Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Physiol Rev ; 103(4): 2679-2757, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37382939

RESUMEN

Mechanisms underlying mechanical overload-induced skeletal muscle hypertrophy have been extensively researched since the landmark report by Morpurgo (1897) of "work-induced hypertrophy" in dogs that were treadmill trained. Much of the preclinical rodent and human resistance training research to date supports that involved mechanisms include enhanced mammalian/mechanistic target of rapamycin complex 1 (mTORC1) signaling, an expansion in translational capacity through ribosome biogenesis, increased satellite cell abundance and myonuclear accretion, and postexercise elevations in muscle protein synthesis rates. However, several lines of past and emerging evidence suggest that additional mechanisms that feed into or are independent of these processes are also involved. This review first provides a historical account of how mechanistic research into skeletal muscle hypertrophy has progressed. A comprehensive list of mechanisms associated with skeletal muscle hypertrophy is then outlined, and areas of disagreement involving these mechanisms are presented. Finally, future research directions involving many of the discussed mechanisms are proposed.


Asunto(s)
Músculo Esquelético , Transducción de Señal , Humanos , Animales , Perros , Músculo Esquelético/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Biosíntesis de Proteínas , Hipertrofia/metabolismo , Mamíferos/metabolismo
2.
J Appl Physiol (1985) ; 133(6): 1273-1283, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36201323

RESUMEN

We investigated the impact of tumor burden on muscle wasting in metastatic (m) and xenograft (x) models of colorectal cancer (CRC). Male Nod SCID γ and CD2F1 mice were injected subcutaneously or intrasplenically with HCT116 or C26 tumor cells, respectively. CRC tumors resulted in significant muscle wasting regardless of tumor type or model, although muscle loss was exacerbated in mHCT116 hosts. The mHCT116 model decreased ribosomal (r)RNA content and rDNA transcription, whereas the mC26 model showed no loss of rRNA and the upregulation of rDNA transcription. The xHCT116 model reduced mTOR, RPS6, and 4E-BP1 phosphorylation, whereas the mHCT116 model had a similar effect on RPS6 and 4E-BP1 without altering mTOR phosphorylation. The C26 models caused a reduction in 4E-BP1 phosphorylation independent of mTOR. Muscle interleukin (IL)-6 mRNA was elevated in all models except xHCT116, and the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) mRNA was induced only in the mC26 model. IL-1ß mRNA increased in all groups with greater expression in metastatic relative to the xenograft model regardless of tumor types. Our findings indicate that HCT116 tumor burden results in more drastic muscle wasting and anabolic deficits, whereas C26 tumor burden causes similar muscle wasting but exhibits a divergent proinflammatory phenotype. These results highlight potentially important divergence in the pathogenesis of muscle wasting among preclinical models of CRC and demonstrate that tumor burden plays a role in determining anabolic deficits and the expression of proinflammatory effectors of muscle wasting in a tumor-type-dependent manner.NEW & NOTEWORTHY We provide evidence demonstrating that colorectal tumor burden plays a role in determining anabolic deficits and the expression of proinflammatory effectors of muscle wasting in a tumor-type-dependent manner.


Asunto(s)
Caquexia , Neoplasias Colorrectales , Ratones , Humanos , Masculino , Animales , Caquexia/metabolismo , Xenoinjertos , Músculo Esquelético/metabolismo , Ratones SCID , Atrofia Muscular/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , ARN Mensajero/metabolismo , ADN Ribosómico/metabolismo , ADN Ribosómico/farmacología
3.
J Appl Physiol (1985) ; 133(6): 1260-1272, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36201324

RESUMEN

Preclinical models have been instrumental to elucidate the mechanisms underlying muscle wasting in lung cancer (LC). We investigated anabolic deficits and the expression of proinflammatory effectors of muscle wasting in the LP07 and Lewis lung carcinoma (LLC) tumor models. Tumor growth resulted in significant weakness in LP07 but not in LLC mice despite similar reductions in gastrocnemius muscle mass in both models. The LP07 tumors caused a reduction in ribosomal (r)RNA and a decrease in rRNA gene (rDNA) transcription elongation, whereas no changes in ribosomal capacity were evident in LLC tumor-bearing mice. Expression of RNA Polymerase I (Pol I) elongation-associated subunits Polr2f, PAF53, and Znrd1 mRNAs was significantly elevated in the LP07 model, whereas Pol I elongation-related factors FACT and Spt4/5 mRNAs were elevated in the LLC mice. Reductions in RPS6 and 4E-BP1 phosphorylation were similar in both models but were independent of mTOR phosphorylation in LP07 mice. Muscle inflammation was also tumor-specific, IL-6 and TNF-α mRNA increased with LLC tumors, and upregulation of NLRP3 mRNA was independent of tumor type. In summary, although both models caused muscle wasting, only the LP07 model displayed muscle weakness with reductions in ribosomal capacity. Intracellular signaling diverged at the mTOR level with similar reductions in RPS6 and 4E-BP1 phosphorylation regardless of tumor type. The increase in proinflammatory factors was more pronounced in the LLC model. Our results demonstrate novel divergent anabolic deficits and expression of proinflammatory effectors of muscle wasting in the LP07 and LLC preclinical models of lung cancer.NEW & NOTEWORTHY We provide novel data demonstrating significant divergence in anabolic deficits and the expression of proinflammatory effectors of muscle wasting consequent to different lung-derived tumors.


Asunto(s)
Carcinoma Pulmonar de Lewis , Neoplasias Pulmonares , Ratones , Animales , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Caquexia/etiología , Atrofia Muscular/metabolismo , Músculo Esquelético/metabolismo , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Serina-Treonina Quinasas TOR/metabolismo , ARN Mensajero/metabolismo , Ratones Endogámicos C57BL
4.
Am J Physiol Cell Physiol ; 321(6): C1000-C1009, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34705587

RESUMEN

Chemotherapeutic agents (CAs) are first-line antineoplastic treatments against a wide variety of cancers. Despite their effectiveness in halting tumor progression, side effects associated with CAs promote muscle loss by incompletely understood mechanisms. To address this problem, we first identified how oxidative stress impairs protein synthesis in C2C12 myotubes. Transient elevations in reactive oxygen species (ROS) resulted in protein synthesis deficits and reduced ribosomal (r)RNA levels. Oxidative stress did not reduce rRNA gene (rDNA) transcription, but it caused an increase in rRNA and protein oxidation. To determine whether CAs affect protein synthesis independent of oxidative stress, we exposed myotubes to Paclitaxel (PTX), Doxorubicin (DXR), or Marizomib (Mzb) at doses that did result in elevated ROS levels (sub-ROS). Exposure to CAs reduced protein synthesis and rRNA levels, but unlike oxidative stress, sub-ROS exposures impaired rDNA transcription. These results indicate that although oxidative stress disrupts protein synthesis by compromising ribosomal quantity and quality, CAs at sub-ROS doses compromise protein synthesis and ribosomal capacity, at least in part, by reducing rDNA transcription. Therefore, CAs negatively impact protein synthesis by causing oxidative stress in addition to directly reducing the ribosomal capacity of myotubes in a ROS-independent manner.


Asunto(s)
Antineoplásicos/toxicidad , Fibras Musculares Esqueléticas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Ribosomas/efectos de los fármacos , Animales , Línea Celular , Peróxido de Hidrógeno/toxicidad , Ratones , Fibras Musculares Esqueléticas/metabolismo , Estrés Oxidativo/fisiología , Biosíntesis de Proteínas/fisiología , Ribosomas/metabolismo
5.
FASEB J ; 35(2): e21335, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33527503

RESUMEN

Muscle wasting in cancer is associated with deficits in protein synthesis, yet, the mechanisms underlying this anabolic impairment remain poorly understood. The capacity for protein synthesis is mainly determined by the abundance of muscle ribosomes, which is in turn regulated by transcription of the ribosomal (r)RNA genes (rDNA). In this study, we investigated whether muscle loss in a preclinical model of ovarian cancer is associated with a reduction in ribosomal capacity and was a consequence of impaired rDNA transcription. Tumor bearing resulted in a significant loss in gastrocnemius muscle weight and protein synthesis capacity, and was consistent with a significant reduction in rDNA transcription and ribosomal capacity. Despite the induction of the ribophagy receptor NUFIP1 mRNA and the loss of NUFIP1 protein, in vitro studies revealed that while inhibition of autophagy rescued NUFIP1, it did not prevent the loss of rRNA. Electrophoretic analysis of rRNA fragmentation from both in vivo and in vitro models showed no evidence of endonucleolytic cleavage, suggesting that rRNA degradation may not play a major role in modulating muscle ribosome abundance. Our results indicate that in this model of ovarian cancer-induced cachexia, the ability of skeletal muscle to synthesize protein is compromised by a reduction in rDNA transcription and consequently a lower ribosomal capacity. Thus, impaired ribosomal production appears to play a key role in the anabolic deficits associated with muscle wasting in cancer cachexia.


Asunto(s)
Caquexia/genética , ADN Ribosómico/genética , Músculo Esquelético/metabolismo , Neoplasias Ováricas/complicaciones , ARN Ribosómico/genética , Ribosomas/metabolismo , Animales , Caquexia/etiología , Caquexia/metabolismo , Línea Celular Tumoral , ADN Ribosómico/metabolismo , Femenino , Ratones , Biosíntesis de Proteínas , ARN Ribosómico/metabolismo , Transcripción Genética
6.
Sci Rep ; 9(1): 18251, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796806

RESUMEN

Carbon dioxide (CO2) is sensed by cells and can trigger signals to modify gene expression in different tissues leading to changes in organismal functions. Despite accumulating evidence that several pathways in various organisms are responsive to CO2 elevation (hypercapnia), it has yet to be elucidated how hypercapnia activates genes and signaling pathways, or whether they interact, are integrated, or are conserved across species. Here, we performed a large-scale transcriptomic study to explore the interaction/integration/conservation of hypercapnia-induced genomic responses in mammals (mice and humans) as well as invertebrates (Caenorhabditis elegans and Drosophila melanogaster). We found that hypercapnia activated genes that regulate Wnt signaling in mouse lungs and skeletal muscles in vivo and in several cell lines of different tissue origin. Hypercapnia-responsive Wnt pathway homologues were similarly observed in secondary analysis of available transcriptomic datasets of hypercapnia in a human bronchial cell line, flies and nematodes. Our data suggest the evolutionarily conserved role of high CO2 in regulating Wnt pathway genes.


Asunto(s)
Caenorhabditis elegans/metabolismo , Dióxido de Carbono/farmacología , Drosophila melanogaster/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Animales , Bronquios/citología , Bronquios/metabolismo , Caenorhabditis elegans/efectos de los fármacos , Línea Celular , Drosophila melanogaster/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Hipercapnia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares
7.
Am J Physiol Renal Physiol ; 317(5): F1122-F1131, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31432706

RESUMEN

Advanced chronic kidney disease (CKD) is characterized by a premature aging phenotype of multifactorial origin. Mitochondrial dysfunction is prevalent in CKD and has been proposed as a major contributor to poor muscle function. Although the mitochondria-derived peptides (MDPs) humanin and mitochondrial open reading frame of 12S rRNA-c (MOTS-c) are involved in cell survival, suppression of apoptosis, and glucose control, the implications of MDP in CKD are unknown. We investigated humanin and MOTS-c protein expression in skeletal muscle and serum levels in CKD at stage 5 (glomerular filtration rate: <15 ml/min) patients and age-matched controls with normal renal function. Whereas circulating levels of humanin were increased in CKD, local muscle expression was reduced. In contrast, MOTS-c levels were reduced in both skeletal muscle and serum in CKD. Humanin in serum correlated positively to circulating TNF levels. Reduced MDP levels in skeletal muscle were associated with lower mitochondrial density and evidence of oxidative stress. These results indicate a differential regulation of MDPs in CKD and suggest an alternative site for humanin production than skeletal muscle in the uremic milieu. MDP levels were linked to systemic inflammation and evidence of oxidative stress in the muscle, two hallmark features of premature aging and uremia.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mitocondrias Musculares/metabolismo , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Insuficiencia Renal Crónica/metabolismo , Adulto , Anciano , Regulación hacia Abajo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Factor 2 Relacionado con NF-E2/genética , Adulto Joven
8.
Exerc Sport Sci Rev ; 47(2): 91-97, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30632998

RESUMEN

An increase in ribosomal capacity is a hallmark of the hypertrophying muscle. We review evidence demonstrating that transcription of ribosomal RNA genes is necessary for the increase in ribosomal capacity, and this is critical for muscle growth in human and animal models of hypertrophy.


Asunto(s)
Músculo Esquelético/crecimiento & desarrollo , Ribosomas/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Humanos
9.
Muscle Nerve ; 58(2): 277-285, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29572878

RESUMEN

INTRODUCTION: Children with cerebral palsy (CP) and acquired brain injury (ABI) commonly develop muscle contractures with advancing age. An underlying growth defect contributing to skeletal muscle contracture formation in CP/ABI has been suggested. METHODS: The biceps muscles of children and adolescents with CP/ABI (n = 20) and typically developing controls (n = 10) were investigated. We used immunohistochemistry, quantitative real-time polymerase chain reaction, and Western blotting to assess gene expression relevant to growth and size homeostasis. RESULTS: Classical pro-inflammatory cytokines and genes involved in extracellular matrix (ECM) production were elevated in skeletal muscle of children with CP/ABI. Intramuscular collagen content was increased and satellite cell number decreased and this was associated with reduced levels of RNA polymerase I transcription factors, 45s pre-rRNA and 28S rRNA. DISCUSSION: The present study provides novel data suggesting a role for pro-inflammatory cytokines and reduced ribosomal production in the development/maintenance of muscle contractures, possibly underlying stunted growth and perimysial ECM expansion. Muscle Nerve 58: 277-285, 2018.


Asunto(s)
Lesiones Encefálicas/patología , Parálisis Cerebral/patología , Matriz Extracelular/patología , Músculo Esquelético/patología , ARN Ribosómico/biosíntesis , Adolescente , Recuento de Células , Niño , Colágeno/metabolismo , Citocinas/biosíntesis , Citocinas/genética , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Fibras Musculares Esqueléticas/patología , ARN Ribosómico/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Ribosomas/genética , Ribosomas/patología , Células Satélite del Músculo Esquelético/patología
10.
Am J Physiol Cell Physiol ; 311(4): C663-C672, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27581648

RESUMEN

Ribosome production is an early event during skeletal muscle hypertrophy and precedes muscle protein accretion. Signaling via mTOR is crucial for ribosome production and hypertrophy; however, the mechanisms by which it regulates these processes remain to be identified. Herein, we investigated the activation of mTOR signaling in hypertrophying myotubes and determined that mTOR coordinates various aspects of gene expression important for ribosome production. First, inhibition of translation with cycloheximide had a more potent effect on protein synthesis than rapamycin indicating that mTOR function during hypertrophy is not on general, but rather on specific protein synthesis. Second, blocking Pol II transcription had a similar effect as Rapamycin and, unexpectedly, revealed the necessity of Pol II transcription for Pol I transcription, suggesting that mTOR may regulate ribosome production also by controlling Class II genes at the transcriptional level. Third, Pol I activity is essential for rDNA transcription and, surprisingly, for protein synthesis as selective Pol I inhibition blunted rDNA transcription, protein synthesis, and the hypertrophic response of myotubes. Finally, mTOR has nuclear localization in muscle, which is not sensitive to rapamycin. Inhibition of mTOR signaling by rapamycin disrupted mTOR-rDNA promoter interaction and resulted in altered histone marks indicative of repressed transcription and formation of higher-order chromatin structure. Thus mTOR signaling appears to regulate muscle hypertrophy by affecting protein synthesis, Class I and II gene expression, and chromatin remodeling.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , ADN Ribosómico/genética , Fibras Musculares Esqueléticas/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Transcripción Genética/genética , Animales , Línea Celular Tumoral , Hipertrofia/genética , Ratones , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Regiones Promotoras Genéticas/genética , Ribosomas/genética
12.
Am J Physiol Regul Integr Comp Physiol ; 311(3): R545-57, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27465734

RESUMEN

The overload-induced increase in muscle mass is accompanied by protein accretion; however, the initiating events are poorly understood. Regulated in Development and DNA Damage 1 (REDD1), a repressor of the mechanistic target of rapamycin in complex 1 (mTORC1), blunts the elevation in protein synthesis induced by acute muscle contractions. Therefore, this study was designed to determine whether REDD1 alters the rate of the overload-induced increase in muscle mass. Wild-type (WT) and REDD1-null mice underwent unilateral functional overload (OV) of the plantaris, while the contralateral sham leg served as a control. After 3 and 5 days of OV, puromycin incorporation was used as a measurement of protein synthesis. The percent increase in plantaris wet weight and protein content was greater in REDD1-null mice after 3, 5, and 10 days OV. The overload-stimulated rate of protein synthesis in the plantaris was similar between genotypes after 3 days OV, but translational capacity was lower in REDD1-null mice, indicating elevated translational efficiency. This was likely due to elevated absolute mTORC1 signaling [phosphorylation of p70S6K1 (Thr-389) and 4E-BP1 (Ser-65)]. By 5 days of OV, the rate of protein synthesis in REDD1-null mice was lower than WT mice with no difference in absolute mTORC1 signaling. Additionally, markers of autophagy (LC3II/I ratio and p62 protein) were decreased to a greater absolute extent after 3 days OV in REDD1-null mice. These data suggest that loss of REDD1 augments the rate of the OV-induced increase in muscle mass by altering multiple protein balance pathways.


Asunto(s)
Contracción Muscular/fisiología , Proteínas Musculares/metabolismo , Músculo Esquelético/fisiología , Biosíntesis de Proteínas/fisiología , Factores de Transcripción/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos/fisiología , Factores de Transcripción/genética
13.
Am J Physiol Endocrinol Metab ; 310(6): E461-72, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26758685

RESUMEN

Mechanisms regulating skeletal muscle growth involve a balance between the activity of serine/threonine protein kinases, including the mammalian target of rapamycin (mTOR) and 5'-AMP-activated protein kinase (AMPK). The contribution of different AMPK subunits to the regulation of cell growth size remains inadequately characterized. Using AMPKγ3 mutant-overexpressing transgenic Tg-Prkag3(225Q) and AMPKγ3-knockout (Prkag3(-/-)) mice, we investigated the requirement for the AMPKγ3 isoform in functional overload-induced muscle hypertrophy. Although the genetic disruption of the γ3 isoform did not impair muscle growth, control sham-operated AMPKγ3-transgenic mice displayed heavier plantaris muscles in response to overload hypertrophy and underwent smaller mass gain and lower Igf1 expression compared with wild-type littermates. The mTOR signaling pathway was upregulated with functional overload but unchanged between genetically modified animals and wild-type littermates. Differences in AMPK-related signaling pathways between transgenic, knockout, and wild-type mice did not impact muscle hypertrophy. Glycogen content was increased following overload in wild-type mice. In conclusion, our functional, transcriptional, and signaling data provide evidence against the involvement of the AMPKγ3 isoform in the regulation of skeletal muscle hypertrophy. Thus, the AMPKγ3 isoform is dispensable for functional overload-induced muscle growth. Mechanical loading can override signaling pathways that act as negative effectors of mTOR signaling and consequently promote skeletal muscle hypertrophy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Hipertrofia/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/metabolismo , Tamaño de los Órganos , Transducción de Señal
14.
Nephrol Dial Transplant ; 31(7): 1070-7, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-25910496

RESUMEN

Muscle wasting (or sarcopenia) is a common feature of the uremic phenotype and predisposes this vulnerable patient population to increased risk of comorbid complications, poor quality of life, frailty and premature death. The old age of dialysis patients is in addition a likely contributor to loss of muscle mass. As recent evidence suggests that assessment of muscle strength (i.e. function) is a better predictor of outcome and comorbidities than muscle mass, this opens new screening, assessment and therapeutic opportunities. Among established treatment strategies, the benefit of resistance exercise and endurance training are increasingly recognized among nephrologists as being effective and should be promoted in sedentary chronic kidney disease patients. Testosterone and growth hormone replacement appear as the most promising among emerging treatments strategies for muscle wasting. As treatment of muscle wasting is difficult and seldom successful in this often old, frail, sedentary and exercise-hesitant patient group, novel treatment strategies are urgently needed. In this review, we summarize recent studies on stimulation of mitochondrial biogenesis, myogenic stem (satellite) cells and manipulation of transforming growth factor family members, all of which hold promise for more effective therapies to target muscle mass loss and function in the future.


Asunto(s)
Fallo Renal Crónico/complicaciones , Mortalidad Prematura , Síndrome Debilitante/terapia , Humanos , Síndrome Debilitante/mortalidad
15.
Nat Commun ; 6: 7916, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26243583

RESUMEN

The ability to repeatedly regenerate limbs during the entire lifespan of an animal is restricted to certain salamander species among vertebrates. This ability involves dedifferentiation of post-mitotic cells into progenitors that in turn form new structures. A long-term enigma has been how injury leads to dedifferentiation. Here we show that skeletal muscle dedifferentiation during newt limb regeneration depends on a programmed cell death response by myofibres. We find that programmed cell death-induced muscle fragmentation produces a population of 'undead' intermediate cells, which have the capacity to resume proliferation and contribute to muscle regeneration. We demonstrate the derivation of proliferating progeny from differentiated, multinucleated muscle cells by first inducing and subsequently intercepting a programmed cell death response. We conclude that cell survival may be manifested by the production of a dedifferentiated cell with broader potential and that the diversion of a programmed cell death response is an instrument to achieve dedifferentiation.


Asunto(s)
Desdiferenciación Celular , Músculo Esquelético/fisiología , Notophthalmus viridescens/fisiología , Regeneración , Animales , Caspasas/metabolismo , Muerte Celular , Proliferación Celular , Femenino , Ratones Endogámicos NOD , Ratones SCID , Fibras Musculares Esqueléticas , Músculo Esquelético/citología
17.
J Biol Chem ; 290(14): 9183-94, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25691571

RESUMEN

Patients with chronic obstructive pulmonary disease, acute lung injury, and critical care illness may develop hypercapnia. Many of these patients often have muscle dysfunction which increases morbidity and impairs their quality of life. Here, we investigated whether hypercapnia leads to skeletal muscle atrophy. Mice exposed to high CO2 had decreased skeletal muscle wet weight, fiber diameter, and strength. Cultured myotubes exposed to high CO2 had reduced fiber diameter, protein/DNA ratios, and anabolic capacity. High CO2 induced the expression of MuRF1 in vivo and in vitro, whereas MuRF1(-/-) mice exposed to high CO2 did not develop muscle atrophy. AMP-activated kinase (AMPK), a metabolic sensor, was activated in myotubes exposed to high CO2, and loss-of-function studies showed that the AMPKα2 isoform is necessary for muscle-specific ring finger protein 1 (MuRF1) up-regulation and myofiber size reduction. High CO2 induced AMPKα2 activation, triggering the phosphorylation and nuclear translocation of FoxO3a, and leading to an increase in MuRF1 expression and myotube atrophy. Accordingly, we provide evidence that high CO2 activates skeletal muscle atrophy via AMPKα2-FoxO3a-MuRF1, which is of biological and potentially clinical significance in patients with lung diseases and hypercapnia.


Asunto(s)
Adenilato Quinasa/metabolismo , Dióxido de Carbono/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/etiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Proteína Forkhead Box O3 , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas de Motivos Tripartitos , Regulación hacia Arriba
19.
J Gerontol A Biol Sci Med Sci ; 68(9): 1035-44, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23418191

RESUMEN

To develop a global view of muscle transcriptional differences between older men and women and sex-specific aging, we obtained muscle biopsies from the biceps brachii of young and older men and women and profiled the whole-genome gene expression using microarray. A logistic regression-based method in combination with an intensity-based Bayesian moderated t test was used to identify significant sex- and aging-related gene functional groups. Our analysis revealed extensive sex differences in the muscle transcriptome of older individuals and different patterns of transcriptional changes with aging in men and women. In older women, we observed a coordinated transcriptional upregulation of immune activation, extracellular matrix remodeling, and lipids storage; and a downregulation of mitochondrial biogenesis and function and muscle regeneration. The effect of aging results in sexual dimorphic alterations in the skeletal muscle transcriptome, which may modify the risk for developing musculoskeletal and metabolic diseases in men and women.


Asunto(s)
Envejecimiento/genética , Músculo Esquelético/metabolismo , Adulto , Brazo , Teorema de Bayes , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/genética , Persona de Mediana Edad , Enfermedades Musculoesqueléticas/etiología , Enfermedades Musculoesqueléticas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Riesgo , Caracteres Sexuales , Transcriptoma , Adulto Joven
20.
Muscle Nerve ; 46(5): 785-92, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22996233

RESUMEN

INTRODUCTION: Upper motor neuron lesions after stroke are a major cause of disability. We aimed to determine whether skeletal muscles from these patients display typical molecular signatures of inflammation, growth arrest, and atrophy. METHODS: Muscle biopsies were analyzed for morphological, histochemical, ultrastructural, and molecular features indicative of changes in gene expression involved in muscle atrophy. RESULTS: Chronic hemiplegia resulted in ~9.5% atrophy, fiber type shifts, and histochemical and ultrastructural signs of impaired remodeling. TNF and TWEAK expressions were unaltered, but MSTN mRNA was lower (-73%, P < 0.05) in paretic tibialis anterior vs. age-matched controls. The expression of autophagy-related genes (BCN-1, LC3, and GABARAPL1) was lower in paretic tibialis anterior (-81%, -48%, and -60%, respectively, P < 0.01) and soleus (-85%, -54%, and -60% respectively, P < 0.01) compared with old controls. CONCLUSIONS: Persistent atrophy in chronic spastic hemiplegia may be associated with impaired remodeling partly due to altered autophagy gene expression.


Asunto(s)
Autofagia/genética , Hemiplejía/genética , Hemiplejía/patología , Músculo Esquelético/patología , Atrofia Muscular/genética , Atrofia Muscular/patología , Adulto , Anciano , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica , Hemiplejía/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA