Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Genome Biol ; 22(1): 310, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34763716

RESUMEN

A modified Chromium 10x droplet-based protocol that subsamples cells for both short-read and long-read (nanopore) sequencing together with a new computational pipeline (FLAMES) is developed to enable isoform discovery, splicing analysis, and mutation detection in single cells. We identify thousands of unannotated isoforms and find conserved functional modules that are enriched for alternative transcript usage in different cell types and species, including ribosome biogenesis and mRNA splicing. Analysis at the transcript level allows data integration with scATAC-seq on individual promoters, improved correlation with protein expression data, and linked mutations known to confer drug resistance to transcriptome heterogeneity.


Asunto(s)
Secuenciación de Nanoporos/métodos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Empalme Alternativo , Animales , Exones , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Empalme del ARN , ARN Mensajero , Transcriptoma
2.
Mol Metab ; 45: 101157, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33359740

RESUMEN

OBJECTIVES: Preferential damage to fast, glycolytic myofibers is common in many muscle-wasting diseases, including Duchenne muscular dystrophy (DMD). Promoting an oxidative phenotype could protect muscles from damage and ameliorate the dystrophic pathology with therapeutic relevance, but developing efficacious strategies requires understanding currently unknown biological roles for dystrophin and utrophin in dystrophic muscle adaptation and plasticity. METHODS: Combining whole transcriptome RNA sequencing and mitochondrial proteomics with assessments of metabolic and contractile function, we investigated the roles of dystrophin and utrophin in fast-to-slow muscle remodeling with low-frequency electrical stimulation (LFS, 10 Hz, 12 h/d, 7 d/wk, 28 d) in mdx (dystrophin null) and dko (dystrophin/utrophin null) mice, two established preclinical models of DMD. RESULTS: Novel biological roles in adaptation were demonstrated by impaired transcriptional activation of estrogen-related receptor alpha-responsive genes supporting oxidative phosphorylation in dystrophic muscles. Further, utrophin expression in dystrophic muscles was required for LFS-induced remodeling of mitochondrial respiratory chain complexes, enhanced fiber respiration, and conferred protection from eccentric contraction-mediated damage. CONCLUSIONS: These findings reveal novel roles for dystrophin and utrophin during LFS-induced metabolic remodeling of dystrophic muscle and highlight the therapeutic potential of LFS to ameliorate the dystrophic pathology and protect from contraction-induced injury with important implications for DMD and related muscle disorders.


Asunto(s)
Adaptación Fisiológica/fisiología , Distrofina/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Utrofina/metabolismo , Animales , Distrofina/genética , Masculino , Ingeniería Metabólica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Mitocondrias/metabolismo , Contracción Muscular , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Utrofina/genética
3.
Biol Open ; 9(7)2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32605905

RESUMEN

In response to injury, skeletal muscle stem cells (MuSCs) undergo myogenesis where they become activated, proliferate rapidly, differentiate and undergo fusion to form multinucleated myotubes. Dramatic changes in cell size, shape, metabolism and motility occur during myogenesis, which cause cellular stress and alter proteostasis. The molecular chaperone heat shock protein 70 (HSP70) maintains proteostasis by regulating protein biosynthesis and folding, facilitating transport of polypeptides across intracellular membranes and preventing stress-induced protein unfolding/aggregation. Although HSP70 overexpression can exert beneficial effects in skeletal muscle diseases and enhance skeletal muscle repair after injury, its effect on myogenesis has not been investigated. Plasmid-mediated overexpression of HSP70 did not affect the rate of C2C12 proliferation or differentiation, but the median number of myonuclei per myotube and median myotube width in differentiated C2C12 myotubes were increased with HSP70 overexpression. These findings reveal that increased HSP70 expression can promote myoblast fusion, identifying a mechanism for its therapeutic potential to enhance muscle repair after injury.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Diferenciación Celular , Proteínas HSP70 de Choque Térmico/genética , Desarrollo de Músculos , Mioblastos/citología , Mioblastos/metabolismo , Animales , Diferenciación Celular/genética , Fusión Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Expresión Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo
4.
Cell Metab ; 31(6): 1052-1067, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32433923

RESUMEN

While metabolism was initially thought to play a passive role in cell biology by generating ATP to meet bioenergetic demands, recent studies have identified critical roles for metabolism in the generation of new biomass and provision of obligate substrates for the epigenetic modification of histones and DNA. This review details how metabolites generated through glycolysis and the tricarboxylic acid cycle are utilized by somatic stem cells to support cell proliferation and lineage commitment. Importantly, we also discuss the evolving hypothesis that histones can act as an energy reservoir during times of energy stress. Finally, we discuss how cells integrate both extrinsic metabolic cues and intrinsic metabolic machinery to regulate cell fate.


Asunto(s)
Células Madre Adultas/metabolismo , Células Madre Adultas/citología , Animales , Proliferación Celular , Ciclo del Ácido Cítrico , Glucólisis , Humanos
5.
Front Cell Dev Biol ; 7: 254, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31737625

RESUMEN

Skeletal muscle has a remarkable capacity to regenerate following injury, a property conferred by a resident population of muscle stem cells (MuSCs). In response to injury, MuSCs must double their cellular content to divide, a process requiring significant new biomass in the form of nucleotides, phospholipids, and amino acids. This new biomass is derived from a series of intracellular metabolic cycles and alternative routing of carbon. In this review, we examine the link between metabolism and skeletal muscle regeneration with particular emphasis on the role of the cellular microenvironment in supporting the production of new biomass and MuSC proliferation.

6.
Stem Cells Int ; 2019: 8195614, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31236115

RESUMEN

Metabolism has been shown to alter cell fate in human pluripotent stem cells (hPSC). However, current understanding is almost exclusively based on work performed at 20% oxygen (air), with very few studies reporting on hPSC at physiological oxygen (5%). In this study, we integrated metabolic, transcriptomic, and epigenetic data to elucidate the impact of oxygen on hPSC. Using 13C-glucose labeling, we show that 5% oxygen increased the intracellular levels of glycolytic intermediates, glycogen, and the antioxidant response in hPSC. In contrast, 20% oxygen increased metabolite flux through the TCA cycle, activity of mitochondria, and ATP production. Acetylation of H3K9 and H3K27 was elevated at 5% oxygen while H3K27 trimethylation was decreased, conforming to a more open chromatin structure. RNA-seq analysis of 5% oxygen hPSC also indicated increases in glycolysis, lysine demethylases, and glucose-derived carbon metabolism, while increased methyltransferase and cell cycle activity was indicated at 20% oxygen. Our findings show that oxygen drives metabolite flux and specifies carbon fate in hPSC and, although the mechanism remains to be elucidated, oxygen was shown to alter methyltransferase and demethylase activity and the global epigenetic landscape.

7.
Cell Stress Chaperones ; 24(4): 749-761, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31098840

RESUMEN

Skeletal myogenesis is a coordinated sequence of events associated with dramatic changes in cell morphology, motility, and metabolism, which causes cellular stress and alters proteostasis. Chaperones, such as heat-shock proteins (HSPs), play important roles in limiting cellular stresses and maintaining proteostasis, but whether HSPs are specifically involved in myogenesis is not well understood. Here, we characterized gene and protein expression and subcellular localization of various HSPs in proliferating C2C12 myoblasts and differentiating myotubes under control conditions and in response to heat stress. Hsp25, Hsp40, and Hsp60 protein expression declined by 48, 35, and 83%, respectively, during differentiation. In contrast, Hsp70 protein levels doubled during early differentiation. Hsp25 was predominantly localized to the cytoplasm of myoblasts and myotubes but formed distinct aggregates in perinuclear spaces of myoblasts after heat-shock. Hsp40 was distributed diffusely throughout the cytoplasm and nucleus and, after heat-shock, translocated to the nucleus of myoblasts but formed aggregates in myotubes. Hsp60 localized to the perinuclear space in myoblasts but was distributed more diffusely across the cytoplasm in myotubes. Hsp70 was expressed diffusely throughout the cytoplasm and nucleus and translocated to the nucleus after heat-shock in myoblasts, but not in myotubes. Hsp90 was expressed diffusely across the cytoplasm in both myoblasts and myotubes under control conditions and did not change in response to heat-shock. These findings reveal distinct and different roles for HSPs in the regulation of myogenic cell proliferation and differentiation.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Desarrollo de Músculos/fisiología , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Respuesta al Choque Térmico/fisiología , Fibras Musculares Esqueléticas/citología , Mioblastos/citología
8.
J Neurochem ; 149(2): 269-283, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30664245

RESUMEN

Contributions of damaged mitochondria to neuropathologies have stimulated interest in mitophagy. We investigated triggers of neuronal mitophagy by disruption of mitochondrial energy metabolism in primary neurons. Mitophagy was examined in cultured murine cerebellar granule cells after inhibition of mitochondrial respiratory chain by drugs rotenone, 3-nitropropionic acid, antimycin A, and potassium cyanide, targeting complexes I, II, III, and IV, respectively. Inhibitor concentrations producing slow cellular demise were determined from analyses of cellular viability, morphology of neuritic damage, plasma membrane permeability, and oxidative phosphorylation. Live cell imaging of dissipation of mitochondrial membrane potential (ΔΨm ) by drugs targeting mitochondrial complexes was referenced to complete depolarization by carbonyl cyanide m-chlorophenyl hydrazone. While inhibition of complexes I, III and IV effected rapid dissipation of ΔΨm , inhibition of complex II using 3-nitropropionic acid led to minimal depolarization of mitochondria. Nonetheless, all respiratory chain inhibitors triggered mitophagy as indicated by increased aggregation of mitochondrially localized PINK1. Mitophagy was further analyzed using a dual fluorescent protein biosensor reporting mitochondrial relocation to acidic lysosomal environment. Significant acidification of mitochondria was observed in neurons treated with rotenone or 3-nitropropionic acid, revealing mitophagy at distal processes. Neurons treated with antimycin A or cyanide failed to show mitochondrial acidification. Minor dissipation of ΔΨm by 3-nitropropionic acid coupled with vigorous triggering of mitophagy suggested depolarization of mitochondria is not a necessary condition to trigger mitophagy. Moreover, weak elicitation of mitophagy by antimycin A, subsequent to loss of ΔΨm , suggested that mitochondrial depolarization is not a sufficient condition for triggering robust neuronal mitophagy. Our findings provide new insight into complexities of mitophagic clearance of neuronal mitochondria.


Asunto(s)
Metabolismo Energético/fisiología , Potencial de la Membrana Mitocondrial/fisiología , Mitofagia/fisiología , Neuronas/metabolismo , Animales , Células Cultivadas , Ratones , Proteínas Quinasas/metabolismo
9.
Curr Opin Clin Nutr Metab Care ; 21(4): 240-245, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29697538

RESUMEN

PURPOSE OF REVIEW: To discuss how innate muscle stem-cell metabolism and nutrient availability can provide temporal regulation of chromatin accessibility and transcription. RECENT FINDINGS: Fluorescence-activated cell sorting coupled with whole transcriptome sequencing revealed for the first time that quiescent and proliferating skeletal muscle stem cells exhibit a process of metabolic reprogramming, from fatty-acid oxidation during quiescence to glycolysis during proliferation. Using a combination of immunofluorescence and chromatin immunoprecipitation sequencing, this shift in metabolism has been linked to altered availability of key metabolites essential for histone (de)acetylation and (de)methylation, including acetyl-CoA, s-adenosylmethionine and α-ketoglutarate. Importantly, these changes in metabolite availability have been linked to muscle stem-cell function. SUMMARY: Together, these results provide greater insight into how muscle stem cells interact with their local environment, with important implications for metabolic diseases, skeletal muscle regeneration and cell-transplantation therapies.


Asunto(s)
Cromatina/genética , Dieta , Epigénesis Genética , Histonas/metabolismo , Músculo Esquelético/citología , Estado Nutricional , Células Madre/metabolismo , Acetilcoenzima A/metabolismo , Acetilación , Cromatina/metabolismo , Humanos , Ácidos Cetoglutáricos/metabolismo , Metilación , Músculo Esquelético/metabolismo , Procesamiento Proteico-Postraduccional , S-Adenosilmetionina/metabolismo , Transcriptoma
10.
Philos Trans R Soc Lond B Biol Sci ; 373(1738)2018 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-29203713

RESUMEN

Duchenne muscular dystrophy is the most common and severe of the muscular dystrophies, a group of inherited myopathies caused by different genetic mutations leading to aberrant expression or complete absence of cytoskeletal proteins. Dystrophic muscles are prone to injury, and regenerate poorly after damage. Remorseless cycles of muscle fibre breakdown and incomplete repair lead to progressive and severe muscle wasting, weakness and premature death. Many other conditions are similarly characterized by muscle wasting, including sarcopenia, cancer cachexia, sepsis, denervation, burns, and chronic obstructive pulmonary disease. Muscle trauma and loss of mass and physical capacity can significantly compromise quality of life for patients. Exercise and nutritional interventions are unlikely to halt or reverse the conditions, and strategies promoting muscle anabolism have limited clinical acceptance. Heat shock proteins (HSPs) are molecular chaperones that help proteins fold back to their original conformation and restore function. Since many muscle wasting conditions have pathophysiologies where inflammation, atrophy and weakness are indicated, increasing HSP expression in skeletal muscle may have therapeutic potential. This review will provide evidence supporting HSP induction for muscular dystrophy and other muscle wasting conditions.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'.


Asunto(s)
Proteínas de Choque Térmico/uso terapéutico , Distrofia Muscular de Duchenne/terapia , Animales , Proteínas de Choque Térmico/genética , Humanos , Ratones , Distrofia Muscular de Duchenne/genética , Ratas
11.
Proc Natl Acad Sci U S A ; 114(40): E8372-E8381, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28916735

RESUMEN

The mammalian heart undergoes maturation during postnatal life to meet the increased functional requirements of an adult. However, the key drivers of this process remain poorly defined. We are currently unable to recapitulate postnatal maturation in human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), limiting their potential as a model system to discover regenerative therapeutics. Here, we provide a summary of our studies, where we developed a 96-well device for functional screening in human pluripotent stem cell-derived cardiac organoids (hCOs). Through interrogation of >10,000 organoids, we systematically optimize parameters, including extracellular matrix (ECM), metabolic substrate, and growth factor conditions, that enhance cardiac tissue viability, function, and maturation. Under optimized maturation conditions, functional and molecular characterization revealed that a switch to fatty acid metabolism was a central driver of cardiac maturation. Under these conditions, hPSC-CMs were refractory to mitogenic stimuli, and we found that key proliferation pathways including ß-catenin and Yes-associated protein 1 (YAP1) were repressed. This proliferative barrier imposed by fatty acid metabolism in hCOs could be rescued by simultaneous activation of both ß-catenin and YAP1 using genetic approaches or a small molecule activating both pathways. These studies highlight that human organoids coupled with higher-throughput screening platforms have the potential to rapidly expand our knowledge of human biology and potentially unlock therapeutic strategies.


Asunto(s)
Factores Biológicos/metabolismo , Puntos de Control del Ciclo Celular , Miocitos Cardíacos/metabolismo , Organoides/metabolismo , Células Madre Pluripotentes/metabolismo , Regeneración/fisiología , Adulto , Animales , Diferenciación Celular , Daño del ADN , Humanos , Masculino , Miocitos Cardíacos/citología , Organoides/citología , Células Madre Pluripotentes/citología , Ratas Sprague-Dawley
12.
Methods Mol Biol ; 1668: 61-73, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28842902

RESUMEN

Skeletal muscle stem cells (MuSCs) derived from the somatic mesoderm play a critical role in successful muscle regeneration following injury and trauma. MuSCs have been found to undergo rapid changes in metabolism following a change in cell state, such as that which occurs during the transition from quiescence to an actively proliferating state. There is mounting evidence that metabolism is critically important in the regulation of quiescence, activation, and differentiation and thus the development of new techniques that aim to further probe the metabolism of MuSCs is essential. The Seahorse XF Bioanalyzer is a powerful tool that simultaneously measures the extracellular rate of change in oxygen partial pressure and pH, providing a method to measure mitochondrial respiration and lactate production. In this chapter, we describe the use of key metabolic inhibitors that allow for the investigation of mitochondrial substrate utilization in primary MuSCs.


Asunto(s)
Mitocondrias Musculares/metabolismo , Mioblastos Esqueléticos/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Compuestos Epoxi/farmacología , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Concentración de Iones de Hidrógeno , Hipoglucemiantes/farmacología , Mitocondrias Musculares/efectos de los fármacos , Mioblastos Esqueléticos/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Smegmamorpha , Sulfuros/farmacología , Tiadiazoles/farmacología
13.
Methods Mol Biol ; 1556: 245-253, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28247354

RESUMEN

Cellular metabolism has recently been identified as an important regulator of cell identity, with several adult stem cell populations having been observed to undergo a shift in metabolism during important changes in cell state, such as during the shift from quiescence to proliferation. In this chapter, a method to characterize the metabolism of quiescent skeletal muscle stem cells is presented. This technique will allow for the comparison of quiescent muscle stem cells isolated from two or more different mouse models.


Asunto(s)
Bioensayo/métodos , Metabolismo Energético , Mitocondrias/metabolismo , Músculo Esquelético/citología , Fase de Descanso del Ciclo Celular , Células Madre/citología , Células Madre/metabolismo , Técnicas Biosensibles , Técnicas de Cultivo de Célula , Separación Celular , Glucólisis , Fosforilación Oxidativa , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo
14.
JCI Insight ; 1(5)2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27182554

RESUMEN

The transforming growth factor-ß (TGF-ß) signaling network is a critical regulator of skeletal muscle mass and function and, thus, is an attractive therapeutic target for combating muscle disease, but the underlying mechanisms of action remain undetermined. We report that follistatin-based interventions (which modulate TGF-ß network activity) can promote muscle hypertrophy that ameliorates aging-associated muscle wasting. However, the muscles of old sarcopenic mice demonstrate reduced response to follistatin compared with healthy young-adult musculature. Quantitative proteomic and transcriptomic analyses of young-adult muscles identified a transcription/translation signature elicited by follistatin exposure, which included repression of ankyrin repeat and SOCS box protein 2 (Asb2). Increasing expression of ASB2 reduced muscle mass, thereby demonstrating that Asb2 is a TGF-ß network-responsive negative regulator of muscle mass. In contrast to young-adult muscles, sarcopenic muscles do not exhibit reduced ASB2 abundance with follistatin exposure. Moreover, preventing repression of ASB2 in young-adult muscles diminished follistatin-induced muscle hypertrophy. These findings provide insight into the program of transcription and translation events governing follistatin-mediated adaptation of skeletal muscle attributes and identify Asb2 as a regulator of muscle mass implicated in the potential mechanistic dysfunction between follistatin-mediated muscle growth in young and old muscles.

15.
Cancer Res ; 76(12): 3618-28, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27197176

RESUMEN

Subpopulations of cancer stem-like cells (CSC) are thought to drive tumor progression and posttreatment recurrence in multiple solid tumors. However, the mechanisms that maintain stable proportions of self-renewing CSC within heterogeneous tumors under homeostatic conditions remain poorly understood. Progastrin is a secreted peptide that exhibits tumor-forming potential in colorectal cancer, where it regulates pathways known to modulate colon CSC behaviors. In this study, we investigated the role of progastrin in regulating CSC phenotype in advanced colorectal cancer. Progastrin expression and secretion were highly enriched in colon CSC isolated from human colorectal cancer cell lines and colon tumor biopsies. Progastrin expression promoted CSC self-renewal and survival, whereas its depletion by RNA interference-mediated or antibody-mediated strategies altered the homeostatic proportions of CSC cells within heterogeneous colorectal cancer tumors. Progastrin downregulation also decreased the frequency of ALDH(high) cells, impairing their tumor-initiating potential, and inhibited the high glycolytic activity of ALDH(high) CSC to limit their self-renewal capability. Taken together, our results show how colorectal CSC maintain their tumor-initiating and self-renewal capabilities by secreting progastrin, thereby contributing to the tumor microenvironment to support malignancy. Cancer Res; 76(12); 3618-28. ©2016 AACR.


Asunto(s)
Neoplasias del Colon/patología , Gastrinas/fisiología , Células Madre Neoplásicas/fisiología , Precursores de Proteínas/fisiología , Aldehído Deshidrogenasa/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Humanos , Ratones , Microambiente Tumoral
16.
Stem Cells Int ; 2016: 5725927, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26880971

RESUMEN

An unresolved and critically important question in skeletal muscle biology is how muscle stem cells initiate and regulate the genetic program during muscle development. Epigenetic dynamics are essential for cellular development and organogenesis in early life and it is becoming increasingly clear that epigenetic remodeling may also be responsible for the cellular adaptations that occur in later life. DNA methylation of cytosine bases within CpG dinucleotide pairs is an important epigenetic modification that reduces gene expression when located within a promoter or enhancer region. Recent advances in the field suggest that epigenetic regulation is essential for skeletal muscle stem cell identity and subsequent cell development. This review summarizes what is currently known about how skeletal muscle stem cells regulate the myogenic program through DNA methylation, discusses a novel role for metabolism in this process, and addresses DNA methylation dynamics in adult skeletal muscle in response to physical activity.

17.
Cell Stem Cell ; 17(6): 651-662, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26637942

RESUMEN

For many years, stem cell metabolism was viewed as a byproduct of cell fate status rather than an active regulatory mechanism; however, there is now a growing appreciation that metabolic pathways influence epigenetic changes associated with lineage commitment, specification, and self-renewal. Here we review how metabolites generated during glycolytic and oxidative processes are utilized in enzymatic reactions leading to epigenetic modifications and transcriptional regulation. We discuss how "metabolic reprogramming" contributes to global epigenetic changes in the context of naive and primed pluripotent states, somatic reprogramming, and hematopoietic and skeletal muscle tissue stem cells, and we discuss the implications for regenerative medicine.


Asunto(s)
Epigénesis Genética , Regulación de la Expresión Génica , Células Madre/citología , Animales , Carbono/química , Diferenciación Celular , Linaje de la Célula , Reprogramación Celular , Ciclo del Ácido Cítrico , Células Madre Embrionarias/citología , Glucólisis , Histonas/química , Humanos , Células Madre Pluripotentes Inducidas/citología , Fosforilación Oxidativa , Oxígeno/química , Células Madre Pluripotentes/citología , Proteínas/química , Transcripción Genética
18.
J Neurosci Res ; 93(7): 1147-56, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25677687

RESUMEN

Silent information regulators (SIRTs) have been shown to deacetylate a range of metabolic enzymes, including those in glycolysis and the Krebs cycle, and thus alter their activity. SIRTs require NAD(+) for their activity, linking cellular energy status to enzyme activity. To examine the impact of SIRT1 modulation on oxidative metabolism, this study tests the effect of ligands that are either SIRT-activating compounds (resveratrol and SRT1720) or SIRT inhibitors (EX527) on the metabolism of (13)C-enriched substrates by guinea pig brain cortical tissue slices with (13)C and (1)H nuclear magnetic resonance spectroscopy. Resveratrol increased lactate labeling but decreased incorporation of (13)C into Krebs cycle intermediates, consistent with effects on AMPK and inhibition of the F0/F1-ATPase. By testing with resveratrol that was directly applied to astrocytes with a Seahorse analyzer, increased glycolytic shift and increased mitochondrial proton leak resulting from interactions of resveratrol with the mitochondrial electron transport chain were revealed. SRT1720, by contrast, stimulated incorporation of (13)C into Krebs cycle intermediates and reduced incorporation into lactate, although the inhibitor EX527 paradoxically also increased Krebs cycle (13)C incorporation. In summary, the various SIRT1 modulators show distinct acute effects on oxidative metabolism. The strong effects of resveratrol on the mitochondrial respiratory chain and on glycolysis suggest that caution should be used in attempts to increase bioavailability of this compound in the CNS.


Asunto(s)
Antioxidantes/farmacología , Encéfalo , Ácido Láctico/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estilbenos/farmacología , Animales , Área Bajo la Curva , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/ultraestructura , Carbazoles/farmacología , Isótopos de Carbono/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Cobayas , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Imagen por Resonancia Magnética , Masculino , Estrés Oxidativo/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Resveratrol , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/metabolismo
19.
Cell Stem Cell ; 16(2): 171-83, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25600643

RESUMEN

Stem cells undergo a shift in metabolic substrate utilization during specification and/or differentiation, a process that has been termed metabolic reprogramming. Here, we report that during the transition from quiescence to proliferation, skeletal muscle stem cells experience a metabolic switch from fatty acid oxidation to glycolysis. This reprogramming of cellular metabolism decreases intracellular NAD(+) levels and the activity of the histone deacetylase SIRT1, leading to elevated H4K16 acetylation and activation of muscle gene transcription. Selective genetic ablation of the SIRT1 deacetylase domain in skeletal muscle results in increased H4K16 acetylation and deregulated activation of the myogenic program in SCs. Moreover, mice with muscle-specific inactivation of the SIRT1 deacetylase domain display reduced myofiber size, impaired muscle regeneration, and derepression of muscle developmental genes. Overall, these findings reveal how metabolic cues can be mechanistically translated into epigenetic modifications that regulate skeletal muscle stem cell biology.


Asunto(s)
Epigénesis Genética , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , NAD/metabolismo , Sirtuina 1/metabolismo , Células Madre/citología , Células Madre/metabolismo , Acetilación , Animales , Epigénesis Genética/genética , Histonas/metabolismo , Ratones
20.
PLoS One ; 9(7): e101379, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25000590

RESUMEN

Muscles can be injured in different ways and the trauma and subsequent loss of function and physical capacity can impact significantly on the lives of patients through physical impairments and compromised quality of life. The relative success of muscle repair after injury will largely determine the extent of functional recovery. Unfortunately, regenerative processes are often slow and incomplete, and so developing novel strategies to enhance muscle regeneration is important. While the capacity to enhance muscle repair by stimulating ß2-adrenoceptors (ß-ARs) using ß2-AR agonists (ß2-agonists) has been demonstrated previously, the exact role ß-ARs play in regulating the regenerative process remains unclear. To investigate ß-AR-mediated signaling in muscle regeneration after myotoxic damage, we examined the regenerative capacity of tibialis anterior and extensor digitorum longus muscles from mice lacking either ß1-AR (ß1-KO) and/or ß2-ARs (ß2-KO), testing the hypothesis that muscles from mice lacking the ß2-AR would exhibit impaired functional regeneration after damage compared with muscles from ß1-KO or ß1/ß2-AR null (ß1/ß2-KO) KO mice. At 7 days post-injury, regenerating muscles from ß1/ß2-KO mice produced less force than those of controls but muscles from ß1-KO or ß2-KO mice did not exhibit any delay in functional restoration. Compared with controls, ß1/ß2-KO mice exhibited an enhanced inflammatory response to injury, which delayed early muscle regeneration, but an enhanced myoblast proliferation later during regeneration ensured a similar functional recovery (to controls) by 14 days post-injury. This apparent redundancy in the ß-AR signaling pathway was unexpected and may have important implications for manipulating ß-AR signaling to improve the rate, extent and efficacy of muscle regeneration to enhance functional recovery after injury.


Asunto(s)
Diferenciación Celular , Músculo Esquelético/fisiología , Mioblastos/citología , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Regeneración , Animales , Proliferación Celular , Técnicas de Inactivación de Genes , Ratones , Fuerza Muscular , Músculo Esquelético/anatomía & histología , Músculo Esquelético/citología , Tamaño de los Órganos , Receptores Adrenérgicos beta 1/deficiencia , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/deficiencia , Receptores Adrenérgicos beta 2/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA