Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 8(24)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-37917194

RESUMEN

Osteoclasts specialize in bone resorption and are critical for bone remodeling. Previous studies have shown that osteoclasts possess abundant mitochondria and derive most energy through oxidative phosphorylation (OXPHOS). However, the energy substrates fueling OXPHOS in osteoclasts remain to be fully defined. Here, we showed that osteoclast differentiation was coupled with increased oxidation of glucose, glutamine, and oleate. Transcriptomic analyses with RNA sequencing revealed marked upregulation of genes participating in OXPHOS and mitochondrial fatty acid oxidation, during osteoclast differentiation. Increased mitochondrial oxidation of long-chain fatty acids was required for osteoclast differentiation in vitro. However, blocking fatty acid oxidation in vivo, by deletion of carnitine palmitoyltransferase 1a (Cpt1a) in osteoclast progenitors, impaired osteoclast formation only in the female mice. The Cpt1a-deficient females were further protected from osteoclast activation by a high-fat diet. The males, on the contrary, exhibited normal bone resorption despite Cpt1a deletion, regardless of the dietary fat content. Moreover, concurrent deletion of mitochondrial pyruvate carrier 1 and Cpt1a, blocking mitochondrial oxidation of both glucose and fatty acids in the osteoclast lineage, failed to impede bone resorption in the males. The study therefore uncovers a female-specific dependence on mitochondrial oxidation of fatty acids and glucose in osteoclasts in vivo.


Asunto(s)
Resorción Ósea , Osteoclastos , Masculino , Ratones , Femenino , Animales , Osteoclastos/metabolismo , Caracteres Sexuales , Resorción Ósea/metabolismo , Mitocondrias , Ácidos Grasos/metabolismo , Glucosa/metabolismo
2.
Cell Chem Biol ; 30(9): 1053-1063.e5, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37562406

RESUMEN

Type I diabetes (T1D) impairs bone accrual in patients, but the mechanism is unclear. Here in a murine monogenic model for T1D, we demonstrate that diabetes suppresses bone formation resulting in a rapid loss of both cortical and trabecular bone. Single-cell RNA sequencing uncovers metabolic dysregulation in bone marrow osteogenic cells of diabetic mice. In vivo stable isotope tracing reveals impaired glycolysis in diabetic bone that is highly responsive to insulin stimulation. Remarkably, deletion of the insulin receptor reduces cortical but not trabecular bone. Increasing glucose uptake by overexpressing Glut1 in osteoblasts exacerbates bone defects in T1D mice. Conversely, activation of glycolysis by Pfkfb3 overexpression preserves both trabecular and cortical bone mass in the face of diabetes. The study identifies defective glucose metabolism in osteoblasts as a pathogenic mechanism for osteopenia in T1D, and furthermore implicates boosting osteoblast glycolysis as a potential bone anabolic therapy.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Humanos , Ratones , Animales , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Osteoblastos/metabolismo , Densidad Ósea , Glucólisis
3.
Elife ; 122023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-37144869

RESUMEN

Skeletal fragility is associated with type 2 diabetes mellitus (T2D), but the underlying mechanism is not well understood. Here, in a mouse model for youth-onset T2D, we show that both trabecular and cortical bone mass is reduced due to diminished osteoblast activity. Stable isotope tracing in vivo with 13C-glucose demonstrates that both glycolysis and glucose fueling of the TCA cycle are impaired in diabetic bones. Similarly, Seahorse assays show suppression of both glycolysis and oxidative phosphorylation by diabetes in bone marrow mesenchymal cells as a whole, whereas single-cell RNA sequencing reveals distinct modes of metabolic dysregulation among the subpopulations. Metformin not only promotes glycolysis and osteoblast differentiation in vitro, but also improves bone mass in diabetic mice. Finally, osteoblast-specific overexpression of either Hif1a, a general inducer of glycolysis, or Pfkfb3 which stimulates a specific step in glycolysis, averts bone loss in T2D mice. The study identifies osteoblast-intrinsic defects in glucose metabolism as an underlying cause of diabetic osteopenia, which may be targeted therapeutically.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Ratones , Masculino , Animales , Osteogénesis , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Experimental/metabolismo , Osteoblastos/metabolismo , Glucosa/metabolismo
4.
bioRxiv ; 2023 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-36711657

RESUMEN

Skeletal fragility is associated with type 2 diabetes mellitus (T2D), but the underlying mechanism is not well understood. Here, in a mouse model for youth-onset T2D, we show that both trabecular and cortical bone mass are reduced due to diminished osteoblast activity. Stable isotope tracing in vivo with 13 C-glucose demonstrates that both glycolysis and glucose fueling of the TCA cycle are impaired in diabetic bones. Similarly, Seahorse assays show suppression of both glycolysis and oxidative phosphorylation by diabetes in bone marrow mesenchymal cells as a whole, whereas single-cell RNA sequencing reveals distinct modes of metabolic dysregulation among the subpopulations. Metformin not only promotes glycolysis and osteoblast differentiation in vitro, but also improves bone mass in diabetic mice. Finally, targeted overexpression of Hif1a or Pfkfb3 in osteoblasts of T2D mice averts bone loss. The study identifies osteoblast-intrinsic defects in glucose metabolism as an underlying cause of diabetic osteopenia, which may be targeted therapeutically.

5.
Cell Death Discov ; 8(1): 204, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35428362

RESUMEN

Iroquois homeobox transcription factor 5 (IRX5) plays a pivotal role in extramedullary adipogenesis, but little is known about the effects of IRX5 on adipogenesis of human bone marrow-derived mesenchymal stem cells (hMSCs). In this study, we aimed to determine the effect of IRX5 on hMSCs adipogenesis. By means of qPCR analysis, we determined that IRX5 expression was elevated during adipogenic commitment of hMSCs. The biologic role of IRX5 was further investigated by employing a gain/loss-of-function strategy using an in vitro lentivirus-based system. IRX5 overexpression promoted adipogenesis whereas IRX5 knockdown reduced the adipogenic phenotype. RNA-seq and metabolomics revealed that IRX5 overexpression repressed glycolysis. Dual-luciferase assay results showed that IRX5 overexpression transcriptionally activates peroxisome proliferator-activated receptor gamma coactivator (PGC-1α). Metformin and PGC-1α inhibitor reversed IRX5-induced adipogenesis and glycolytic inhibition. Collectively, IRX5 facilitates adipogenic differentiation of hMSCs by transcriptionally regulating PGC-1α and inhibiting glycolysis, revealing a potential target to control bone marrow-derived mesenchymal stem cells (BMSCs) fate decision and bone homeostasis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA