Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Commun ; 15(1): 451, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38200005

RESUMEN

Immune cells must adapt to different environments during the course of an immune response. Here we study the adaptation of CD8+ T cells to the intestinal microenvironment and how this process shapes the establishment of the CD8+ T cell pool. CD8+ T cells progressively remodel their transcriptome and surface phenotype as they enter the gut wall, and downregulate expression of mitochondrial genes. Human and mouse intestinal CD8+ T cells have reduced mitochondrial mass, but maintain a viable energy balance to sustain their function. We find that the intestinal microenvironment is rich in prostaglandin E2 (PGE2), which drives mitochondrial depolarization in CD8+ T cells. Consequently, these cells engage autophagy to clear depolarized mitochondria, and enhance glutathione synthesis to scavenge reactive oxygen species (ROS) that result from mitochondrial depolarization. Impairing PGE2 sensing promotes CD8+ T cell accumulation in the gut, while tampering with autophagy and glutathione negatively impacts the T cell pool. Thus, a PGE2-autophagy-glutathione axis defines the metabolic adaptation of CD8+ T cells to the intestinal microenvironment, to ultimately influence the T cell pool.


Asunto(s)
Autofagia , Linfocitos T CD8-positivos , Humanos , Animales , Ratones , Dinoprostona , Genes Mitocondriales , Glutatión
2.
bioRxiv ; 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36993703

RESUMEN

Immune cells must adapt to different environments during the course of an immune response. We studied the adaptation of CD8 + T cells to the intestinal microenvironment and how this process shapes their residency in the gut. CD8 + T cells progressively remodel their transcriptome and surface phenotype as they acquire gut residency, and downregulate expression of mitochondrial genes. Human and mouse gut-resident CD8 + T cells have reduced mitochondrial mass, but maintain a viable energy balance to sustain their function. We found that the intestinal microenvironment is rich in prostaglandin E 2 (PGE 2 ), which drives mitochondrial depolarization in CD8 + T cells. Consequently, these cells engage autophagy to clear depolarized mitochondria, and enhance glutathione synthesis to scavenge reactive oxygen species (ROS) that result from mitochondrial depolarization. Impairing PGE 2 sensing promotes CD8 + T cell accumulation in the gut, while tampering with autophagy and glutathione negatively impacts the T cell population. Thus, a PGE 2 -autophagy-glutathione axis defines the metabolic adaptation of CD8 + T cells to the intestinal microenvironment, to ultimately influence the T cell pool.

3.
J Clin Invest ; 131(11)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34060483

RESUMEN

Skeletal muscle wasting is commonly associated with chronic kidney disease (CKD), resulting in increased morbidity and mortality. However, the link between kidney and muscle function remains poorly understood. Here, we took a complementary interorgan approach to investigate skeletal muscle wasting in CKD. We identified increased production and elevated blood levels of soluble pro-cachectic factors, including activin A, directly linking experimental and human CKD to skeletal muscle wasting programs. Single-cell sequencing data identified the expression of activin A in specific kidney cell populations of fibroblasts and cells of the juxtaglomerular apparatus. We propose that persistent and increased kidney production of pro-cachectic factors, combined with a lack of kidney clearance, facilitates a vicious kidney/muscle signaling cycle, leading to exacerbated blood accumulation and, thereby, skeletal muscle wasting. Systemic pharmacological blockade of activin A using soluble activin receptor type IIB ligand trap as well as muscle-specific adeno-associated virus-mediated downregulation of its receptor ACVR2A/B prevented muscle wasting in different mouse models of experimental CKD, suggesting that activin A is a key factor in CKD-induced cachexia. In summary, we uncovered a crosstalk between kidney and muscle and propose modulation of activin signaling as a potential therapeutic strategy for skeletal muscle wasting in CKD.


Asunto(s)
Caquexia/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Insuficiencia Renal Crónica/metabolismo , Síndrome Debilitante/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Activinas/genética , Activinas/metabolismo , Animales , Caquexia/etiología , Caquexia/genética , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Atrofia Muscular/etiología , Atrofia Muscular/genética , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/genética , Síndrome Debilitante/etiología , Síndrome Debilitante/genética
4.
Acta Physiol (Oxf) ; 230(1): e13496, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32408395

RESUMEN

AIM: Resistance exercise increases muscle mass over time. However, the early signalling events leading to muscle growth are not yet well-defined. Here, we aim to identify new signalling pathways important for muscle remodelling after exercise. METHODS: We performed a phosphoproteomics screen after a single bout of exercise in mice. As an exercise model we used unilateral electrical stimulation in vivo and treadmill running. We analysed muscle biopsies from human subjects to verify if our findings in murine muscle also translate to exercise in humans. RESULTS: We identified a new phosphorylation site on Myocardin-Related Transcription Factor B (MRTF-B), a co-activator of serum response factor (SRF). Phosphorylation of MRTF-B is required for its nuclear translocation after exercise and is accompanied by the transcription of the SRF target gene Fos. In addition, high-intensity exercise also remodels chromatin at specific SRF target gene loci through the phosphorylation of histone 3 on serine 10 in myonuclei of both mice and humans. Ablation of the MAP kinase member MSK1/2 is sufficient to prevent this histone phosphorylation, reduce induction of SRF-target genes, and prevent increases in protein synthesis after exercise. CONCLUSION: Our results identify a new exercise signalling fingerprint in vivo, instrumental for exercise-induced protein synthesis and potentially muscle growth.


Asunto(s)
Cromatina/química , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal , Factor de Respuesta Sérica , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Ejercicio Físico , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Biosíntesis de Proteínas , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo
5.
J Cachexia Sarcopenia Muscle ; 11(1): 208-225, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31651100

RESUMEN

BACKGROUND: Skeletal muscle is a plastic tissue that can adapt to different stimuli. It is well established that Mammalian Target of Rapamycin Complex 1 (mTORC1) signalling is a key modulator in mediating increases in skeletal muscle mass and function. However, the role of mTORC1 signalling in adult skeletal muscle homeostasis is still not well defined. METHODS: Inducible, muscle-specific Raptor and mTOR k.o. mice were generated. Muscles at 1 and 7 months after deletion were analysed to assess muscle histology and muscle force. RESULTS: We found no change in muscle size or contractile properties 1 month after deletion. Prolonging deletion of Raptor to 7 months, however, leads to a very marked phenotype characterized by weakness, muscle regeneration, mitochondrial dysfunction, and autophagy impairment. Unexpectedly, reduced mTOR signalling in muscle fibres is accompanied by the appearance of markers of fibre denervation, like the increased expression of the neural cell adhesion molecule (NCAM). Both muscle-specific deletion of mTOR or Raptor, or the use of rapamycin, was sufficient to induce 3-8% of NCAM-positive fibres (P < 0.01), muscle fibrillation, and neuromuscular junction (NMJ) fragmentation in 24% of examined fibres (P < 0.001). Mechanistically, reactivation of autophagy with the small peptide Tat-beclin1 is sufficient to prevent mitochondrial dysfunction and the appearance of NCAM-positive fibres in Raptor k.o. muscles. CONCLUSIONS: Our study shows that mTOR signalling in skeletal muscle fibres is critical for maintaining proper fibre innervation, preserving the NMJ structure in both the muscle fibre and the motor neuron. In addition, considering the beneficial effects of exercise in most pathologies affecting the NMJ, our findings suggest that part of these beneficial effects of exercise are through the well-established activation of mTORC1 in skeletal muscle during and after exercise.


Asunto(s)
Músculo Esquelético/fisiopatología , Unión Neuromuscular/fisiopatología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados
6.
J Cachexia Sarcopenia Muscle ; 10(3): 662-686, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30916493

RESUMEN

BACKGROUND: One of the principles underpinning our understanding of ageing is that DNA damage induces a stress response that shifts cellular resources from growth towards maintenance. A contrasting and seemingly irreconcilable view is that prompting growth of, for example, skeletal muscle confers systemic benefit. METHODS: To investigate the robustness of these axioms, we induced muscle growth in a murine progeroid model through the use of activin receptor IIB ligand trap that dampens myostatin/activin signalling. Progeric mice were then investigated for neurological and muscle function as well as cellular profiling of the muscle, kidney, liver, and bone. RESULTS: We show that muscle of Ercc1Δ/- progeroid mice undergoes severe wasting (decreases in hind limb muscle mass of 40-60% compared with normal mass), which is largely protected by attenuating myostatin/activin signalling using soluble activin receptor type IIB (sActRIIB) (increase of 30-62% compared with untreated progeric). sActRIIB-treated progeroid mice maintained muscle activity (distance travel per hour: 5.6 m in untreated mice vs. 13.7 m in treated) and increased specific force (19.3 mN/mg in untreated vs. 24.0 mN/mg in treated). sActRIIb treatment of progeroid mice also improved satellite cell function especially their ability to proliferate on their native substrate (2.5 cells per fibre in untreated progeroids vs. 5.4 in sActRIIB-treated progeroids after 72 h in culture). Besides direct protective effects on muscle, we show systemic improvements to other organs including the structure and function of the kidneys; there was a major decrease in the protein content in urine (albumin/creatinine of 4.9 sActRIIB treated vs. 15.7 in untreated), which is likely to be a result in the normalization of podocyte foot processes, which constitute the filtration apparatus (glomerular basement membrane thickness reduced from 224 to 177 nm following sActRIIB treatment). Treatment of the progeric mice with the activin ligand trap protected against the development of liver abnormalities including polyploidy (18.3% untreated vs. 8.1% treated) and osteoporosis (trabecular bone volume; 0.30 mm3 in treated progeroid mice vs. 0.14 mm3 in untreated mice, cortical bone volume; 0.30 mm3 in treated progeroid mice vs. 0.22 mm3 in untreated mice). The onset of neurological abnormalities was delayed (by ~5 weeks) and their severity reduced, overall sustaining health without affecting lifespan. CONCLUSIONS: This study questions the notion that tissue growth and maintaining tissue function during ageing are incompatible mechanisms. It highlights the need for future investigations to assess the potential of therapies based on myostatin/activin blockade to compress morbidity and promote healthy ageing.


Asunto(s)
Activinas/antagonistas & inhibidores , Envejecimiento/patología , Músculo Esquelético/patología , Transducción de Señal/efectos de los fármacos , Síndrome Debilitante/prevención & control , Receptores de Activinas Tipo II/administración & dosificación , Receptores de Activinas Tipo II/genética , Activinas/metabolismo , Animales , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Endonucleasas/genética , Femenino , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Miostatina/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Índice de Severidad de la Enfermedad , Síndrome Debilitante/diagnóstico , Síndrome Debilitante/genética , Síndrome Debilitante/patología
7.
Sci Rep ; 7: 40993, 2017 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-28106121

RESUMEN

Endoplasmic reticulum (ER) and oxidative stress are two related phenomena that have important metabolic consequences. As many skeletal muscle diseases are triggered by oxidative stress, we explored the chain of events linking a hyperoxidized ER (which causes ER and oxidative stress) with skeletal muscle dysfunction. An unbiased exon expression array showed that the combined genetic modulation of the two master ER redox proteins, selenoprotein N (SEPN1) and endoplasmic oxidoreductin 1 (ERO1), led to an SEPN1-related myopathic phenotype due to excessive signalling of transforming growth factor (TGF)-beta. The increased TGF-beta activity in the genetic mutants was caused by accelerated turnover of the ER localized (anti-oxidant) ascorbic acid that affected collagen deposition in the extracellular matrix. In a mouse mutant of SEPN1, which is dependent on exogenous ascorbic acid, a limited intake of ascorbic acid revealed a myopathic phenotype as a consequence of an altered TGF-beta signalling. Indeed, systemic antagonism of TGF-beta re-established skeletal muscle function in SEPN1 mutant mice. In conclusion, this study sheds new light on the molecular mechanism of SEPN1-related myopathies and indicates that the TGF-beta/ERO1/ascorbic acid axis offers potential for their treatment.


Asunto(s)
Antioxidantes/metabolismo , Ácido Ascórbico/metabolismo , Retículo Endoplásmico/fisiología , Músculo Esquelético/fisiopatología , Enfermedades Musculares/fisiopatología , Estrés Oxidativo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Perfilación de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Histocitoquímica , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Microscopía Electrónica , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/patología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Oxidorreductasas , Selenoproteínas/genética , Selenoproteínas/metabolismo , Transducción de Señal
8.
Front Physiol ; 8: 968, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29255421

RESUMEN

Skeletal muscle mass is a result of the balance between protein breakdown and protein synthesis. It has been shown that multiple conditions of muscle atrophy are characterized by the common regulation of a specific set of genes, termed atrogenes. It is not known whether various models of muscle hypertrophy are similarly regulated by a common transcriptional program. Here, we characterized gene expression changes in three different conditions of muscle growth, examining each condition during acute and chronic phases. Specifically, we compared the transcriptome of Extensor Digitorum Longus (EDL) muscles collected (1) during the rapid phase of postnatal growth at 2 and 4 weeks of age, (2) 24 h or 3 weeks after constitutive activation of AKT, and (3) 24 h or 3 weeks after overload hypertrophy caused by tenotomy of the Tibialis Anterior muscle. We observed an important overlap between significantly regulated genes when comparing each single condition at the two different timepoints. Furthermore, examining the transcriptional changes occurring 24 h after a hypertrophic stimulus, we identify an important role for genes linked to a stress response, despite the absence of muscle damage in the AKT model. However, when we compared all different growth conditions, we did not find a common transcriptional fingerprint. On the other hand, all conditions showed a marked increase in mTORC1 signaling and increased ribosome biogenesis, suggesting that muscle growth is characterized more by translational, than transcriptional regulation.

9.
Cell Metab ; 25(1): 182-196, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28011087

RESUMEN

The transcription factor EB (TFEB) is an essential component of lysosomal biogenesis and autophagy for the adaptive response to food deprivation. To address the physiological function of TFEB in skeletal muscle, we have used muscle-specific gain- and loss-of-function approaches. Here, we show that TFEB controls metabolic flexibility in muscle during exercise and that this action is independent of peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α). Indeed, TFEB translocates into the myonuclei during physical activity and regulates glucose uptake and glycogen content by controlling expression of glucose transporters, glycolytic enzymes, and pathways related to glucose homeostasis. In addition, TFEB induces the expression of genes involved in mitochondrial biogenesis, fatty acid oxidation, and oxidative phosphorylation. This coordinated action optimizes mitochondrial substrate utilization, thus enhancing ATP production and exercise capacity. These findings identify TFEB as a critical mediator of the beneficial effects of exercise on metabolism.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Metabolismo , Condicionamiento Físico Animal , Adenilato Quinasa/metabolismo , Animales , Autofagia/genética , Núcleo Celular/metabolismo , Metabolismo Energético/genética , Genes Mitocondriales , Genoma , Glucosa/metabolismo , Homeostasis/genética , Insulina/metabolismo , Metabolismo/genética , Ratones Noqueados , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Transporte de Proteínas , Transducción de Señal/genética
10.
Cell Rep ; 17(2): 501-513, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27705797

RESUMEN

Loss of skeletal muscle mass and force aggravates age-related sarcopenia and numerous pathologies, such as cancer and diabetes. The AKT-mTORC1 pathway plays a major role in stimulating adult muscle growth; however, the functional role of its downstream mediators in vivo is unknown. Here, we show that simultaneous inhibition of mTOR signaling to both S6K1 and 4E-BP1 is sufficient to reduce AKT-induced muscle growth and render it insensitive to the mTORC1-inhibitor rapamycin. Surprisingly, lack of mTOR signaling to 4E-BP1 only, or deletion of S6K1 alone, is not sufficient to reduce muscle hypertrophy or alter its sensitivity to rapamycin. However, we report that, while not required for muscle growth, S6K1 is essential for maintaining muscle structure and force production. Hypertrophy in the absence of S6K1 is characterized by compromised ribosome biogenesis and the formation of p62-positive protein aggregates. These findings identify S6K1 as a crucial player for maintaining muscle function during hypertrophy.


Asunto(s)
Proteínas Portadoras/genética , Hipertrofia/genética , Músculo Esquelético/metabolismo , Fosfoproteínas/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Serina-Treonina Quinasas TOR/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Factores Eucarióticos de Iniciación , Humanos , Hipertrofia/metabolismo , Hipertrofia/patología , Ratones , Ratones Noqueados , Músculo Esquelético/crecimiento & desarrollo , Proteína Oncogénica v-akt/genética , Péptidos/genética , Fosfoproteínas/metabolismo , Fosforilación , Agregado de Proteínas/genética , Ribosomas/genética , Ribosomas/metabolismo , Sarcopenia/genética , Sarcopenia/metabolismo , Sarcopenia/patología , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
11.
Mol Metab ; 3(1): 29-41, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24567902

RESUMEN

Circadian rhythms control metabolism and energy homeostasis, but the role of the skeletal muscle clock has never been explored. We generated conditional and inducible mouse lines with muscle-specific ablation of the core clock gene Bmal1. Skeletal muscles from these mice showed impaired insulin-stimulated glucose uptake with reduced protein levels of GLUT4, the insulin-dependent glucose transporter, and TBC1D1, a Rab-GTPase involved in GLUT4 translocation. Pyruvate dehydrogenase (PDH) activity was also reduced due to altered expression of circadian genes Pdk4 and Pdp1, coding for PDH kinase and phosphatase, respectively. PDH inhibition leads to reduced glucose oxidation and diversion of glycolytic intermediates to alternative metabolic pathways, as revealed by metabolome analysis. The impaired glucose metabolism induced by muscle-specific Bmal1 knockout suggests that a major physiological role of the muscle clock is to prepare for the transition from the rest/fasting phase to the active/feeding phase, when glucose becomes the predominant fuel for skeletal muscle.

13.
J Gen Physiol ; 140(2): 235-41, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22802359

RESUMEN

The presence and role of functional inositol 1,4,5-trisphosphate (IP(3)) receptors (IP(3)Rs) in adult skeletal muscle are controversial. The current consensus is that, in adult striated muscle, the relative amount of IP(3)Rs is too low and the kinetics of Ca(2+) release from IP(3)R is too slow compared with ryanodine receptors to contribute to the Ca(2+) transient during excitation-contraction coupling. However, it has been suggested that IP(3)-dependent Ca(2+) release may be involved in signaling cascades leading to regulation of muscle gene expression. We have reinvestigated IP(3)-dependent Ca(2+) release in isolated flexor digitorum brevis (FDB) muscle fibers from adult mice. Although Ca(2+) transients were readily induced in cultured C2C12 muscle cells by (a) UTP stimulation, (b) direct injection of IP(3), or (c) photolysis of membrane-permeant caged IP(3), no statistically significant change in calcium signal was detected in adult FDB fibers. We conclude that the IP(3)-IP(3)R system does not appear to affect global calcium levels in adult mouse skeletal muscle.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Inositol 1,4,5-Trifosfato/farmacología , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Ratones , Ratones Endogámicos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fotólisis , Uridina Trifosfato/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA