Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
J Cell Physiol ; 237(4): 2271-2287, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35141958

RESUMEN

The physiological functions and downstream effectors of the atypical mitogen-activated protein kinase extracellular signal-regulated kinase 3 (ERK3) remain to be characterized. We recently reported that mice expressing catalytically-inactive ERK3 (Mapk6KD/KD ) exhibit a reduced postnatal growth rate as compared to control mice. Here, we show that genetic inactivation of ERK3 impairs postnatal skeletal muscle growth and adult muscle regeneration after injury. Loss of MAPK-activated protein kinase 5 (MK5) phenocopies the muscle phenotypes of Mapk6KD/KD mice. At the cellular level, genetic or pharmacological inactivation of ERK3 or MK5 induces precocious differentiation of C2C12 or primary myoblasts, concomitant with MyoD activation. Reciprocally, ectopic expression of activated MK5 inhibits myogenic differentiation. Mechanistically, we show that MK5 directly phosphorylates FoxO3, promoting its degradation and reducing its association with MyoD. Depletion of FoxO3 rescues in part the premature differentiation of C2C12 myoblasts observed upon inactivation of ERK3 or MK5. Our findings reveal that ERK3 and its substrate MK5 act in a linear signaling pathway to control postnatal myogenic differentiation.


Asunto(s)
Proteína Forkhead Box O3/metabolismo , Transducción de Señal , Animales , Péptidos y Proteínas de Señalización Intracelular , Ratones , Proteína Quinasa 6 Activada por Mitógenos/metabolismo , Músculos , Proteínas Serina-Treonina Quinasas/metabolismo
4.
Immunol Cell Biol ; 100(5): 338-351, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35285071

RESUMEN

Pou2af1 encodes for OCA-B, a coactivator of OCT-1/2 transcription factors, which plays a key role in B-cell maturation. The function of OCA-B has also been studied in T cells, where T cells from Pou2af1-/- mice have impaired functions, such as cytokine production and T follicular helper (Tfh) differentiation. Arguably, some of these T-cell phenotypes may result from impaired T-B interactions, secondary to the well-documented B-cell defects in Pou2af1-/- mice. Yet, Pou2af1 is actively transcribed in activated T cells, suggesting a T-cell-intrinsic role. To isolate the T-cell-intrinsic impact of Pou2af1, we generated Pou2af1fl/fl mice with specific genetic disruption of Pou2af1 either in all hematopoietic cells or exclusively in T cells. While we confirm that Pou2af1 is expressed in activated T cells, we surprisingly find that T-cell cytokine production is not impaired in Pou2af1-deficient T cells. Moreover, Pou2af1-sufficient and Pou2af1-deficient T cells have comparable transcriptome profiles, arguing against a T-cell-intrinsic role for Pou2af1. In line with these observations, we demonstrate that Tfh maturation is influenced by T-cell-extrinsic deletion of Pou2af1, as observed both in competitive bone marrow chimeras and in Pou2af1fl/fl mice with specific deletion in B cells. Overall, this study provides strong evidence that Pou2af1 does not act as a transcriptional coactivator in T cells, and conclusively demonstrates that loss of OCA-B in B cells indirectly impacts Tfh differentiation, clarifying the role of OCA-B in the immune system.


Asunto(s)
Linfocitos T , Factores de Transcripción , Animales , Linfocitos B , Diferenciación Celular/genética , Citocinas , Centro Germinal , Ratones , Linfocitos T Colaboradores-Inductores , Transactivadores/genética
5.
Mol Genet Metab ; 133(2): 211-221, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33814269

RESUMEN

Previously we have shown that young mice with a dominant severe form of osteogenesis imperfecta (OI), caused by mutated collagen type I, exhibit an altered glucose/insulin metabolism and energy expenditure along with elevated levels of osteocalcin, a bone-derived hormone involved in the regulation of whole-body metabolism. This study aimed to examine the long-term effects of a western diet in these OI mice. Male and female OI mice and wild type littermates (WT) were fed a high-fat diet (HFD) or a matched low-fat diet (LFD) for 26 weeks. HFD-induced obesity was observed in male and female WT and female OI mice, but not in male OI mice. HFD-fed WT and OI mice of both sexes developed hyperglycemia and glucose intolerance, but the degree of glucose intolerance was significantly lower in male and female OI mice compared to sex- and diet-matched WT mice. Indirect calorimetry revealed increased movement of male OI mice on HFD compared to LFD and, while HFD lowered energy expenditure in WT mice, energy expenditure was not changed in OI mice. Further, HFD-fed male OI mice demonstrated a diet-induced increased expression of the thermogenesis genes, Ucp1 and Pgc1α, in brown adipose tissue. On LFD, total and Gla-13 osteocalcin levels were similar in 30-week-old WT and OI mice, but on HFD, both were significantly higher in OI mice than WT. Thus, male OI mice respond to HFD with increased movement, energy expenditure, brown adipose tissue thermogenesis, and higher levels of osteocalcin, resulting in partial protection against HFD-induced obesity.


Asunto(s)
Obesidad/metabolismo , Osteocalcina/genética , Osteogénesis Imperfecta/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Proteína Desacopladora 1/genética , Tejido Adiposo Pardo , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético/genética , Femenino , Regulación de la Expresión Génica/genética , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Masculino , Ratones , Obesidad/complicaciones , Obesidad/genética , Obesidad/patología , Osteogénesis Imperfecta/complicaciones , Osteogénesis Imperfecta/genética , Osteogénesis Imperfecta/patología , Caracteres Sexuales
6.
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
7.
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
8.
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
9.
J Biol Chem ; 292(27): 11400-11412, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28487368

RESUMEN

Genetic and environmental factors may lead to abnormal growth of the orofacial skeleton, affecting the overall structure of the face. In this study, we investigated the craniofacial abnormalities in a mouse model for Keutel syndrome, a rare genetic disease caused by loss-of-function mutations in the matrix Gla protein (MGP) gene. Keutel syndrome patients show diffuse ectopic calcification of cartilaginous tissues and impaired midface development. Our comparative cephalometric analyses of micro-computed tomography images revealed a severe midface hypoplasia in Mgp-/- mice. In vivo reporter studies demonstrated that the Mgp promoter is highly active at the cranial sutures, cranial base synchondroses, and nasal septum. Interestingly, the cranial sutures of the mutant mice showed normal anatomical features. Although we observed a mild increase in mineralization of the spheno-occipital synchondrosis, it did not reduce the relative length of the cranial base in comparison with total skull length. Contrary to this, we found the nasal septum to be abnormally mineralized and shortened in Mgp-/- mice. Transgenic restoration of Mgp expression in chondrocytes fully corrected the craniofacial anomalies caused by MGP deficiency, suggesting a local role for MGP in the developing nasal septum. Although there was no up-regulation of markers for hypertrophic chondrocytes, a TUNEL assay showed a marked increase in apoptotic chondrocytes in the calcified nasal septum. Transmission electron microscopy confirmed unusual mineral deposits in the septal extracellular matrix of the mutant mice. Of note, the systemic reduction of the inorganic phosphate level was sufficient to prevent abnormal mineralization of the nasal septum in Mgp-/-;Hyp compound mutants. Our work provides evidence that modulation of local and systemic factors regulating extracellular matrix mineralization can be possible therapeutic strategies to prevent ectopic cartilage calcification and some forms of congenital craniofacial anomalies in humans.


Asunto(s)
Calcinosis , Proteínas de Unión al Calcio/deficiencia , Condrocitos , Anomalías Craneofaciales , Proteínas de la Matriz Extracelular/deficiencia , Tabique Nasal , Animales , Calcinosis/embriología , Calcinosis/genética , Calcinosis/metabolismo , Calcinosis/patología , Condrocitos/metabolismo , Condrocitos/patología , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Anomalías Craneofaciales/patología , Humanos , Ratones , Ratones Noqueados , Tabique Nasal/embriología , Tabique Nasal/metabolismo , Tabique Nasal/patología , Proteína Gla de la Matriz
10.
Nature ; 481(7381): 314-20, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22258610

RESUMEN

The mouse genetic revolution has shown repeatedly that most organs have more functions than expected. This has led to the realization that, in addition to a molecular and cellular approach, there is a need for a whole-organism study of physiology. The skeleton is an example of how a whole-organism approach to physiology can broaden the functions of a given organ, reveal connections of this organ with others such as the brain, pancreas and gut, and shed new light on the pathogenesis of degenerative diseases affecting multiple organs.


Asunto(s)
Huesos/fisiología , Fenómenos Fisiológicos , Animales , Apetito/fisiología , Enfermedades Óseas/metabolismo , Enfermedades Óseas/patología , Huesos/patología , Metabolismo Energético , Humanos , Leptina/metabolismo , Especificidad de Órganos , Osteocalcina/metabolismo
11.
EMBO J ; 31(5): 1095-108, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22343943

RESUMEN

The lysosome plays a key role in cellular homeostasis by controlling both cellular clearance and energy production to respond to environmental cues. However, the mechanisms mediating lysosomal adaptation are largely unknown. Here, we show that the Transcription Factor EB (TFEB), a master regulator of lysosomal biogenesis, colocalizes with master growth regulator mTOR complex 1 (mTORC1) on the lysosomal membrane. When nutrients are present, phosphorylation of TFEB by mTORC1 inhibits TFEB activity. Conversely, pharmacological inhibition of mTORC1, as well as starvation and lysosomal disruption, activates TFEB by promoting its nuclear translocation. In addition, the transcriptional response of lysosomal and autophagic genes to either lysosomal dysfunction or pharmacological inhibition of mTORC1 is suppressed in TFEB-/- cells. Interestingly, the Rag GTPase complex, which senses lysosomal amino acids and activates mTORC1, is both necessary and sufficient to regulate starvation- and stress-induced nuclear translocation of TFEB. These data indicate that the lysosome senses its content and regulates its own biogenesis by a lysosome-to-nucleus signalling mechanism that involves TFEB and mTOR.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Núcleo Celular/fisiología , Lisosomas/fisiología , Proteínas/metabolismo , Transducción de Señal , Animales , Línea Celular , Humanos , Inmunoprecipitación , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Microscopía Fluorescente , Modelos Biológicos , Complejos Multiproteicos , Unión Proteica , Mapeo de Interacción de Proteínas , Serina-Treonina Quinasas TOR
12.
Arch Biochem Biophys ; 561: 137-46, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24893146

RESUMEN

The skeleton has recently emerged as an endocrine organ implicated in the regulation of glucose and energy metabolism. This function of bone is mediated, at least in part, by osteocalcin, an osteoblast-derived protein acting as a hormone stimulating insulin sensitivity, insulin secretion and energy expenditure. Osteocalcin secretion and bioactivity is in turn regulated by several hormonal cues including insulin, leptin, the sympathetic nervous system and glucocorticoids. Recent findings support the notion that osteocalcin functions and regulations are conserved between mice and humans. Moreover, studies in mice suggest that osteocalcin could represent a viable therapeutic approach for the treatment of obesity and insulin resistance. In this review, we summarize the current knowledge on osteocalcin functions, its various modes of action and the mechanisms implicated in the control of this hormone.


Asunto(s)
Huesos/metabolismo , Metabolismo Energético/fisiología , Glucosa/metabolismo , Homeostasis/fisiología , Resistencia a la Insulina/fisiología , Insulina/sangre , Osteocalcina/metabolismo , Animales , Humanos , Ratones
13.
Artículo en Inglés | MEDLINE | ID: mdl-38429160

RESUMEN

Vitamin K is an essential micronutrient and a cofactor for the enzyme γ-glutamyl carboxylase, which adds a carboxyl group to specific glutamic acid residues in proteins transiting through the secretory pathway. Higher vitamin K intake has been linked to a reduced incidence of type 2 diabetes (T2D) in humans. Preclinical work suggests that this effect depends on the γ-carboxylation of specific proteins in ß-cells, including endoplasmic reticulum Gla protein (ERGP), implicated in the control of intracellular Ca2+ levels. In this review we discuss these recent advances linking vitamin K and glucose metabolism, and argue that identification of γ-carboxylated proteins in ß-cells is pivotal to better understand how vitamin K protects from T2D and to design targeted therapies for this disease.

14.
J Biol Chem ; 287(12): 8757-68, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22298775

RESUMEN

The Forkhead transcription factor FoxO1 inhibits through its expression in osteoblasts ß-cell proliferation, insulin secretion, and sensitivity. At least part of the FoxO1 metabolic functions result from its ability to suppress the activity of osteocalcin, an osteoblast-derived hormone favoring glucose metabolism and energy expenditure. In searching for mechanisms mediating the metabolic actions of FoxO1, we focused on ATF4, because this transcription factor also affects glucose metabolism through its expression in osteoblasts. We show here that FoxO1 co-localizes with ATF4 in the osteoblast nucleus, and physically interacts with and promotes the transcriptional activity of ATF4. Genetic experiments demonstrate that FoxO1 and ATF4 cooperate to increase glucose levels and decrease glucose tolerance. These effects result from a synergistic effect of the two transcription factors to suppress the activity of osteocalcin through up-regulating expression of the phosphatase catalyzing osteocalcin inactivation. As a result, insulin production by ß-cells and insulin signaling in the muscle, liver and white adipose tissue are compromised and fat weight increases by the FoxO1/ATF4 interaction. Taken together these observations demonstrate that FoxO1 and ATF4 cooperate in osteoblasts to regulate glucose homeostasis.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Factores de Transcripción Forkhead/metabolismo , Glucosa/metabolismo , Osteoblastos/metabolismo , Factor de Transcripción Activador 4/genética , Animales , Proliferación Celular , Células Cultivadas , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Homeostasis , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Osteoblastos/citología , Unión Proteica
15.
Am J Physiol Endocrinol Metab ; 304(12): E1321-30, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23612996

RESUMEN

Among women, the polycystic ovarian syndrome (PCOS) is considered a form of metabolic syndrome with reproductive abnormalities. Women with PCOS show increased sympathetic tone, visceral adiposity with enlarged adipocytes, hypoadiponectinemia, insulin resistance, glucose intolerance, increased inactive osteocalcin, and hypertension. Excess fetal exposure to androgens has been hypothesized to play a role in the pathogenesis of PCOS. Previously, we showed that neonatal exposure to the androgen testosterone (NT) programs leptin resistance in adult female mice. Here, we studied the impact of NT on lean and adipose tissues, sympathetic tone in cardiometabolic tissues, and the development of metabolic dysfunction in mice. Neonatally androgenized adult female mice (NTF) displayed masculinization of lean tissues with increased cardiac and skeletal muscle as well as kidney masses. NTF mice showed increased and dysfunctional white adipose tissue with increased sympathetic tone in both visceral and subcutaneous fat as well as increased number of enlarged and insulin-resistant adipocytes that displayed altered expression of developmental genes and hypoadiponectinemia. NTF exhibited dysfunctional brown adipose tissue with increased mass and decreased energy expenditure. They also displayed decreased undercarboxylated and active osteocalcin and were predisposed to obesity during chronic androgen excess. NTF showed increased renal sympathetic tone associated with increased blood pressure, and they developed glucose intolerance and insulin resistance. Thus, developmental exposure to testosterone in female mice programs features of cardiometabolic dysfunction, as can be observed in women with PCOS, including increased sympathetic tone, visceral adiposity, insulin resistance, prediabetes, and hypertension.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Hipertensión Renal/metabolismo , Síndrome Metabólico/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Sistema Nervioso Simpático/metabolismo , Testosterona/metabolismo , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/crecimiento & desarrollo , Andrógenos/metabolismo , Andrógenos/farmacología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Humanos , Resistencia a la Insulina/fisiología , Grasa Intraabdominal/metabolismo , Ratones , Ratones Endogámicos C57BL , Estado Prediabético/metabolismo , Sistema Nervioso Simpático/crecimiento & desarrollo , Testosterona/farmacología
16.
Appl Physiol Nutr Metab ; 48(4): 321-330, 2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-36680800

RESUMEN

Patients with cystic fibrosis (CF) are at high risk of fat-soluble vitamin deficiencies, even with supplementation. The contribution of a suboptimal vitamin K status to respiratory and endocrine pathophysiology in CF has been inadequately characterized. This is a cross-sectional study in adult CF patients (≥18 years old) from the Montreal Cystic Fibrosis Cohort. Vitamin K1 (VK1) was measured with high-performance liquid chromatography, using fasted serum samples collected during an oral glucose tolerance test (OGTT: 2 h with plasma glucose and insulin every 30 min) (n = 168). Patients were categorized according to VK1 status (suboptimal defined as <0.30 nmol/L). Suboptimal VK1 levels were observed in 66% of patients. Patients with a suboptimal VK1 status have a higher risk of colonization with Pseudomonas aeruginosa (p = 0.001), have lower body mass index (BMI) (p = 0.003), and were more likely to have exocrine pancreatic insufficiency (p = 0.002). Using an established threshold for VK1, we did show significantly reduced OGTT-derived measures of insulin secretion in patients with a VK1 status below 0.30 nmol/L (first- and second-phase area under the curve (AUC)INS/GLU (p = 0.002 and p = 0.006), AUCINS (p = 0.012) and AUCINS/GLU (p = 0.004)). Subclinical vitamin K deficiency is more common than other fat-soluble vitamin deficiencies in patients with CF. We demonstrate an association between a suboptimal VK1 status and measures of insulin secretion. We highlight the potential associations of mild vitamin K deficiency with pseudomonal colonization and lower BMI, although these need to be validated in prospective studies.


Asunto(s)
Avitaminosis , Fibrosis Quística , Deficiencia de Vitamina K , Adulto , Humanos , Avitaminosis/complicaciones , Índice de Masa Corporal , Estudios Transversales , Fibrosis Quística/complicaciones , Secreción de Insulina , Estudios Prospectivos , Vitamina K , Deficiencia de Vitamina K/complicaciones , Vitaminas
17.
Cell Rep ; 42(5): 112500, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37171959

RESUMEN

Vitamin K is a micronutrient necessary for γ-carboxylation of glutamic acids. This post-translational modification occurs in the endoplasmic reticulum (ER) and affects secreted proteins. Recent clinical studies implicate vitamin K in the pathophysiology of diabetes, but the underlying molecular mechanism remains unknown. Here, we show that mouse ß cells lacking γ-carboxylation fail to adapt their insulin secretion in the context of age-related insulin resistance or diet-induced ß cell stress. In human islets, γ-carboxylase expression positively correlates with improved insulin secretion in response to glucose. We identify endoplasmic reticulum Gla protein (ERGP) as a γ-carboxylated ER-resident Ca2+-binding protein expressed in ß cells. Mechanistically, γ-carboxylation of ERGP protects cells against Ca2+ overfilling by diminishing STIM1 and Orai1 interaction and restraining store-operated Ca2+ entry. These results reveal a critical role of vitamin K-dependent carboxylation in regulation of Ca2+ flux in ß cells and in their capacity to adapt to metabolic stress.


Asunto(s)
Procesamiento Proteico-Postraduccional , Vitamina K , Ratones , Animales , Humanos , Vitamina K/farmacología , Vitamina K/fisiología , Osteocalcina/metabolismo , Insulina/metabolismo , Estrés Fisiológico , Calcio/metabolismo
18.
Nat Med ; 9(4): 399-406, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12627228

RESUMEN

The spontaneous mouse grey-lethal (gl) mutation is responsible for a coat color defect and for the development of the most severe autosomal recessive form of osteopetrosis. Using a positional cloning approach, we have mapped and isolated the gl locus from a approximately 1.5 cM genetic interval. The gl locus was identified in a bacterial artificial chromosome (BAC) contig by functional genetic complementation in transgenic mice. Genomic sequence analysis revealed that the gl mutation is a deletion resulting in complete loss of function. The unique approximately 3 kb wild-type transcript is expressed primarily in osteoclasts and melanocytes as well as in brain, kidney, thymus and spleen. The gl gene is predicted to encode a 338-amino acid type I transmembrane protein that localizes to the intracellular compartment. Mutation in the human GL gene leads to severe recessive osteopetrosis. Our studies show that mouse Gl protein function is absolutely required for osteoclast and melanocyte maturation and function.


Asunto(s)
Genes Letales , Genes Recesivos , Proteínas de la Membrana/genética , Osteopetrosis/genética , Trastornos de la Pigmentación/genética , Secuencia de Aminoácidos , Animales , Cromosomas Artificiales Bacterianos , Eliminación de Gen , Humanos , Líquido Intracelular/metabolismo , Melanocitos/fisiología , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Osteoclastos/fisiología , Osteopetrosis/patología , Mapeo Físico de Cromosoma , Homología de Secuencia de Aminoácido
19.
Proc Natl Acad Sci U S A ; 105(13): 5266-70, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18362359

RESUMEN

The osteoblast-specific secreted molecule osteocalcin behaves as a hormone regulating glucose metabolism and fat mass in two mutant mouse strains. Here, we ask two questions: is the action of osteocalcin on beta cells and adipocytes elicited by the same concentrations of the molecule, and more importantly, does osteocalcin regulate energy metabolism in WT mice? Cell-based assays using isolated pancreatic islets, a beta cell line, and primary adipocytes showed that picomolar amounts of osteocalcin are sufficient to regulate the expression of the insulin genes and beta cell proliferation markers, whereas nanomolar amounts affect adiponectin and Pgc1alpha expression in white and brown adipocytes, respectively. In vivo the same difference exists in osteocalcin's ability to regulate glucose metabolism on the one hand and affect insulin sensitivity and fat mass on the other hand. Furthermore, we show that long-term treatment of WT mice with osteocalcin can significantly weaken the deleterious effect on body mass and glucose metabolism of gold thioglucose-induced hyperphagia and high-fat diet. These results establish in WT mice the importance of this novel molecular player in the regulation of glucose metabolism and fat mass and suggest that osteocalcin may be of value in the treatment of metabolic diseases.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Enfermedades Metabólicas/metabolismo , Osteocalcina/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus/metabolismo , Diabetes Mellitus/prevención & control , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/genética , Ratones , Ratones Endogámicos C57BL , Obesidad/prevención & control , Sensibilidad y Especificidad
20.
Front Endocrinol (Lausanne) ; 12: 690681, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34149625

RESUMEN

Fibroblast growth factor 23 (FGF23) is a hormone secreted from fully differentiated osteoblasts and osteocytes that inhibits phosphate reabsorption by kidney proximal tubules. The full-length (i.e., intact) protein mediates FGF23 endocrine functions, while endoproteolytic cleavage at a consensus cleavage sequence for the proprotein convertases (PCs) inactivates FGF23. Two PCs, furin and PC5, were shown to cleave FGF23 in vitro at RHTR179↓, but whether they are fulfilling this function in vivo is currently unknown. To address this question, we used here mice lacking either or both furin and PC5 in cell-specific manners and mice lacking the paired basic amino acid-cleaving enzyme 4 (PACE4) in all cells. Our analysis shows that furin inactivation in osteoblasts and osteocytes results in a 25% increase in circulating intact FGF23, without any significant impact on serum phosphate levels, whether mice are maintained on a normal or a low phosphate diet. Under conditions of iron deficiency, FGF23 is normally processed in control mice, but its processing is impaired in mice lacking furin in osteoblasts and osteocytes. In contrast, FGF23 is normally cleaved following erythropoietin or IL-1ß injections in mice lacking furin or both furin and PC5, and in PACE4-deficient mice. Altogether, these studies suggest that furin is only partially responsible for FGF23 cleavage under certain conditions in vivo. The processing of FGF23 may therefore involve the redundant action of multiple PCs or of other peptidases in osteoblasts, osteocytes and hematopoietic cells.


Asunto(s)
Factor-23 de Crecimiento de Fibroblastos/metabolismo , Furina/metabolismo , Osteoblastos/metabolismo , Osteocitos/metabolismo , Proproteína Convertasa 5/metabolismo , Animales , Médula Ósea/metabolismo , Factor-23 de Crecimiento de Fibroblastos/genética , Furina/genética , Deficiencias de Hierro/genética , Deficiencias de Hierro/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Proproteína Convertasa 5/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA