Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 214
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 164(6): 1248-1256, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26967290

RESUMEN

Most physiological functions originate with the communication between organs. Mouse genetics has revived this holistic view of physiology through the identification of inter-organ communications that are unanticipated, functionally important, and would have been difficult to uncover otherwise. This Review highlights this point by showing how two tissues usually not seen as endocrine ones, bone and striated muscles, influence several physiological processes in a significant manner.


Asunto(s)
Huesos/fisiología , Músculo Esquelético/fisiología , Animales , Citocinas/metabolismo , Metabolismo Energético , Humanos , Ratones , Osteocalcina , Fosfatos/metabolismo
2.
Cell ; 165(4): 882-95, 2016 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-27133169

RESUMEN

High-fat diet (HFD) feeding induces rapid reprogramming of systemic metabolism. Here, we demonstrate that HFD feeding of mice downregulates glucose transporter (GLUT)-1 expression in blood-brain barrier (BBB) vascular endothelial cells (BECs) and reduces brain glucose uptake. Upon prolonged HFD feeding, GLUT1 expression is restored, which is paralleled by increased expression of vascular endothelial growth factor (VEGF) in macrophages at the BBB. In turn, inducible reduction of GLUT1 expression specifically in BECs reduces brain glucose uptake and increases VEGF serum concentrations in lean mice. Conversely, myeloid-cell-specific deletion of VEGF in VEGF(Δmyel) mice impairs BBB-GLUT1 expression, brain glucose uptake, and memory formation in obese, but not in lean mice. Moreover, obese VEGF(Δmyel) mice exhibit exaggerated progression of cognitive decline and neuroinflammation on an Alzheimer's disease background. These experiments reveal that transient, HFD-elicited reduction of brain glucose uptake initiates a compensatory increase of VEGF production and assign obesity-associated macrophage activation a homeostatic role to restore cerebral glucose metabolism, preserve cognitive function, and limit neurodegeneration in obesity.


Asunto(s)
Encéfalo/metabolismo , Dieta Alta en Grasa , Glucosa/metabolismo , Obesidad/fisiopatología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Cognición , Células Endoteliales/metabolismo , Ácidos Grasos/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Ratones , Células Mieloides/metabolismo
3.
Cell ; 161(7): 1576-1591, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26091038

RESUMEN

The synthesis of type I collagen, the main component of bone matrix, precedes the expression of Runx2, the earliest determinant of osteoblast differentiation. We hypothesized that the energetic needs of osteoblasts might explain this apparent paradox. We show here that glucose, the main nutrient of osteoblasts, is transported in these cells through Glut1, whose expression precedes that of Runx2. Glucose uptake favors osteoblast differentiation by suppressing the AMPK-dependent proteasomal degradation of Runx2 and promotes bone formation by inhibiting another function of AMPK. While RUNX2 cannot induce osteoblast differentiation when glucose uptake is compromised, raising blood glucose levels restores collagen synthesis in Runx2-null osteoblasts and initiates bone formation in Runx2-deficient embryos. Moreover, RUNX2 favors Glut1 expression, and this feedforward regulation between RUNX2 and Glut1 determines the onset of osteoblast differentiation during development and the extent of bone formation throughout life. These results reveal an unexpected intricacy between bone and glucose metabolism.


Asunto(s)
Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Glucosa/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Secuencia de Aminoácidos , Animales , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Homeostasis , Ratones , Osteoblastos/citología , Alineación de Secuencia , Cráneo/citología
4.
Cell ; 160(1-2): 269-84, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25594183

RESUMEN

The stem cells that maintain and repair the postnatal skeleton remain undefined. One model suggests that perisinusoidal mesenchymal stem cells (MSCs) give rise to osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, although the existence of these cells has not been proven through fate-mapping experiments. We demonstrate here that expression of the bone morphogenetic protein (BMP) antagonist gremlin 1 defines a population of osteochondroreticular (OCR) stem cells in the bone marrow. OCR stem cells self-renew and generate osteoblasts, chondrocytes, and reticular marrow stromal cells, but not adipocytes. OCR stem cells are concentrated within the metaphysis of long bones not in the perisinusoidal space and are needed for bone development, bone remodeling, and fracture repair. Grem1 expression also identifies intestinal reticular stem cells (iRSCs) that are cells of origin for the periepithelial intestinal mesenchymal sheath. Grem1 expression identifies distinct connective tissue stem cells in both the bone (OCR stem cells) and the intestine (iRSCs).


Asunto(s)
Huesos/citología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Intestino Delgado/citología , Células Madre Mesenquimatosas/citología , Animales , Cartílago/metabolismo , Intestino Delgado/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
5.
Immunity ; 52(4): 606-619.e6, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32160524

RESUMEN

Group 2 innate lymphoid cells (ILC2s) regulate immunity, inflammation, and tissue homeostasis. Two distinct subsets of ILC2s have been described: steady-state natural ILC2s and inflammatory ILC2s, which are elicited following helminth infection. However, how tissue-specific cues regulate these two subsets of ILC2s and their effector functions remains elusive. Here, we report that interleukin-33 (IL-33) promotes the generation of inflammatory ILC2s (ILC2INFLAM) via induction of the enzyme tryptophan hydroxylase 1 (Tph1). Tph1 expression was upregulated in ILC2s upon activation with IL-33 or following helminth infection in an IL-33-dependent manner. Conditional deletion of Tph1 in lymphocytes resulted in selective impairment of ILC2INFLAM responses and increased susceptibility to helminth infection. Further, RNA sequencing analysis revealed altered gene expression in Tph1 deficient ILC2s including inducible T cell co-stimulator (Icos). Collectively, these data reveal a previously unrecognized function for IL-33, Tph1, and ICOS in promoting inflammatory ILC2 responses and type 2 immunity at mucosal barriers.


Asunto(s)
Inmunidad Celular , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Interleucina-33/inmunología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Subgrupos de Linfocitos T/inmunología , Triptófano Hidroxilasa/inmunología , Animales , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Inmunidad Mucosa , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucina-33/genética , Larva/crecimiento & desarrollo , Larva/inmunología , Larva/patogenicidad , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nippostrongylus/crecimiento & desarrollo , Nippostrongylus/patogenicidad , Cultivo Primario de Células , Transducción de Señal , Infecciones por Strongylida/genética , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología , Subgrupos de Linfocitos T/clasificación , Subgrupos de Linfocitos T/parasitología , Triptófano Hidroxilasa/genética
6.
Cell ; 155(1): 228-41, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24074871

RESUMEN

The powerful regulation of bone mass exerted by the brain suggests the existence of bone-derived signals modulating this regulation or other functions of the brain. We show here that the osteoblast-derived hormone osteocalcin crosses the blood-brain barrier, binds to neurons of the brainstem, midbrain, and hippocampus, enhances the synthesis of monoamine neurotransmitters, inhibits GABA synthesis, prevents anxiety and depression, and favors learning and memory independently of its metabolic functions. In addition to these postnatal functions, maternal osteocalcin crosses the placenta during pregnancy and prevents neuronal apoptosis before embryos synthesize this hormone. As a result, the severity of the neuroanatomical defects and learning and memory deficits of Osteocalcin(-/-) mice is determined by the maternal genotype, and delivering osteocalcin to pregnant Osteocalcin(-/-) mothers rescues these abnormalities in their Osteocalcin(-/-) progeny. This study reveals that the skeleton via osteocalcin influences cognition and contributes to the maternal influence on fetal brain development.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Osteocalcina/metabolismo , Transducción de Señal , Envejecimiento , Animales , Encéfalo/embriología , Encéfalo/fisiología , Femenino , Feto/metabolismo , Ratones , Neurotransmisores/metabolismo , Embarazo
7.
EMBO Rep ; 25(2): 593-615, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38228788

RESUMEN

Many physiological osteocalcin-regulated functions are affected in adult offspring of mothers experiencing unhealthy pregnancy. Furthermore, osteocalcin signaling during gestation influences cognition and adrenal steroidogenesis in adult mice. Together these observations suggest that osteocalcin may broadly function during pregnancy to determine organismal homeostasis in adult mammals. To test this hypothesis, we analyzed in unchallenged wildtype and Osteocalcin-deficient, newborn and adult mice of various genotypes and origin maintained on different genetic backgrounds, the functions of osteocalcin in the pancreas, liver and testes and their molecular underpinnings. This analysis revealed that providing mothers are Osteocalcin-deficient, Osteocalcin haploinsufficiency in embryos hampers insulin secretion, liver gluconeogenesis, glucose homeostasis, testes steroidogenesis in adult offspring; inhibits cell proliferation in developing pancreatic islets and testes; and disrupts distinct programs of gene expression in these organs and in the brain. This study indicates that osteocalcin exerts dominant functions in most organs it influences. Furthermore, through their synergistic regulation of multiple physiological functions, osteocalcin of maternal and embryonic origins contributes to the establishment and maintenance of organismal homeostasis in newborn and adult offspring.


Asunto(s)
Glucemia , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Humanos , Ratones , Embarazo , Glucemia/análisis , Glucemia/metabolismo , Homeostasis , Insulina/metabolismo , Secreción de Insulina , Mamíferos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo
8.
Cell ; 144(5): 796-809, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21333348

RESUMEN

Interactions between bone and the reproductive system have until now been thought to be limited to the regulation of bone remodeling by the gonads. We now show that, in males, bone acts as a regulator of fertility. Using coculture assays, we demonstrate that osteoblasts are able to induce testosterone production by the testes, though they fail to influence estrogen production by the ovaries. Analyses of cell-specific loss- and gain-of-function models reveal that the osteoblast-derived hormone osteocalcin performs this endocrine function. By binding to a G protein-coupled receptor expressed in the Leydig cells of the testes, osteocalcin regulates in a CREB-dependent manner the expression of enzymes that is required for testosterone synthesis, promoting germ cell survival. This study expands the physiological repertoire of osteocalcin and provides the first evidence that the skeleton is an endocrine regulator of reproduction.


Asunto(s)
Huesos/fisiología , Fertilidad , Osteocalcina/fisiología , Animales , Células Cultivadas , Humanos , Células Intersticiales del Testículo/fisiología , Masculino , Ratones , Osteoblastos/fisiología , Testículo/fisiología
10.
Cell ; 142(2): 296-308, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20655470

RESUMEN

The broad expression of the insulin receptor suggests that the spectrum of insulin function has not been fully described. A cell type expressing this receptor is the osteoblast, a bone-specific cell favoring glucose metabolism through a hormone, osteocalcin, that becomes active once uncarboxylated. We show here that insulin signaling in osteoblasts is necessary for whole-body glucose homeostasis because it increases osteocalcin activity. To achieve this function insulin signaling in osteoblasts takes advantage of the regulation of osteoclastic bone resorption exerted by osteoblasts. Indeed, since bone resorption occurs at a pH acidic enough to decarboxylate proteins, osteoclasts determine the carboxylation status and function of osteocalcin. Accordingly, increasing or decreasing insulin signaling in osteoblasts promotes or hampers glucose metabolism in a bone resorption-dependent manner in mice and humans. Hence, in a feed-forward loop, insulin signals in osteoblasts activate a hormone, osteocalcin, that promotes glucose metabolism.


Asunto(s)
Remodelación Ósea , Metabolismo Energético , Insulina/metabolismo , Osteoblastos/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Matriz Extracelular , Glucosa/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Osteocalcina/metabolismo
11.
Nature ; 568(7753): 541-545, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30971820

RESUMEN

Osteoclasts are multinucleated giant cells that resorb bone, ensuring development and continuous remodelling of the skeleton and the bone marrow haematopoietic niche. Defective osteoclast activity leads to osteopetrosis and bone marrow failure1-9, whereas excess activity can contribute to bone loss and osteoporosis10. Osteopetrosis can be partially treated by bone marrow transplantation in humans and mice11-18, consistent with a haematopoietic origin of osteoclasts13,16,19 and studies that suggest that they develop by fusion of monocytic precursors derived from haematopoietic stem cells in the presence of CSF1 and RANK ligand1,20. However, the developmental origin and lifespan of osteoclasts, and the mechanisms that ensure maintenance of osteoclast function throughout life in vivo remain largely unexplored. Here we report that osteoclasts that colonize fetal ossification centres originate from embryonic erythro-myeloid progenitors21,22. These erythro-myeloid progenitor-derived osteoclasts are required for normal bone development and tooth eruption. Yet, timely transfusion of haematopoietic-stem-cell-derived monocytic cells in newborn mice is sufficient to rescue bone development in early-onset autosomal recessive osteopetrosis. We also found that the postnatal maintenance of osteoclasts, bone mass and the bone marrow cavity involve iterative fusion of circulating blood monocytic cells with long-lived osteoclast syncytia. As a consequence, parabiosis or transfusion of monocytic cells results in long-term gene transfer in osteoclasts in the absence of haematopoietic-stem-cell chimerism, and can rescue an adult-onset osteopetrotic phenotype caused by cathepsin K deficiency23,24. In sum, our results identify the developmental origin of osteoclasts and a mechanism that controls their maintenance in bones after birth. These data suggest strategies to rescue osteoclast deficiency in osteopetrosis and to modulate osteoclast activity in vivo.


Asunto(s)
Células Madre Hematopoyéticas/citología , Osteoclastos/citología , Osteoclastos/metabolismo , Osteopetrosis/genética , Animales , Animales Recién Nacidos , Desarrollo Óseo , Femenino , Genes Recesivos , Masculino , Ratones , Osteopetrosis/patología , Erupción Dental
12.
Annu Rev Nutr ; 43: 55-71, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37603430

RESUMEN

Together, loss- and gain-of-function experiments have identified the bone-derived secreted molecule osteocalcin as a hormone with a broad reach in rodents and primates. Following its binding to one of three receptors, osteocalcin exerts a profound influence on various aspects of energy metabolism as well as steroidogenesis, neurotransmitter biosynthesis and thereby male fertility, electrolyte homeostasis, cognition, the acute stress response, and exercise capacity. Although this review focuses mostly on the regulation of energy metabolism by osteocalcin, it also touches on its other functions. Lastly, it proposes what could be a common theme between the functions of osteocalcin and between these functions and the structural functions of bone.


Asunto(s)
Cognición , Trastornos de Estrés Traumático Agudo , Animales , Masculino , Transporte Biológico , Metabolismo Energético , Osteocalcina , Humanos
13.
Cell ; 138(5): 976-89, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19737523

RESUMEN

Leptin inhibition of bone mass accrual requires the integrity of specific hypothalamic neurons but not expression of its receptor on these neurons. The same is true for its regulation of appetite and energy expenditure. This suggests that leptin acts elsewhere in the brain to achieve these three functions. We show here that brainstem-derived serotonin (BDS) favors bone mass accrual following its binding to Htr2c receptors on ventromedial hypothalamic neurons and appetite via Htr1a and 2b receptors on arcuate neurons. Leptin inhibits these functions and increases energy expenditure because it reduces serotonin synthesis and firing of serotonergic neurons. Accordingly, while abrogating BDS synthesis corrects the bone, appetite and energy expenditure phenotypes caused by leptin deficiency, inactivation of the leptin receptor in serotonergic neurons recapitulates them fully. This study modifies the map of leptin signaling in the brain and identifies a molecular basis for the common regulation of bone and energy metabolisms. For a video summary of this article, see the PaperFlick file with the Supplemental Data available online.


Asunto(s)
Apetito , Densidad Ósea , Metabolismo Energético , Leptina/metabolismo , Serotonina/metabolismo , Tronco Encefálico/metabolismo , Hipotálamo/metabolismo , Receptores de Leptina/metabolismo , Transducción de Señal
14.
Cell ; 135(5): 825-37, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-19041748

RESUMEN

Loss- and gain-of-function mutations in the broadly expressed gene Lrp5 affect bone formation, causing osteoporosis and high bone mass, respectively. Although Lrp5 is viewed as a Wnt coreceptor, osteoblast-specific disruption of beta-Catenin does not affect bone formation. Instead, we show here that Lrp5 inhibits expression of Tph1, the rate-limiting biosynthetic enzyme for serotonin in enterochromaffin cells of the duodenum. Accordingly, decreasing serotonin blood levels normalizes bone formation and bone mass in Lrp5-deficient mice, and gut- but not osteoblast-specific Lrp5 inactivation decreases bone formation in a beta-Catenin-independent manner. Moreover, gut-specific activation of Lrp5, or inactivation of Tph1, increases bone mass and prevents ovariectomy-induced bone loss. Serotonin acts on osteoblasts through the Htr1b receptor and CREB to inhibit their proliferation. By identifying duodenum-derived serotonin as a hormone inhibiting bone formation in an Lrp5-dependent manner, this study broadens our understanding of bone remodeling and suggests potential therapies to increase bone mass.


Asunto(s)
Duodeno/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Osteogénesis , Serotonina/metabolismo , Animales , Proteína de Unión a CREB/metabolismo , Femenino , Proteínas Relacionadas con Receptor de LDL/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Receptor de Serotonina 5-HT1B/metabolismo , Triptófano Hidroxilasa/metabolismo
15.
Annu Rev Cell Dev Biol ; 25: 629-48, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19575648

RESUMEN

In the past few years, our molecular understanding of bone formation has continued to increase. This review aims to present a comprehensive view of the current state of knowledge in the field. Thus, it will cover our current knowledge of chondrogenesis and osteoblastogenesis. It will also cover the most salient aspects of osteoblast function.


Asunto(s)
Condrogénesis , Regulación del Desarrollo de la Expresión Génica , Osteogénesis , Animales , Humanos , Osteoblastos/metabolismo , Factores de Transcripción/metabolismo
16.
J Cell Physiol ; 236(6): 4614-4624, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33305372

RESUMEN

Supraphysiological levels of the osteoblast-enriched mineralization regulator ectonucleotide pyrophosphatase or phosphodiesterase-1 (NPP1) is associated with type 2 diabetes mellitus. We determined the impact of osteoblast-specific Enpp1 ablation on skeletal structure and metabolic phenotype in mice. Female, but not male, 6-week-old mice lacking osteoblast NPP1 expression (osteoblast-specific knockout [KO]) exhibited increased femoral bone volume or total volume (17.50% vs. 11.67%; p < .01), and reduced trabecular spacing (0.187 vs. 0.157 mm; p < .01) compared with floxed (control) mice. Furthermore, an enhanced ability of isolated osteoblasts from the osteoblast-specific KO to calcify their matrix in vitro compared to fl/fl osteoblasts was observed (p < .05). Male osteoblast-specific KO and fl/fl mice showed comparable glucose and insulin tolerance despite increased levels of insulin-sensitizing under-carboxylated osteocalcin (195% increase; p < .05). However, following high-fat-diet challenge, osteoblast-specific KO mice showed impaired glucose and insulin tolerance compared with fl/fl mice. These data highlight a crucial local role for osteoblast NPP1 in skeletal development and a secondary metabolic impact that predominantly maintains insulin sensitivity.


Asunto(s)
Huesos/enzimología , Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina , Osteoblastos/enzimología , Osteogénesis , Hidrolasas Diéster Fosfóricas/deficiencia , Pirofosfatasas/deficiencia , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Huesos/patología , Hueso Esponjoso/enzimología , Hueso Esponjoso/patología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fémur/enzimología , Fémur/patología , Insulina/sangre , Masculino , Ratones Noqueados , Osteoblastos/patología , Osteocalcina/sangre , Hidrolasas Diéster Fosfóricas/genética , Pirofosfatasas/genética , Factores Sexuales , Cráneo/enzimología , Cráneo/patología , Tibia/enzimología , Tibia/patología
17.
Proc Natl Acad Sci U S A ; 115(25): 6458-6463, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29866843

RESUMEN

The enteric nervous system (ENS) is crucial for essential gastrointestinal physiologic functions such as motility, fluid secretion, and blood flow. The gut is colonized by trillions of bacteria that regulate host production of several signaling molecules including serotonin (5-HT) and other hormones and neurotransmitters. Approximately 90% of 5-HT originates from the intestine, and activation of the 5-HT4 receptor in the ENS has been linked to adult neurogenesis and neuroprotection. Here, we tested the hypothesis that the gut microbiota could induce maturation of the adult ENS through release of 5-HT and activation of 5-HT4 receptors. Colonization of germ-free mice with a microbiota from conventionally raised mice modified the neuroanatomy of the ENS and increased intestinal transit rates, which was associated with neuronal and mucosal 5-HT production and the proliferation of enteric neuronal progenitors in the adult intestine. Pharmacological modulation of the 5-HT4 receptor, as well as depletion of endogenous 5-HT, identified a mechanistic link between the gut microbiota and maturation of the adult ENS through the release of 5-HT and activation of the 5-HT4 receptor. Taken together, these findings show that the microbiota modulates the anatomy of the adult ENS in a 5-HT-dependent fashion with concomitant changes in intestinal transit.


Asunto(s)
Sistema Nervioso Entérico/microbiología , Sistema Nervioso Entérico/fisiología , Microbioma Gastrointestinal/fisiología , Intestino Delgado/microbiología , Serotonina/metabolismo , Animales , Sistema Nervioso Entérico/metabolismo , Femenino , Motilidad Gastrointestinal/fisiología , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Microbiota/fisiología , Neurogénesis/fisiología , Neuronas/metabolismo , Neuronas/microbiología , Receptores de Serotonina 5-HT4/metabolismo
18.
BMC Biol ; 18(1): 149, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-33092598

RESUMEN

BACKGROUND: The classical functions of the skeleton encompass locomotion, protection and mineral homeostasis. However, cell-specific gene deletions in the mouse and human genetic studies have identified the skeleton as a key endocrine regulator of metabolism. The bone-specific phosphatase, Phosphatase, Orphan 1 (PHOSPHO1), which is indispensable for bone mineralisation, has been recently implicated in the regulation of energy metabolism in humans, but its role in systemic metabolism remains unclear. Here, we probe the mechanism underlying metabolic regulation by analysing Phospho1 mutant mice. RESULTS: Phospho1-/- mice exhibited improved basal glucose homeostasis and resisted high-fat-diet-induced weight gain and diabetes. The metabolic protection in Phospho1-/- mice was manifested in the absence of altered levels of osteocalcin. Osteoblasts isolated from Phospho1-/- mice were enriched for genes associated with energy metabolism and diabetes; Phospho1 both directly and indirectly interacted with genes associated with glucose transport and insulin receptor signalling. Canonical thermogenesis via brown adipose tissue did not underlie the metabolic protection observed in adult Phospho1-/- mice. However, the decreased serum choline levels in Phospho1-/- mice were normalised by feeding a 2% choline rich diet resulting in a normalisation in insulin sensitivity and fat mass. CONCLUSION: We show that mice lacking the bone mineralisation enzyme PHOSPHO1 exhibit improved basal glucose homeostasis and resist high-fat-diet-induced weight gain and diabetes. This study identifies PHOSPHO1 as a potential bone-derived therapeutic target for the treatment of obesity and diabetes.


Asunto(s)
Metabolismo Energético , Resistencia a la Insulina/genética , Obesidad/genética , Monoéster Fosfórico Hidrolasas/genética , Animales , Colina/metabolismo , Glucosa/metabolismo , Homeostasis , Masculino , Ratones , Monoéster Fosfórico Hidrolasas/metabolismo
19.
Genes Dev ; 27(8): 955-69, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23599343

RESUMEN

Bone resorption by osteoclasts requires a large number of lysosomes that release proteases in the resorption lacuna. Whether lysosomal biogenesis is a consequence of the action of transcriptional regulators of osteoclast differentiation or is under the control of a different and specific transcriptional pathway remains unknown. We show here, through cell-based assays and cell-specific gene deletion experiments in mice, that the osteoclast differentiation factor RANKL promotes lysosomal biogenesis once osteoclasts are differentiated through the selective activation of TFEB, a member of the MITF/TFE family of transcription factors. This occurs following PKCß phosphorylation of TFEB on three serine residues located in its last 15 amino acids. This post-translational modification stabilizes and increases the activity of this transcription factor. Supporting these biochemical observations, mice lacking in osteoclasts--either TFEB or PKCß--show decreased lysosomal gene expression and increased bone mass. Altogether, these results uncover a RANKL-dependent signaling pathway taking place in differentiated osteoclasts and culminating in the activation of TFEB to enhance lysosomal biogenesis-a necessary step for proper bone resorption.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Regulación de la Expresión Génica , Lisosomas/metabolismo , Osteoclastos/fisiología , Proteína Quinasa C/metabolismo , Ligando RANK/metabolismo , Transducción de Señal , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Diferenciación Celular , Línea Celular , Femenino , Eliminación de Gen , Ratones , Ratones Endogámicos C57BL , Osteoclastos/citología , Osteoclastos/metabolismo , Fosforilación , Proteína Quinasa C/genética , Proteína Quinasa C beta , Procesamiento Proteico-Postraduccional , Ligando RANK/genética
20.
Am J Physiol Endocrinol Metab ; 318(3): E381-E391, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31935114

RESUMEN

Osteocalcin (OCN) is a bone-derived hormone involved in the regulation of glucose metabolism. In serum, OCN exists in carboxylated and uncarboxylated forms (ucOCN), and studies in rodents suggest that ucOCN is the bioactive form of this hormone. Whether this is also the case in humans is unclear, because a reliable assay to measure ucOCN is not available. Here, we established and validated a new immunoassay (ELISA) measuring human ucOCN and used it to determine the level of bioactive OCN in two cohorts of overweight or obese subjects, with or without type 2 diabetes (T2D). The ELISA could specifically detect ucOCN concentrations ranging from 0.037 to 1.8 ng/mL. In a first cohort of overweight or obese postmenopausal women without diabetes (n = 132), ucOCN correlated negatively with fasting glucose (r = -0.18, P = 0.042) and insulin resistance assessed by the homeostatic model assessment of insulin resistance (r = -0.18, P = 0.038) and positively with insulin sensitivity assessed by a hyperinsulinemic-euglycemic clamp (r = 0.18, P = 0.043) or insulin sensitivity index derived from an oral glucose tolerance test (r = 0.26, P = 0.003). In a second cohort of subjects with severe obesity (n = 16), ucOCN was found to be lower in subjects with T2D compared with those without T2D (2.76 ± 0.38 versus 4.52 ± 0.06 ng/mL, P = 0.009) and to negatively correlate with fasting glucose (r = -0.50, P = 0.046) and glycated hemoglobin (r = -0.57, P = 0.021). Moreover, the subjects with ucOCN levels below 3 ng/mL had a reduced insulin secretion rate during a hyperglycemic clamp (P = 0.03). In conclusion, ucOCN measured with this novel and specific assay is inversely associated with insulin resistance and ß-cell dysfunction in humans.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Osteocalcina/análisis , Osteocalcina/metabolismo , Pruebas de Función Pancreática , Adolescente , Adulto , Anciano , Animales , Glucemia , Estudios de Cohortes , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Técnica de Clampeo de la Glucosa , Hemoglobina Glucada/análisis , Humanos , Inmunoensayo/métodos , Resistencia a la Insulina , Masculino , Ratones Endogámicos BALB C , Persona de Mediana Edad , Obesidad/metabolismo , Sobrepeso/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA