Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 8(7)2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-36862514

RESUMEN

Hypothalamic neurons regulate body homeostasis by sensing and integrating changes in the levels of key hormones and primary nutrients (amino acids, glucose, and lipids). However, the molecular mechanisms that enable hypothalamic neurons to detect primary nutrients remain elusive. Here, we identified l-type amino acid transporter 1 (LAT1) in hypothalamic leptin receptor-expressing (LepR-expressing) neurons as being important for systemic energy and bone homeostasis. We observed LAT1-dependent amino acid uptake in the hypothalamus, which was compromised in a mouse model of obesity and diabetes. Mice lacking LAT1 (encoded by solute carrier transporter 7a5, Slc7a5) in LepR-expressing neurons exhibited obesity-related phenotypes and higher bone mass. Slc7a5 deficiency caused sympathetic dysfunction and leptin insensitivity in LepR-expressing neurons before obesity onset. Importantly, restoring Slc7a5 expression selectively in LepR-expressing ventromedial hypothalamus neurons rescued energy and bone homeostasis in mice deficient for Slc7a5 in LepR-expressing cells. Mechanistic target of rapamycin complex-1 (mTORC1) was found to be a crucial mediator of LAT1-dependent regulation of energy and bone homeostasis. These results suggest that the LAT1/mTORC1 axis in LepR-expressing neurons controls energy and bone homeostasis by fine-tuning sympathetic outflow, thus providing in vivo evidence of the implications of amino acid sensing by hypothalamic neurons in body homeostasis.


Asunto(s)
Hipotálamo , Transportador de Aminoácidos Neutros Grandes 1 , Ratones , Animales , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo , Neuronas/metabolismo , Homeostasis/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
2.
Spine Surg Relat Res ; 6(4): 402-407, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36051676

RESUMEN

Introduction: Amino acid transporters are transmembrane proteins that are known to mediate the transfer of amino acids. As one of the amino acid transporters, LAT1, which is encoded by Slc7a5, mediates the cellular uptake of the essential amino acids. Recently, most studies have focused on examining the relationship between LAT1 and skeletal formation in terms of development. However, little is known regarding the clinical features of LAT1 in the cartilage, which might result in the development of skeletal deformities such as scoliosis. Thus, the aim of this study was to investigate the expression of L-type amino acid transporter 1 (LAT1) and its solute carrier transporter 7a5 (Slc7a5) in patients with pediatric scoliosis and to compare with the relationship between LAT1 and Slc7a5 expression and their clinical features. Methods: We have prospectively recruited 56 patients who underwent corrective spinal fusion for scoliosis. The patients comprised 40 girls and 16 boys, with a mean age of 13.1 years at the time of surgery. There were 34 idiopathic scoliosis (IS) patients, whereas 22 were congenital scoliosis (CS) patients. During the surgery, an epiphyseal part of the spinous process at apical vertebra was harvested; then, LAT1 and Slc7a5 expressions in the cartilage were evaluated. Results: As per our findings, LAT1 expression was observed in the cartilage in 60.7% (34 out of 56) of the patients. LAT1 expression in IS patients was 76%, which were statistically higher compared to 36% in CS patients. When compared with LAT1 expression, no statistical difference was noted in terms of age, gender, body mass index (BMI), Cobb angle, and Risser grade. Meanwhile, the mean Slc7a5 expression in IS patients was determined to be significantly higher than that in CS patients. No significant correlation was observed between Slc7a5 expression and age, BMI, and Cobb angle. Conclusions: LAT1 and Slc7a5 expression in IS and CS patients showed significant differences. These expressions were found to be not correlated with age, stature, and severity of the deformity.

3.
J Cell Physiol ; 237(11): 4292-4302, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36161979

RESUMEN

Scoliosis, usually diagnosed in childhood and early adolescence, is an abnormal lateral curvature of the spine. L-type amino acid transporter 1 (LAT1), encoded by solute carrier transporter 7a5 (Slc7a5), plays a crucial role in amino acid sensing and signaling in specific cell types. We previously demonstrated the pivotal role of LAT1 on bone homeostasis in mice, and the expression of LAT1/SLC7A5 in vertebral cartilage of pediatric scoliosis patients; however, its role in chondrocytes on spinal homeostasis and implications regarding the underlying mechanisms during the onset and progression of scoliosis, remain unknown. Here, we identified LAT1 in mouse chondrocytes as an important regulator of postnatal spinal homeostasis. Conditional inactivation of LAT1 in chondrocytes resulted in a postnatal-onset severe thoracic scoliosis at the early adolescent stage with normal embryonic spinal development. Histological analyses revealed that Slc7a5 deletion in chondrocytes led to general disorganization of chondrocytes in the vertebral growth plate, along with an increase in apoptosis and a decrease in cell proliferation. Furthermore, loss of Slc7a5 in chondrocytes activated the general amino acid control (GAAC) pathway but inactivated the mechanistic target of rapamycin complex 1 (mTORC1) pathway in the vertebrae. The spinal deformity in Slc7a5-deficient mice was corrected by genetic inactivation of the GAAC pathway, but not by genetic activation of the mTORC1 pathway. These findings suggest that the LAT1-GAAC pathway in chondrocytes plays a critical role in the maintenance of proper spinal homeostasis by modulating cell proliferation and survivability.


Asunto(s)
Transportador de Aminoácidos Neutros Grandes 1 , Escoliosis , Animales , Ratones , Aminoácidos , Condrocitos/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/genética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Escoliosis/genética , Escoliosis/metabolismo , Escoliosis/patología , Modelos Animales de Enfermedad
4.
Stem Cell Reports ; 17(7): 1576-1588, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35777359

RESUMEN

Bone marrow mesenchymal stem cells (MSCs) are critical regulators of postnatal bone homeostasis. Osteoporosis is characterized by bone volume and strength deterioration, partly due to MSC dysfunction. Cyclin-dependent kinase 8 (CDK8) belongs to the transcription-related CDK family. Here, CDK8 in MSCs was identified as important for bone homeostasis. CDK8 level was increased in aged MSCs along with the association with aging-related signals. Mouse genetic studies revealed that CDK8 in MSCs plays a crucial role in bone resorption and homeostasis. Mechanistically, CDK8 in MSCs extrinsically controls osteoclastogenesis through the signal transducer and transcription 1 (STAT1)-receptor activator of the nuclear factor κ Β ligand (RANKL) axis. Moreover, aged MSCs have high osteoclastogenesis-supporting activity, partly through a CDK8-dependent manner. Finally, pharmacological inhibition of CDK8 effectively repressed MSC-dependent osteoclastogenesis and prevented ovariectomy-induced osteoclastic activation and bone loss. These findings highlight that the CDK8-STAT1-RANKL axis in MSCs could play a crucial role in bone resorption and homeostasis.


Asunto(s)
Resorción Ósea , Quinasa 8 Dependiente de Ciclina/metabolismo , Células Madre Mesenquimatosas , Animales , Resorción Ósea/genética , Diferenciación Celular , Quinasa 8 Dependiente de Ciclina/genética , Femenino , Homeostasis , Células Madre Mesenquimatosas/metabolismo , Ratones , FN-kappa B/metabolismo , Osteoclastos , Osteogénesis/genética , Ligando RANK/metabolismo , Ligando RANK/farmacología
5.
Stem Cells ; 40(4): 411-422, 2022 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-35304894

RESUMEN

Extracellular signal-regulated kinase 5 (Erk5) belongs to the mitogen-activated protein kinase (MAPK) family. Previously, we demonstrated that Erk5 directly phosphorylates Smad-specific E3 ubiquitin protein ligase 2 (Smurf2) at Thr249 (Smurf2Thr249) to activate its E3 ubiquitin ligase activity. Although we have clarified the importance of Erk5 in embryonic mesenchymal stem cells (MSCs) on skeletogenesis, its role in adult bone marrow (BM)-MSCs on bone homeostasis remains unknown. Leptin receptor-positive (LepR+) BM-MSCs represent a major source of bone in adult bone marrow and are critical regulators of postnatal bone homeostasis. Here, we identified Erk5 in BM-MSCs as an important regulator of bone homeostasis in adulthood. Bone marrow tissue was progressively osteosclerotic in mice lacking Erk5 in LepR+ BM-MSCs with age, accompanied by increased bone formation and normal bone resorption in vivo. Erk5 deficiency increased the osteogenic differentiation of BM-MSCs along with a higher expression of Runx2 and Osterix, essential transcription factors for osteogenic differentiation, without affecting their stemness in vitro. Erk5 deficiency decreased Smurf2Thr249 phosphorylation and subsequently increased Smad1/5/8-dependent signaling in BM-MSCs. The genetic introduction of the Smurf2T249E mutant (a phosphomimetic mutant) suppressed the osteosclerotic phenotype in Erk5-deficient mice. These findings suggest that the Erk5-Smurf2Thr249 axis in BM-MSCs plays a critical role in the maintenance of proper bone homeostasis by preventing excessive osteogenesis in adult bone marrow.


Asunto(s)
Células Madre Mesenquimatosas , Osteogénesis , Animales , Células de la Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Homeostasis , Células Madre Mesenquimatosas/metabolismo , Ratones , Proteína Quinasa 7 Activada por Mitógenos/genética , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Osteogénesis/genética
6.
Commun Biol ; 5(1): 22, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35017630

RESUMEN

Glioma stem cells (GSCs) contribute to the pathogenesis of glioblastoma, the most malignant form of glioma. The implication and underlying mechanisms of SMAD specific E3 ubiquitin protein ligase 2 (SMURF2) on the GSC phenotypes remain unknown. We previously demonstrated that SMURF2 phosphorylation at Thr249 (SMURF2Thr249) activates its E3 ubiquitin ligase activity. Here, we demonstrate that SMURF2Thr249 phosphorylation plays an essential role in maintaining GSC stemness and tumorigenicity. SMURF2 silencing augmented the self-renewal potential and tumorigenicity of patient-derived GSCs. The SMURF2Thr249 phosphorylation level was low in human glioblastoma pathology specimens. Introduction of the SMURF2T249A mutant resulted in increased stemness and tumorigenicity of GSCs, recapitulating the SMURF2 silencing. Moreover, the inactivation of SMURF2Thr249 phosphorylation increases TGF-ß receptor (TGFBR) protein stability. Indeed, TGFBR1 knockdown markedly counteracted the GSC phenotypes by SMURF2T249A mutant. These findings highlight the importance of SMURF2Thr249 phosphorylation in maintaining GSC phenotypes, thereby demonstrating a potential target for GSC-directed therapy.


Asunto(s)
Glioblastoma , Receptores de Factores de Crecimiento Transformadores beta/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Femenino , Glioblastoma/genética , Glioblastoma/patología , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Mutación/genética , Fosforilación/genética
7.
Nihon Yakurigaku Zasshi ; 156(5): 271-274, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34470930

RESUMEN

Vertebral bone and limb bone are formed by endochondral ossification, which is replaced with bone tissue by osteoblasts after cartilage formation. Bone growth is regulated by the balance between epiphyseal chondrocyte proliferation and ossification. We attempted to elucidate the mechanism of chondrocyte differentiation and maturation regulated by the Extracellular-signal-regulated kinase 5 (Erk5) signal. Erk5 is a serine/threonine kinase belonging to the mitogen-activated protein kinase (MAPK) family, which includes Erk1/2, JNK, and p38. Mesenchymal stem cell-specific Erk5-deficient mice exhibited the phenotype of deformities of the metatarsal bones, enlargement of the long bones in limbs, and overgrowth of cartilage tissue. Based on this result, we searched for factors that directly phosphorylate Erk5, and We demonstrated that Erk5 directly phosphorylates and activates Smurf2 (a ubiquitin E3 ligase) at Thr249 to activate its function and promotes ubiquitination-mediated degradation. The TGF-ß-Smad signal suppresses the proliferation of many cells and regulates the production of extracellular matrix. Our findings may lead to the development of novel drugs targeting TGF-ß associated diseases. In this paper, we investigated the function of Smurf2Thr249 phosphorylation and the possibility as new therapeutic target for various diseases.


Asunto(s)
Proteína Quinasa 7 Activada por Mitógenos , Factor de Crecimiento Transformador beta , Ubiquitina-Proteína Ligasas , Animales , Diferenciación Celular , Ratones , Proteína Quinasa 7 Activada por Mitógenos/genética , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Fosforilación , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
8.
Oncogene ; 40(15): 2803-2815, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33727660

RESUMEN

Glioblastoma (GBM) is the most malignant form of glioma. Glioma stem cells (GSCs) contribute to the initiation, progression, and recurrence of GBM as a result of their self-renewal potential and tumorigenicity. Cyclin-dependent kinase 8 (CDK8) belongs to the transcription-related CDK family. Although CDK8 has been shown to be implicated in the malignancy of several types of cancer, its functional role and mechanism in gliomagenesis remain largely unknown. Here, we demonstrate how CDK8 plays an essential role in maintaining stemness and tumorigenicity in GSCs. The genetic inhibition of CDK8 by shRNA or CRISPR interference resulted in an abrogation of the self-renewal potential and tumorigenicity of patient-derived GSCs, which could be significantly rescued by the ectopic expression of c-MYC, a stem cell transcription factor. Moreover, we demonstrated that the pharmacological inhibition of CDK8 significantly attenuated the self-renewal potential and tumorigenicity of GSCs. CDK8 expression was significantly higher in human GBM tissues than in normal brain tissues, and its expression was positively correlated with stem cell markers including c-MYC and SOX2 in human GBM specimens. Additionally, CDK8 expression is associated with poor survival in GBM patients. Collectively, these findings highlight the importance of the CDK8-c-MYC axis in maintaining stemness and tumorigenicity in GSCs; these findings also identify the CDK8-c-MYC axis as a potential target for GSC-directed therapy.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Quinasa 8 Dependiente de Ciclina/metabolismo , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Quinasa 8 Dependiente de Ciclina/genética , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal
9.
J Pharmacol Sci ; 145(1): 1-5, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33357767

RESUMEN

Bone remodeling is sophisticatedly regulated by two different cell types: bone-resorbing osteoclasts and bone-forming osteoblasts. Hochu-Ekki-To, a Japanese traditional herbal medicine, is commonly used for the treatment of chronic diseases or frailty after an illness; however, its effects on metabolic bone diseases such as osteoporosis are not well known. We herein report that daily oral Hochu-Ekki-To administration significantly inhibits osteoclast activation as well as the reduction in bone volume in ovariectomized mice. Our results suggest that supplementation with Hochu-Ekki-To might be beneficial for the prophylaxis and treatment of metabolic bone diseases associated with abnormal osteoclast activation.


Asunto(s)
Conservadores de la Densidad Ósea , Resorción Ósea/etiología , Resorción Ósea/prevención & control , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/farmacología , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Osteoporosis Posmenopáusica/etiología , Osteoporosis Posmenopáusica/prevención & control , Ovariectomía/efectos adversos , Fitoterapia , Administración Oral , Animales , Femenino , Humanos , Ratones Endogámicos
10.
Biol Pharm Bull ; 43(12): 1983-1986, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33268720

RESUMEN

The mechanistic/mammalian target of rapamycin complex-1 (mTORC1) integrates multiple signaling pathways and regulates various cellular processes. Tuberous sclerosis complex 1 (Tsc1) and complex 2 (Tsc2) are critical negative regulators of mTORC1. Mouse genetic studies, including ours, have revealed that inactivation of mTORC1 in undifferentiated mesenchymal cells and chondrocytes leads to severe skeletal abnormalities, indicating a pivotal role for mTORC1 in skeletogenesis. Here, we show that hyperactivation of mTORC1 influences skeletal development through its expression in undifferentiated mesenchymal cells at the embryonic stage. Inactivation of Tsc1 in undifferentiated mesenchymal cells by paired-related homeobox 1 (Prx1)-Cre-mediated recombination led to skeletal abnormalities in appendicular skeletons. In contrast, Tsc1 deletion in chondrocytes using collagen type II α1 (Col2a1)-Cre or in osteoprogenitors using Osterix (Osx)-Cre did not result in skeletal defects in either appendicular or axial skeletons. These findings indicate that Tsc complex-mediated chronic overactivation of mTORC1 influences skeletal development at the embryonic stage through its expression in undifferentiated mesenchymal cells but not in chondrocytes or osteoprogenitors.


Asunto(s)
Desarrollo Óseo/fisiología , Condrocitos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/deficiencia , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética
11.
Biochem Biophys Res Commun ; 533(1): 30-35, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-32917361

RESUMEN

The mechanistic/mammalian target of rapamycin (mTOR) regulates various cellular processes, in part through incorporation into distinct protein complexes. The mTOR complex 1 (mTORC1) contains the Raptor subunit, while mTORC2 specifically contains the Rictor subunit. Mouse genetic studies, including ours, have revealed a critical role for mTOR in skeletogenesis through its expression in undifferentiated mesenchymal cells. In addition, we have recently revealed that mTORC1 expression in chondrocytes is crucial for skeletogenesis. Recent work indicates that mTOR regulates cellular functions, depending on the context, through both complex-dependent (canonical pathway) and complex-independent roles (noncanonical pathway). Here, we determined that mTOR regulates skeletal development through the noncanonical pathway, as well as the canonical pathway, in a cell-type and context-specific manner. Inactivation of Mtor in undifferentiated mesenchymal cells or chondrocytes led to either severe hypoplasia in appendicular skeletons or a severe and generalized chondrodysplasia, respectively. Moreover, Rictor deletion in undifferentiated mesenchymal cells or chondrocytes led to mineralization defects in some skeletal components. Finally, we revealed that simultaneous deletion of Raptor and Rictor in undifferentiated mesenchymal cells recapitulated the appendicular skeletal phenotypes of Mtor deficiency, whereas chondrocyte-specific Raptor and Rictor double-mutants exhibited milder hypoplasia of appendicular and axial skeletons than those seen upon Mtor deletion. These findings indicate that mTOR regulates skeletal development mainly through the canonical pathway in undifferentiated mesenchymal cells, but at least in part through the noncanonical pathway in chondrocytes.


Asunto(s)
Condrocitos/citología , Células Madre Mesenquimatosas/citología , Esqueleto/embriología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Condrocitos/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Ratones , Transducción de Señal , Esqueleto/metabolismo , Serina-Treonina Quinasas TOR/genética
12.
Endocrinology ; 160(12): 2837-2848, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31555819

RESUMEN

Extracellular signal-regulated kinase 5 (Erk5), a member of the MAPK family, is specifically phosphorylated and activated by MAPK/Erk kinase-5. Although it has been implicated in odor discrimination and long-term memory via its expression in the central nervous system, little is known regarding the physiological importance of neuronal Erk5 in body weight and energy homeostasis. In the current study, systemic insulin injection significantly induced phosphorylation of Erk5 in the hypothalamus. Moreover, Erk5 deficiency in leptin receptor (LepR)‒expressing neurons led to an obesity phenotype, with increased white adipose tissue mass due to increased adipocyte size, only in female mice fed a normal chow diet. Furthermore, Erk5 deficiency in LepR-expressing neurons showed impaired glucose tolerance along with decreased physical activity, food intake, and energy expenditure. These results suggest that Erk5 controls body weight and systemic energy homeostasis probably via its expression in hypothalamic neurons in female mice, thereby providing a target for metabolic diseases such as obesity and type 2 diabetes mellitus.


Asunto(s)
Peso Corporal , Metabolismo Energético , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Neuronas/metabolismo , Receptores de Leptina/metabolismo , Tejido Adiposo Blanco , Animales , Glucemia , Ingestión de Alimentos , Femenino , Homeostasis , Hipotálamo/metabolismo , Insulina , Masculino , Ratones Endogámicos C57BL , Actividad Motora , Fosforilación
13.
Sci Signal ; 12(589)2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31289211

RESUMEN

L-type amino acid transporter 1 (LAT1), which is encoded by solute carrier transporter 7a5 (Slc7a5), plays a crucial role in amino acid sensing and signaling in specific cell types, contributing to the pathogenesis of cancer and neurological disorders. Amino acid substrates of LAT1 have a beneficial effect on bone health directly and indirectly, suggesting a potential role for LAT1 in bone homeostasis. Here, we identified LAT1 in osteoclasts as important for bone homeostasis. Slc7a5 expression was substantially reduced in osteoclasts in a mouse model of ovariectomy-induced osteoporosis. The osteoclast-specific deletion of Slc7a5 in mice led to osteoclast activation and bone loss in vivo, and Slc7a5 deficiency increased osteoclastogenesis in vitro. Loss of Slc7a5 impaired activation of the mechanistic target of rapamycin complex 1 (mTORC1) pathway in osteoclasts, whereas genetic activation of mTORC1 corrected the enhanced osteoclastogenesis and bone loss in Slc7a5-deficient mice. Last, Slc7a5 deficiency increased the expression of nuclear factor of activated T cells, cytoplasmic 1 (Nfatc1) and the nuclear accumulation of NFATc1, a master regulator of osteoclast function, possibly through the canonical nuclear factor κB pathway and the Akt-glycogen synthase kinase 3ß signaling axis, respectively. These findings suggest that the LAT1-mTORC1 axis plays a pivotal role in bone resorption and bone homeostasis by modulating NFATc1 in osteoclasts, thereby providing a molecular connection between amino acid intake and skeletal integrity.


Asunto(s)
Sistema de Transporte de Aminoácidos y+L/genética , Huesos/metabolismo , Homeostasis/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Osteoclastos/metabolismo , Osteogénesis/genética , Sistema de Transporte de Aminoácidos y+L/deficiencia , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Huesos/citología , Células Cultivadas , Femenino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Ovariectomía , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética
14.
Front Pharmacol ; 10: 684, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31263418

RESUMEN

The mechanistic/mammalian target of rapamycin (mTOR) is widely implicated in the pathogenesis of various diseases, including cancer, obesity, and cardiovascular disease. Bone homeostasis is maintained by the actions of bone-resorbing osteoclasts and bone-forming osteoblasts. An imbalance in the sophisticated regulation of osteoclasts and osteoblasts leads to the pathogenesis as well as etiology of certain metabolic bone diseases, including osteoporosis and osteopetrosis. Here, we identified mTOR complex 1 (mTORC1) as a pivotal mediator in the regulation of bone resorption and bone homeostasis under pathological conditions through its expression in osteoclasts. The activity of mTORC1, which was indicated by the phosphorylation level of its downstream target p70S6 kinase, was reduced during osteoclast differentiation, in accordance with the upregulation of Hamartin (encoded by tuberous sclerosis complex 1 [Tsc1]), a negative regulator of mTORC1. Receptor activator of nuclear factor-κB ligand (RANKL)-dependent osteoclastogenesis was impaired in Tsc1-deficient bone marrow macrophages. By contrast, osteoclastogenesis was markedly enhanced by Raptor deficiency but was unaffected by Rictor deficiency. The deletion of Tsc1 in osteoclast lineage cells in mice prevented bone resorption and bone loss in a RANKL-induced mouse model of osteoporosis, although neither bone volume nor osteoclastic parameter was markedly altered in these knockout mice under physiological conditions. Therefore, these findings suggest that mTORC1 is a key potential target for the treatment of bone diseases.

15.
FEBS Open Bio ; 9(2): 241-247, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30761250

RESUMEN

An imbalance of branched-chain amino acids (BCAAs) in the brain may result in neuropathological conditions, such as autism spectrum disorders. The L-type amino acid transporter 1 (LAT1), encoded by the solute carrier transporter 7a5 (Slc7a5) gene, is critical for maintaining normal levels of BCAAs in the brain. However, our understanding of the mechanisms that regulate the expression of LAT1/Slc7a5 in neurons is currently limited. Here, we demonstrate that hypoxic conditions result in upregulated expression of Slc7a5 in differentiated neuronal cells (Neuro2A cells induced to differentiate using all-trans retinoic acid). Mechanistically, hypoxia-induced expression of Slc7a5 is markedly reduced by short hairpin RNA (shRNA)-mediated knockdown of hypoxia-inducible factor 2α (HIF-2α), but not by shRNA targeting HIF-1α, in differentiated neuronal cells. Moreover, hypoxia increased the binding of HIF-2α to the proximal promoter of Slc7a5 in differentiated neuronal cells. These results indicate that hypoxia directly enhances the recruitment of HIF-2α to the proximal promoter of Slc7a5, resulting in its upregulated expression in differentiated neuronal cells. These findings indicate that Slc7a5 may be a novel gene responsive to hypoxia in a HIF-2α-dependent manner in differentiated neuronal cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Hipoxia de la Célula/genética , Transportador de Aminoácidos Neutros Grandes 1/genética , Neuronas/citología , Neuronas/metabolismo , Animales , Perfilación de la Expresión Génica , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Ratones , Células Tumorales Cultivadas
16.
Yakugaku Zasshi ; 139(1): 15-18, 2019.
Artículo en Japonés | MEDLINE | ID: mdl-30606922

RESUMEN

Although the transcriptional modulator interferon-related developmental regulator 1 (Ifrd1) has been identified as a transcriptional coactivator/repressor in various cells, including bone-resorbing osteoclasts, no attention has been paid to its role in bone-forming osteoblasts. Therefore, in this study we show that Ifrd1 is a critical mediator of both osteoblastogenesis and osteoclastogenesis through its expression in osteoblasts. Ifrd1 deficiency enhanced both osteoblast differentiation and maturation, and increased the expression of Runt-related transcription factor 2 and Osterix. A coculture experiment revealed that Ifrd1 deficient osteoblasts have higher osteoprotegerin (OPG) expression and less ability to support osteoclastogenesis. These findings suggest that Ifrd1 plays a pivotal role in bone homeostasis through its expression in osteoblasts, and represents a therapeutic target for bone disease.


Asunto(s)
Huesos/metabolismo , Diferenciación Celular/genética , Homeostasis/genética , Proteínas Inmediatas-Precoces/fisiología , Proteínas de la Membrana/fisiología , Osteoblastos/citología , Osteogénesis/genética , Animales , Enfermedades Óseas/genética , Enfermedades Óseas/terapia , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Expresión Génica , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Terapia Molecular Dirigida , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Factor de Transcripción Sp7/genética , Factor de Transcripción Sp7/metabolismo
17.
Bio Protoc ; 9(1): e3133, 2019 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33654761

RESUMEN

Mesenchymal stem cells have the ability to differentiate into multiple lineages, including adipocytes, osteoblasts and chondrocytes. Mesenchymal stem cells can be induced to differentiate into chondrocytes in extracellular matrices, such as alginate or collagen gel. Mesenchymal stem cells in a cell pellet or micromass culture can be also induced to form cartilages in a defined medium containing chondrogenic cytokines, such as transforming growth factor-ß (TGF-ß). Here, we describe a simple method to form cartilage by seeding mesenchymal cells derived from limb-bud cells at high cell density. First, we dissected the limb buds from embryonic mice (embryonic day 12.5) and digested them with enzymes (dispase and collagenase). After filtration using a cell strainer, we seeded the cells at high density. Unlike other methods, the method described here is simple and does not require the use of specialized equipment, expensive materials or complex reagents.

18.
Development ; 145(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-29986870

RESUMEN

Erk5 belongs to the mitogen-activated protein kinase (MAPK) family. Following its phosphorylation by Mek5, Erk5 modulates several signaling pathways in a number of cell types. In this study, we demonstrated that Erk5 inactivation in mesenchymal cells causes abnormalities in skeletal development by inducing Sox9, an important transcription factor of skeletogenesis. We further demonstrate that Erk5 directly phosphorylates and activates Smurf2 (a ubiquitin E3 ligase) at Thr249, which promotes the proteasomal degradation of Smad proteins and phosphorylates Smad1 at Ser206 in the linker region known to trigger its proteasomal degradation by Smurf1. Smads transcriptionally activated the expression of Sox9 in mesenchymal cells. Accordingly, removal of one Sox9 allele in mesenchymal cells from Erk5-deficient mice rescued some abnormalities of skeletogenesis. These findings highlight the importance of the Mek5-Erk5-Smurf-Smad-Sox9 axis in mammalian skeletogenesis.


Asunto(s)
Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Osteogénesis , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular , Condrogénesis , Humanos , Mesodermo/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteolisis , Cráneo/anomalías , Ubiquitina/metabolismo , Ubiquitinación
19.
Stem Cell Reports ; 11(1): 228-241, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-30008325

RESUMEN

The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) regulates cellular function in various cell types. Although the role of mTORC1 in skeletogenesis has been investigated previously, here we show a critical role of mTORC1/4E-BPs/SOX9 axis in regulating skeletogenesis through its expression in undifferentiated mesenchymal cells. Inactivation of Raptor, a component of mTORC1, in limb buds before mesenchymal condensations resulted in a marked loss of both cartilage and bone. Mechanistically, we demonstrated that mTORC1 selectively controls the RNA translation of Sox9, which harbors a 5' terminal oligopyrimidine tract motif, via inhibition of the 4E-BPs. Indeed, introduction of Sox9 or a knockdown of 4E-BP1/2 in undifferentiated mesenchymal cells markedly rescued the deficiency of the condensation observed in Raptor-deficient mice. Furthermore, introduction of the Sox9 transgene rescued phenotypes of deficient skeletal growth in Raptor-deficient mice. These findings highlight a critical role of mTORC1 in mammalian skeletogenesis, at least in part, through translational control of Sox9 RNA.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Osteogénesis/genética , Biosíntesis de Proteínas , Factor de Transcripción SOX9/genética , Esqueleto/metabolismo , Animales , Diferenciación Celular/genética , Expresión Génica , Ratones , Ratones Transgénicos , Fenotipo , Factor de Transcripción SOX9/metabolismo , Esqueleto/embriología
20.
Pharmacology ; 101(1-2): 64-71, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29065407

RESUMEN

The availability of amino acid in the brown adipose tissue (BAT) has been shown to be altered under various conditions; however, little is known about the possible expression and pivotal role of amino acid transporters in BAT under physiological and pathological conditions. The present study comprehensively investigated whether amino acid transporters are regulated by obesogenic conditions in BAT in vivo. Moreover, we investigated the mechanism underlying the regulation of the expression of amino acid transporters by various stressors in brown adipocytes in vitro. The expression of solute carrier family 38 member 1 (Slc38a1; gene encoding sodium-coupled neutral amino acid transporter 1) was preferentially upregulated in the BAT of both genetic and acquired obesity mice in vivo. Moreover, the expression of Slc38a1 was induced by hypoxic stress through hypoxia-inducible factor-1α, which is a master transcription factor of the adaptive response to hypoxic stress, in brown adipocytes in vitro. These results indicate that Slc38a1 is an obesity-associated gene in BAT and a hypoxia-responsive gene in brown adipocytes.


Asunto(s)
Adipocitos Marrones/metabolismo , Sistema de Transporte de Aminoácidos A/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hipoxia/genética , Obesidad/genética , Animales , Línea Celular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...