Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 95(3): 809-52, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26109340

RESUMEN

The neuromuscular junction is the chemical synapse between motor neurons and skeletal muscle fibers. It is designed to reliably convert the action potential from the presynaptic motor neuron into the contraction of the postsynaptic muscle fiber. Diseases that affect the neuromuscular junction may cause failure of this conversion and result in loss of ambulation and respiration. The loss of motor input also causes muscle wasting as muscle mass is constantly adapted to contractile needs by the balancing of protein synthesis and protein degradation. Finally, neuromuscular activity and muscle mass have a major impact on metabolic properties of the organisms. This review discusses the mechanisms involved in the development and maintenance of the neuromuscular junction, the consequences of and the mechanisms involved in its dysfunction, and its role in maintaining muscle mass during aging. As life expectancy is increasing, loss of muscle mass during aging, called sarcopenia, has emerged as a field of high medical need. Interestingly, aging is also accompanied by structural changes at the neuromuscular junction, suggesting that the mechanisms involved in neuromuscular junction maintenance might be disturbed during aging. In addition, there is now evidence that behavioral paradigms and signaling pathways that are involved in longevity also affect neuromuscular junction stability and sarcopenia.


Asunto(s)
Músculo Esquelético/inervación , Unión Neuromuscular/crecimiento & desarrollo , Sarcopenia/fisiopatología , Transmisión Sináptica , Acetilcolina/metabolismo , Factores de Edad , Animales , Humanos , Modelos Animales , Contracción Muscular , Fuerza Muscular , Músculo Esquelético/patología , Unión Neuromuscular/metabolismo , Receptores Colinérgicos/metabolismo , Sarcopenia/metabolismo , Sarcopenia/patología
2.
Am J Physiol Endocrinol Metab ; 323(2): E133-E144, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35723227

RESUMEN

Mammalian target of rapamycin (mTOR) kinase is an essential hub where nutrients and growth factors converge to control cellular metabolism. mTOR interacts with different accessory proteins to form complexes 1 and 2 (mTORC), and each complex has different intracellular targets. Although mTORC1's role in ß-cells has been extensively studied, less is known about mTORC2's function in ß-cells. Here, we show that mice with constitutive and inducible ß-cell-specific deletion of RICTOR (ßRicKO and ißRicKO mice, respectively) are glucose intolerant due to impaired insulin secretion when glucose is injected intraperitoneally. Decreased insulin secretion in ßRicKO islets was caused by abnormal actin polymerization. Interestingly, when glucose was administered orally, no difference in glucose homeostasis and insulin secretion were observed, suggesting that incretins are counteracting the mTORC2 deficiency. Mechanistically, glucagon-like peptide-1 (GLP-1), but not gastric inhibitory polypeptide (GIP), rescued insulin secretion in vivo and in vitro by improving actin polymerization in ßRicKO islets. In conclusion, mTORC2 regulates glucose-stimulated insulin secretion by promoting actin filament remodeling.NEW & NOTEWORTHY The current studies uncover a novel mechanism linking mTORC2 signaling to glucose-stimulated insulin secretion by modulation of the actin filaments. This work also underscores the important role of GLP-1 in rescuing defects in insulin secretion by modulating actin polymerization and suggests that this effect is independent of mTORC2 signaling.


Asunto(s)
Actinas , Insulina , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Secreción de Insulina , Mamíferos/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Serina-Treonina Quinasas TOR/metabolismo
3.
Hum Mol Genet ; 29(8): 1253-1273, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32129442

RESUMEN

Some mutations affecting dynamin 2 (DNM2) can cause dominantly inherited Charcot-Marie-Tooth (CMT) neuropathy. Here, we describe the analysis of mice carrying the DNM2 K562E mutation which has been associated with dominant-intermediate CMT type B (CMTDIB). Contrary to our expectations, heterozygous DNM2 K562E mutant mice did not develop definitive signs of an axonal or demyelinating neuropathy. Rather, we found a primary myopathy-like phenotype in these mice. A likely interpretation of these results is that the lack of a neuropathy in this mouse model has allowed the unmasking of a primary myopathy due to the DNM2 K562E mutation which might be overshadowed by the neuropathy in humans. Consequently, we hypothesize that a primary myopathy may also contribute to the disease mechanism in some CMTDIB patients. We propose that these findings should be considered in the evaluation of patients, the determination of the underlying disease processes and the development of tailored potential treatment strategies.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Dinamina II/deficiencia , Enfermedades Musculares/genética , Miopatías Estructurales Congénitas/genética , Animales , Axones/metabolismo , Axones/patología , Enfermedad de Charcot-Marie-Tooth/patología , Dinamina II/genética , Heterocigoto , Humanos , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/patología , Mutación/genética , Miopatías Estructurales Congénitas/patología , Fenotipo
4.
Development ; 146(7)2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30872276

RESUMEN

The formation of multi-nucleated muscle fibers from progenitors requires the fine-tuned and coordinated regulation of proliferation, differentiation and fusion, both during development and after injury in the adult. Although some of the key factors that are involved in the different steps are well known, how intracellular signals are coordinated and integrated is largely unknown. Here, we investigated the role of the cell-growth regulator mTOR by eliminating essential components of the mTOR complexes 1 (mTORC1) and 2 (mTORC2) in mouse muscle progenitors. We show that inactivation of mTORC1, but not mTORC2, in developing muscle causes perinatal death. In the adult, mTORC1 deficiency in muscle stem cells greatly impinges on injury-induced muscle regeneration. These phenotypes are because of defects in the proliferation and fusion capacity of the targeted muscle progenitors. However, mTORC1-deficient muscle progenitors partially retain their myogenic function. Hence, our results show that mTORC1 and not mTORC2 is an important regulator of embryonic and adult myogenesis, and they point to alternative pathways that partially compensate for the loss of mTORC1.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Immunoblotting , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Desarrollo de Músculos/genética , Desarrollo de Músculos/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasas TOR/genética
5.
Proc Natl Acad Sci U S A ; 116(32): 16111-16120, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31320589

RESUMEN

Brain-derived neurotrophic factor (BDNF) influences the differentiation, plasticity, and survival of central neurons and likewise, affects the development of the neuromuscular system. Besides its neuronal origin, BDNF is also a member of the myokine family. However, the role of skeletal muscle-derived BDNF in regulating neuromuscular physiology in vivo remains unclear. Using gain- and loss-of-function animal models, we show that muscle-specific ablation of BDNF shifts the proportion of muscle fibers from type IIB to IIX, concomitant with elevated slow muscle-type gene expression. Furthermore, BDNF deletion reduces motor end plate volume without affecting neuromuscular junction (NMJ) integrity. These morphological changes are associated with slow muscle function and a greater resistance to contraction-induced fatigue. Conversely, BDNF overexpression promotes a fast muscle-type gene program and elevates glycolytic fiber number. These findings indicate that BDNF is required for fiber-type specification and provide insights into its potential modulation as a therapeutic target in muscle diseases.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Glucólisis , Fibras Musculares Esqueléticas/metabolismo , Animales , Marcha , Regulación de la Expresión Génica , Locomoción , Ratones Noqueados , Modelos Biológicos , Placa Motora/metabolismo , Contracción Muscular , Fatiga Muscular , Especificidad de Órganos , Oxidación-Reducción , Condicionamiento Físico Animal , Transducción de Señal
6.
Nucleic Acids Res ; 47(14): 7618-7632, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31127278

RESUMEN

Spinal Muscular Atrophy results from loss-of-function mutations in SMN1 but correcting aberrant splicing of SMN2 offers hope of a cure. However, current splice therapy requires repeated infusions and is expensive. We previously rescued SMA mice by promoting the inclusion of a defective exon in SMN2 with germline expression of Exon-Specific U1 snRNAs (ExspeU1). Here we tested viral delivery of SMN2 ExspeU1s encoded by adeno-associated virus AAV9. Strikingly the virus increased SMN2 exon 7 inclusion and SMN protein levels and rescued the phenotype of mild and severe SMA mice. In the severe mouse, the treatment improved the neuromuscular function and increased the life span from 10 to 219 days. ExspeU1 expression persisted for 1 month and was effective at around one five-hundredth of the concentration of the endogenous U1snRNA. RNA-seq analysis revealed our potential drug rescues aberrant SMA expression and splicing profiles, which are mostly related to DNA damage, cell-cycle control and acute phase response. Vastly overexpressing ExspeU1 more than 100-fold above the therapeutic level in human cells did not significantly alter global gene expression or splicing. These results indicate that AAV-mediated delivery of a modified U1snRNP particle may be a novel therapeutic option against SMA.


Asunto(s)
Terapia Genética/métodos , Atrofia Muscular Espinal/terapia , Distrofia Muscular Animal/terapia , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Exones/genética , Células HEK293 , Humanos , Ratones Noqueados , Atrofia Muscular Espinal/genética , Distrofia Muscular Animal/genética , Mutación , Empalme del ARN , Ribonucleoproteína Nuclear Pequeña U1/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo
7.
J Allergy Clin Immunol ; 145(1): 283-300.e8, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31401286

RESUMEN

BACKGROUND: Perturbation of epidermal barrier formation will profoundly compromise overall skin function, leading to a dry and scaly, ichthyosis-like skin phenotype that is the hallmark of a broad range of skin diseases, including ichthyosis, atopic dermatitis, and a multitude of clinical eczema variants. An overarching molecular mechanism that orchestrates the multitude of factors controlling epidermal barrier formation and homeostasis remains to be elucidated. OBJECTIVE: Here we highlight a specific role of mammalian target of rapamycin complex 2 (mTORC2) signaling in epidermal barrier formation. METHODS: Epidermal mTORC2 signaling was specifically disrupted by deleting rapamycin-insensitive companion of target of rapamycin (Rictor), encoding an essential subunit of mTORC2 in mouse epidermis (epidermis-specific homozygous Rictor deletion [RicEKO] mice). Epidermal structure and barrier function were investigated through a combination of gene expression, biochemical, morphological and functional analysis in RicEKO and control mice. RESULTS: RicEKO newborns displayed an ichthyosis-like phenotype characterized by dysregulated epidermal de novo lipid synthesis, altered lipid lamellae structure, and aberrant filaggrin (FLG) processing. Despite a compensatory transcriptional epidermal repair response, the protective epidermal function was impaired in RicEKO mice, as revealed by increased transepidermal water loss, enhanced corneocyte fragility, decreased dendritic epidermal T cells, and an exaggerated percutaneous immune response. Restoration of Akt-Ser473 phosphorylation in mTORC2-deficient keratinocytes through expression of constitutive Akt rescued FLG processing. CONCLUSION: Our findings reveal a critical metabolic signaling relay of barrier formation in which epidermal mTORC2 activity controls FLG processing and de novo epidermal lipid synthesis during cornification. Our findings provide novel mechanistic insights into epidermal barrier formation and could open up new therapeutic opportunities to restore defective epidermal barrier conditions.


Asunto(s)
Epidermis , Proteínas de Filamentos Intermediarios , Lípidos , Procesamiento Proteico-Postraduccional/inmunología , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal/inmunología , Animales , Epidermis/inmunología , Epidermis/metabolismo , Proteínas Filagrina , Ictiosis/genética , Ictiosis/inmunología , Ictiosis/metabolismo , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/inmunología , Proteínas de Filamentos Intermediarios/metabolismo , Lípidos/biosíntesis , Lípidos/genética , Lípidos/inmunología , Ratones , Ratones Noqueados , Procesamiento Proteico-Postraduccional/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/inmunología , Transducción de Señal/genética
8.
Development ; 144(24): 4604-4615, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29061639

RESUMEN

The low-density lipoprotein receptor-related protein 4 (LRP4) is essential in muscle fibers for the establishment of the neuromuscular junction. Here, we show that LRP4 is also expressed by embryonic cortical and hippocampal neurons, and that downregulation of LRP4 in these neurons causes a reduction in density of synapses and number of primary dendrites. Accordingly, overexpression of LRP4 in cultured neurons had the opposite effect inducing more but shorter primary dendrites with an increased number of spines. Transsynaptic tracing mediated by rabies virus revealed a reduced number of neurons presynaptic to the cortical neurons in which LRP4 was knocked down. Moreover, neuron-specific knockdown of LRP4 by in utero electroporation of LRP4 miRNA in vivo also resulted in neurons with fewer primary dendrites and a lower density of spines in the developing cortex and hippocampus. Collectively, our results demonstrate an essential and novel role of neuronal LRP4 in dendritic development and synaptogenesis in the CNS.


Asunto(s)
Corteza Cerebral/metabolismo , Dendritas/metabolismo , Hipocampo/metabolismo , Receptores de LDL/metabolismo , Sinapsis/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Técnicas de Inactivación de Genes , Hipocampo/citología , Hipocampo/embriología , Proteínas Relacionadas con Receptor de LDL , Ratones , Ratones Endogámicos C57BL , Rabia/patología , Virus de la Rabia/crecimiento & desarrollo , Receptores de LDL/genética
9.
J Neurosci ; 38(17): 4243-4258, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29626165

RESUMEN

Collagen XIII occurs as both a transmembrane-bound and a shed extracellular protein and is able to regulate the formation and function of neuromuscular synapses. Its absence results in myasthenia: presynaptic and postsynaptic defects at the neuromuscular junction (NMJ), leading to destabilization of the motor nerves, muscle regeneration and atrophy. Mutations in COL13A1 have recently been found to cause congenital myasthenic syndrome, characterized by fatigue and chronic muscle weakness, which may be lethal. We show here that muscle defects in collagen XIII-deficient mice stabilize in adulthood, so that the disease is not progressive until very late. Sciatic nerve crush was performed to examine how the lack of collagen XIII or forced expression of its transmembrane form affects the neuromuscular synapse regeneration and functional recovery following injury. We show that collagen XIII-deficient male mice are unable to achieve complete NMJ regeneration and functional recovery. This is mainly attributable to presynaptic defects that already existed in the absence of collagen XIII before injury. Shedding of the ectodomain is not required, as the transmembrane form of collagen XIII alone fully rescues the phenotype. Thus, collagen XIII could serve as a therapeutic agent in cases of injury-induced PNS regeneration and functional recovery. We conclude that intrinsic alterations at the NMJ in Col13a1-/- mice contribute to impaired and incomplete NMJ regeneration and functional recovery after peripheral nerve injury. However, such alterations do not progress once they have stabilized in early adulthood, emphasizing the role of collagen XIII in NMJ maturation.SIGNIFICANCE STATEMENT Collagen XIII is required for gaining and maintaining the normal size, complexity, and functional capacity of neuromuscular synapses. Loss-of-function mutations in COL13A1 cause congenital myasthenic syndrome 19, characterized by postnatally progressive muscle fatigue, which compromises patients' functional capacity. We show here in collagen XIII-deficient mice that the disease stabilizes in adulthood once the NMJs have matured. This study also describes a relevant contribution of the altered NMJ morphology and function to neuromuscular synapses, and PNS regeneration and functional recovery in collagen XIII-deficient mice after peripheral nerve injury. Correlating the animal model data on collagen XIII-associated congenital myasthenic syndrome, it can be speculated that neuromuscular connections in congenital myasthenic syndrome patients are not able to fully regenerate and restore normal functionality if exposed to peripheral nerve injury.


Asunto(s)
Colágeno Tipo XIII/metabolismo , Regeneración Nerviosa , Unión Neuromuscular/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Animales , Colágeno Tipo XIII/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Neuromuscular/fisiología , Traumatismos de los Nervios Periféricos/fisiopatología , Recuperación de la Función
10.
Am J Physiol Cell Physiol ; 313(6): C604-C611, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28971834

RESUMEN

Mechanistic target of rapamycin (mTOR) resides as two complexes within skeletal muscle. mTOR complex 1 [mTORC1-regulatory associated protein of mTOR (Raptor) positive] regulates skeletal muscle growth, whereas mTORC2 [rapamycin-insensitive companion of mTOR (Rictor) positive] regulates insulin sensitivity. To examine the regulation of these complexes in human skeletal muscle, we utilized immunohistochemical analysis to study the localization of mTOR complexes before and following protein-carbohydrate feeding (FED) and resistance exercise plus protein-carbohydrate feeding (EXFED) in a unilateral exercise model. In basal samples, mTOR and the lysosomal marker lysosomal associated membrane protein 2 (LAMP2) were highly colocalized and remained so throughout. In the FED and EXFED states, mTOR/LAMP2 complexes were redistributed to the cell periphery [wheat germ agglutinin (WGA)-positive staining] (time effect; P = 0.025), with 39% (FED) and 26% (EXFED) increases in mTOR/WGA association observed 1 h post-feeding/exercise. mTOR/WGA colocalization continued to increase in EXFED at 3 h (48% above baseline) whereas colocalization decreased in FED (21% above baseline). A significant effect of condition (P = 0.05) was noted suggesting mTOR/WGA colocalization was greater during EXFED. This pattern was replicated in Raptor/WGA association, where a significant difference between EXFED and FED was noted at 3 h post-exercise/feeding (P = 0.014). Rictor/WGA colocalization remained unaltered throughout the trial. Alterations in mTORC1 cellular location coincided with elevated S6K1 kinase activity, which rose to a greater extent in EXFED compared with FED at 1 h post-exercise/feeding (P < 0.001), and only remained elevated in EXFED at the 3 h time point (P = 0.037). Collectively these data suggest that mTORC1 redistribution within the cell is a fundamental response to resistance exercise and feeding, whereas mTORC2 is predominantly situated at the sarcolemma and does not alter localization.


Asunto(s)
Ingestión de Alimentos , Metabolismo Energético , Ejercicio Físico , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Músculo Cuádriceps/enzimología , Adulto , Carbohidratos de la Dieta/administración & dosificación , Proteínas en la Dieta/administración & dosificación , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/enzimología , Masculino , Contracción Muscular , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Proteína Reguladora Asociada a mTOR/metabolismo , Entrenamiento de Fuerza , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sarcolema/enzimología , Factores de Tiempo , Adulto Joven
11.
Am J Pathol ; 186(2): 324-36, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26683665

RESUMEN

The mammalian target of rapamycin complex 1 (mTORC1) inhibitor rapamycin and its analogs are being increasingly used in solid-organ transplantation. A commonly reported side effect is male subfertility to infertility, yet the precise mechanisms of mTOR interference with male fertility remain obscure. With the use of a conditional mouse genetic approach we demonstrate that deficiency of mTORC1 in the epithelial derivatives of the Wolffian duct is sufficient to cause male infertility. Analysis of spermatozoa from Raptor fl/fl*KspCre mice revealed an overall decreased motility pattern. Both epididymis and seminal vesicles displayed extensive organ regression with increasing age. Histologic and ultrastructural analyses demonstrated increased amounts of destroyed and absorbed spermatozoa in different segments of the epididymis. Mechanistically, genetic and pharmacologic mTORC1 inhibition was associated with an impaired cellular metabolism and a disturbed protein secretion of epididymal epithelial cells. Collectively, our data highlight the role of mTORC1 to preserve the function of the epididymis, ductus deferens, and the seminal vesicles. We thus reveal unexpected new insights into the frequently observed mTORC1 inhibitor side effect of male infertility in transplant recipients.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Fertilidad/efectos de los fármacos , Complejos Multiproteicos/efectos de los fármacos , Vesículas Seminales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/efectos de los fármacos , Animales , Masculino , Mamíferos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Transgénicos , Fosforilación , Vesículas Seminales/metabolismo , Factores de Transcripción/metabolismo
12.
PLoS Genet ; 10(1): e1004145, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24497849

RESUMEN

WNT signaling has been implicated in both embryonic and postnatal bone formation. However, the pertinent WNT ligands and their downstream signaling mechanisms are not well understood. To investigate the osteogenic capacity of WNT7B and WNT5A, both normally expressed in the developing bone, we engineered mouse strains to express either protein in a Cre-dependent manner. Targeted induction of WNT7B, but not WNT5A, in the osteoblast lineage dramatically enhanced bone mass due to increased osteoblast number and activity; this phenotype began in the late-stage embryo and intensified postnatally. Similarly, postnatal induction of WNT7B in Runx2-lineage cells greatly stimulated bone formation. WNT7B activated mTORC1 through PI3K-AKT signaling. Genetic disruption of mTORC1 signaling by deleting Raptor in the osteoblast lineage alleviated the WNT7B-induced high-bone-mass phenotype. Thus, WNT7B promotes bone formation in part through mTORC1 activation.


Asunto(s)
Complejos Multiproteicos/genética , Osteogénesis/genética , Proteínas Proto-Oncogénicas/genética , Serina-Treonina Quinasas TOR/genética , Proteínas Wnt/genética , Animales , Diferenciación Celular , Linaje de la Célula , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/biosíntesis , Osteoblastos/citología , Proteínas Proto-Oncogénicas/biosíntesis , Serina-Treonina Quinasas TOR/biosíntesis , Proteínas Wnt/biosíntesis , Vía de Señalización Wnt , Proteína Wnt-5a
13.
Proc Natl Acad Sci U S A ; 111(27): E2817-26, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958889

RESUMEN

Mammalian target of rapamycin complex 1 (mTORC1) is a key regulator of cell metabolism and autophagy. Despite widespread clinical use of mTORC1 inhibitors, the role of mTORC1 in renal tubular function and kidney homeostasis remains elusive. By using constitutive and inducible deletion of conditional Raptor alleles in renal tubular epithelial cells, we discovered that mTORC1 deficiency caused a marked concentrating defect, loss of tubular cells, and slowly progressive renal fibrosis. Transcriptional profiling revealed that mTORC1 maintains renal tubular homeostasis by controlling mitochondrial metabolism and biogenesis as well as transcellular transport processes involved in countercurrent multiplication and urine concentration. Although mTORC2 partially compensated for the loss of mTORC1, exposure to ischemia and reperfusion injury exaggerated the tubular damage in mTORC1-deficient mice and caused pronounced apoptosis, diminished proliferation rates, and delayed recovery. These findings identify mTORC1 as an important regulator of tubular energy metabolism and as a crucial component of ischemic stress responses.


Asunto(s)
Homeostasis/fisiología , Isquemia/fisiopatología , Túbulos Renales/fisiología , Complejos Multiproteicos/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Western Blotting , Túbulos Renales/irrigación sanguínea , Imagen por Resonancia Magnética , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/genética , Poliuria/genética , Serina-Treonina Quinasas TOR/genética , Transcripción Genética
14.
Stem Cells ; 33(4): 1359-65, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25537496

RESUMEN

Adipocytes (AdCs) and osteoblasts (OBs) are derived from mesenchymal stem cells (MSCs) and differentiation toward either lineage is both mutually exclusive and transcriptionally controlled. Recent studies implicate the mammalian target of rapamycin (mTOR) pathway as important in determining MSC fate, with inhibition of mTOR promoting OB differentiation and suppressing AdC differentiation. mTOR functions within two distinct multiprotein complexes, mTORC1 and mTORC2, each of which contains the unique adaptor protein, raptor or rictor, respectively. While compounds used to study mTOR signaling, such as rapamycin and related analogs, primarily inhibit mTORC1, prolonged exposure can also disrupt mTORC2 function, confounding interpretation of inhibitor studies. As a result, the relative contribution of mTORC1 and mTORC2 to MSC fate determination remains unclear. In this study, we generated primary mouse MSCs deficient in either Rptor (RapKO) or Rictor (RicKO) using the Cre/loxP system. Cre-mediated deletion of Rptor or Rictor resulted in impaired mTORC1 and mTORC2 signaling, respectively. Under lineage-inductive culture conditions, RapKO MSCs displayed a reduced capacity to form lipid-laden AdCs and an increased capacity to form a mineralized matrix. In contrast, RicKO MSCs displayed reduced osteogenic differentiation capacity and enhanced adipogenic differentiation potential. Taken together, our findings reveal distinct roles for mTORC1 and mTORC2 in MSC lineage commitment.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Complejos Multiproteicos/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Proliferación Celular/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Noqueados
15.
J Immunol ; 193(4): 1759-65, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25015820

RESUMEN

Invariant NKT (iNKT) cells play critical roles in bridging innate and adaptive immunity. The Raptor containing mTOR complex 1 (mTORC1) has been well documented to control peripheral CD4 or CD8 T cell effector or memory differentiation. However, the role of mTORC1 in iNKT cell development and function remains largely unknown. By using mice with T cell-restricted deletion of Raptor, we show that mTORC1 is selectively required for iNKT but not for conventional T cell development. Indeed, Raptor-deficient iNKT cells are mostly blocked at thymic stage 1-2, resulting in a dramatic decrease of terminal differentiation into stage 3 and severe reduction of peripheral iNKT cells. Moreover, residual iNKT cells in Raptor knockout mice are impaired in their rapid cytokine production upon αGalcer challenge. Bone marrow chimera studies demonstrate that mTORC1 controls iNKT differentiation in a cell-intrinsic manner. Collectively, our data provide the genetic evidence that iNKT cell development and effector functions are under the control of mTORC1 signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Diferenciación Celular/inmunología , Complejos Multiproteicos/genética , Células T Asesinas Naturales/citología , Serina-Treonina Quinasas TOR/genética , Animales , Antígenos CD/biosíntesis , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Linfocitos T CD8-positivos/inmunología , Citocinas/biosíntesis , Memoria Inmunológica , Interferón gamma/biosíntesis , Lectinas Tipo C/biosíntesis , Activación de Linfocitos/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Proteína Reguladora Asociada a mTOR , Transducción de Señal/inmunología , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
16.
J Immunol ; 193(3): 1162-70, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24981454

RESUMEN

An efficient immune response relies on the presence of T cells expressing a functional TCR. Whereas the mechanisms generating TCR diversity for antigenic recognition are well defined, what controls its surface expression is less known. In this study, we found that deletion of the mammalian target of rapamycin complex (mTORC) 2 component rictor at early stages of T cell development led to aberrant maturation and increased proteasomal degradation of nascent TCRs. Although CD127 expression became elevated, the levels of TCRs as well as CD4, CD8, CD69, Notch, and CD147 were significantly attenuated on the surface of rictor-deficient thymocytes. Diminished expression of these receptors led to suboptimal signaling, partial CD4(-)CD8(-) double-negative 4 (CD25(-)CD44(-)) proliferation, and CD4(+)CD8(+) double-positive activation as well as developmental blocks at the CD4(-)CD8(-) double-negative 3 (CD25(+)CD44(-)) and CD8-immature CD8(+) single-positive stages. Because CD147 glycosylation was also defective in SIN1-deficient fibroblasts, our findings suggest that mTORC2 is involved in the co/posttranslational processing of membrane receptors. Thus, mTORC2 impacts development via regulation of the quantity and quality of receptors important for cell differentiation.


Asunto(s)
Proteínas Portadoras/fisiología , Regulación del Desarrollo de la Expresión Génica/inmunología , Complejos Multiproteicos/fisiología , Procesamiento Proteico-Postraduccional/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/fisiología , Animales , Proteínas Portadoras/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Eliminación de Gen , Humanos , Células Jurkat , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Complejos Multiproteicos/deficiencia , Procesamiento Proteico-Postraduccional/genética , Subunidades de Proteína/deficiencia , Subunidades de Proteína/fisiología , Proteína Asociada al mTOR Insensible a la Rapamicina , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/deficiencia
17.
Biochem J ; 466(1): 123-35, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25431931

RESUMEN

The protein mammalian target of rapamycin (mTOR) is a serine/threonine kinase regulating a number of biochemical pathways controlling cell growth. mTOR exists in two complexes termed mTORC1 and mTORC2. Regulatory associated protein of mTOR (raptor) is associated with mTORC1 and is essential for its function. Ablation of raptor in skeletal muscle results in several phenotypic changes including decreased life expectancy, increased glycogen deposits and alterations of the twitch kinetics of slow fibres. In the present paper, we show that in muscle-specific raptor knockout (RamKO), the bulk of glycogen phosphorylase (GP) is mainly associated in its cAMP-non-stimulated form with sarcoplasmic reticulum (SR) membranes. In addition, 3[H]-ryanodine and 3[H]-PN200-110 equilibrium binding show a ryanodine to dihydropyridine receptors (DHPRs) ratio of 0.79 and 1.35 for wild-type (WT) and raptor KO skeletal muscle membranes respectively. Peak amplitude and time to peak of the global calcium transients evoked by supramaximal field stimulation were not different between WT and raptor KO. However, the increase in the voltage sensor-uncoupled RyRs leads to an increase of both frequency and mass of elementary calcium release events (ECRE) induced by hyper-osmotic shock in flexor digitorum brevis (FDB) fibres from raptor KO. The present study shows that the protein composition and function of the molecular machinery involved in skeletal muscle excitation-contraction (E-C) coupling is affected by mTORC1 signalling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Canales de Calcio Tipo L/genética , Complejos Multiproteicos/genética , Músculo Esquelético/metabolismo , Retículo Sarcoplasmático/metabolismo , Serina-Treonina Quinasas TOR/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/metabolismo , Potenciales Evocados/fisiología , Acoplamiento Excitación-Contracción/fisiología , Regulación de la Expresión Génica , Glucógeno Fosforilasa/genética , Glucógeno Fosforilasa/metabolismo , Contracción Isométrica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Proteína Reguladora Asociada a mTOR , Rianodina/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
18.
Proc Natl Acad Sci U S A ; 110(31): E2915-24, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23847204

RESUMEN

We describe a unique extracellular matrix (ECM) niche in the spleen, the marginal zone (MZ), characterized by the basement membrane glycoproteins, laminin α5 and agrin, that promotes formation of a specialized population of MZ B lymphocytes that respond rapidly to blood-borne antigens. Mice with reduced laminin α5 expression show reduced MZ B cells and increased numbers of newly formed (NF) transitional B cells that migrate from the bone marrow, without changes in other immune or stromal cell compartments. Transient integrin α6ß1-mediated interaction of NF B cells with laminin α5 in the MZ supports the MZ B-cell population, their long-term survival, and antibody response. Data suggest that the unique 3D structure and biochemical composition of the ECM of lymphoid organs impacts on immune cell fate.


Asunto(s)
Linfocitos B/inmunología , Médula Ósea/inmunología , Movimiento Celular/inmunología , Matriz Extracelular/inmunología , Bazo/inmunología , Agrina/genética , Agrina/inmunología , Animales , Linfocitos B/citología , Movimiento Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Matriz Extracelular/genética , Integrina alfa6beta1/genética , Integrina alfa6beta1/inmunología , Laminina/genética , Laminina/inmunología , Ratones , Ratones Noqueados , Bazo/citología
19.
J Neurosci ; 34(25): 8432-48, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24948799

RESUMEN

The mammalian target of rapamycin (mTOR) pathway integrates multiple signals and regulates crucial cell functions via the molecular complexes mTORC1 and mTORC2. These complexes are functionally dependent on their raptor (mTORC1) or rictor (mTORC2) subunits. mTOR has been associated with oligodendrocyte differentiation and myelination downstream of the PI3K/Akt pathway, but the functional contributions of individual complexes are largely unknown. We show, by oligodendrocyte-specific genetic deletion of Rptor and/or Rictor in the mouse, that CNS myelination is mainly dependent on mTORC1 function, with minor mTORC2 contributions. Myelin-associated lipogenesis and protein gene regulation are strongly reliant on mTORC1. We found that also oligodendrocyte-specific overactivation of mTORC1, via ablation of tuberous sclerosis complex 1 (TSC1), causes hypomyelination characterized by downregulation of Akt signaling and lipogenic pathways. Our data demonstrate that a delicately balanced regulation of mTORC1 activation and action in oligodendrocytes is essential for CNS myelination, which has practical overtones for understanding CNS myelin disorders.


Asunto(s)
Complejos Multiproteicos/metabolismo , Fibras Nerviosas Mielínicas/metabolismo , Oligodendroglía/metabolismo , Médula Espinal/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Femenino , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Ratones Transgénicos , Fibras Nerviosas Mielínicas/patología , Oligodendroglía/patología , Médula Espinal/patología
20.
Carcinogenesis ; 36(4): 487-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25740823

RESUMEN

Activation of signaling dependent on the mammalian target of rapamycin (mTOR) has been demonstrated in a variety of human malignancies, and our previous work suggests that mTOR complex (mTORC) 1 and mTORC2 may play unique roles in skin tumorigenesis. The purpose of these studies was to investigate the function of mTORC2-dependent pathways in skin tumor development and the maintenance of established tumors. Using mice that allow spatial and temporal control of mTORC2 in epidermis by conditional knockout of its essential component Rictor, we studied the effect of mTORC2 loss on both epidermal proliferation and chemical carcinogenesis. The results demonstrate that mTORC2 is dispensable for both normal epidermal proliferation and the hyperproliferative response to treatment with tetradecanoyl phorbol acetate (TPA). In contrast, deletion of epidermal Rictor prior to initiation in DMBA/TPA chemical carcinogenesis was sufficient to dramatically delay tumor development and resulted in reduced tumor number and size compared with control groups. Silencing of Rictor expression in tumor-bearing animals triggered regression of established tumors and increased caspase-3 cleavage without changes in proliferation. In vitro experiments demonstrate an increased sensitivity to caspase-dependent apoptosis in the absence of rictor, which is dependent on mTORC2 signaling. These studies demonstrate that mTORC2 activation is essential for keratinocyte survival, and suggest that inhibition of mTORC2 has value in chemoprevention by eliminating carcinogen-damaged cells during the early stages of tumorigenesis, and in therapy of existing tumors by restricting critical pro-survival pathways.


Asunto(s)
Proteínas Portadoras/genética , Transformación Celular Neoplásica/genética , Complejos Multiproteicos/genética , Neoplasias Cutáneas/genética , Serina-Treonina Quinasas TOR/genética , 9,10-Dimetil-1,2-benzantraceno/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/efectos de la radiación , Carcinógenos/farmacología , Caspasa 3/metabolismo , Proliferación Celular/genética , Transformación Celular Neoplásica/efectos de los fármacos , Células Cultivadas , Quimioprevención , Queratinocitos/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal , Neoplasias Cutáneas/inducido químicamente , Acetato de Tetradecanoilforbol/farmacología , Rayos Ultravioleta/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA