at autopsy. We conclude that tachycardia-induced heart failure in dogs is characterized by an early activation of the cardiac and renal tissue endothelin systems, which occurs before any changes in circulating and urinary ET-1 and is closely related to altered ventricular function.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Riñón/fisiopatología , Miocardio/metabolismo , Receptor de Endotelina A/genética , Receptor de Endotelina B/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Biopsia , Presión Sanguínea , Perros , Endotelina-1/sangre , Endotelina-1/genética , Endotelina-1/metabolismo , Endotelina-1/orina , Enzimas Convertidoras de Endotelina , Expresión Génica/fisiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Frecuencia Cardíaca , Riñón/metabolismo , Riñón/patología , Masculino , Metaloendopeptidasas , Marcapaso Artificial , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Respiración
19.
FEBS Lett ; 544(1-3): 214-7, 2003 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-12782319

RESUMEN

MafBx and Murf are two new rat E3 ubiquitin ligases induced in muscle atrophy. Our goal was to investigate whether lipopolysaccharide (LPS) injection, a model of muscle catabolism, is associated with increased expression of MafBx and Murf. LPS (750 microg/100 g body weight) induces MafBx and Murf mRNA (respectively, 23-fold and 33-fold after 12 h; P<0.001). A transient induction of tumor necrosis factor-alpha mRNA (21-fold; P<0.001 at 3 h) and a decrease of insulin like growth factor-I mRNA (50%; P<0.001 at 6 h), two potential regulators of the ubiquitin-proteasome system were also demonstrated. In summary, MafBx and Murf mRNA are up-regulated in response to LPS and might play a role in the muscle proteolysis observed.


Asunto(s)
Ligasas/metabolismo , Lipopolisacáridos/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Proteínas Ligasas SKP Cullina F-box , Ubiquitina-Proteína Ligasas , Animales , Peso Corporal , Músculo Esquelético/efectos de los fármacos , Músculos/metabolismo , ARN/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteínas de Motivos Tripartitos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA