Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Int J Obes (Lond) ; 45(1): 235-246, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32848203

RESUMEN

OBJECTIVES: Little is known about the long-term skeletal impact of bariatric procedures, particularly the increasingly commonly performed gastric sleeve surgery (GS). We examined bone density (BMD) change following three types of bariatric surgery Roux-en-Y gastric bypass (RYGB), GS and laparoscopic adjustable gastric banding (LAGB), compared with diet, over 36 months. METHODS: Non-randomized, prospective study of participants with severe obesity (n = 52), undergoing weight-loss interventions: RYGB (n = 7), GS (n = 21), LAGB (n = 11) and diet (n = 13). Measurements of calciotropic indices, gut hormones (fasting and post prandial) peptide YY (PYY), glucagon-like peptide 1 (GLP1) and adiponectin together with dual-X-ray absorptiometry and quantitative computed tomography scans were performed thorough the study. RESULTS: All groups lost weight during the first 12 months. Despite weight stability from 12 to 36 months and supplementation of calcium and vitamin D, there was progressive bone loss at the total hip (TH) over 36 months in RYGB -14% (95% CI: -12, -17) and GS -9% (95% CI: -7, -10). In RYGB forearm BMD also declined over 36 months -9% (95% CI: -6, -12) and LS BMD declined over the first 12 months -7% (95% CI: -3, -12). RYGB and GS groups experienced significantly greater bone loss until 36 months than LAGB and diet groups, which experienced no significant BMD loss. These bone losses remained significant after adjustment for weight loss and age. RYGB and GS procedures resulted in elevated postprandial PYY, adiponectin and bone turnover markers up to 36 months without such changes among LAGB and diet participants. CONCLUSIONS: RYGB and GS but not LAGB resulted in ongoing TH bone loss for three postoperative years. For RYGB, bone loss was also observed at LS and non-weight-bearing forearms. These BMD changes were independent of weight and age differences. We, therefore, recommend close monitoring of bone health following RYGB and GS surgeries.


Asunto(s)
Densidad Ósea/fisiología , Derivación Gástrica , Obesidad Mórbida/cirugía , Pérdida de Peso/fisiología , Adulto , Femenino , Derivación Gástrica/efectos adversos , Derivación Gástrica/estadística & datos numéricos , Humanos , Persona de Mediana Edad , Estudios Prospectivos
2.
Int J Obes (Lond) ; 44(10): 2149-2164, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32152498

RESUMEN

BACKGROUND/OBJECTIVES: Maintaining energy balance is important to ensure a healthy organism. However, energy partitioning, coordinating the distribution of sufficient energy to different organs and tissues is equally important, but the control of this process is largely unknown. In obesity, an increase in fat mass necessitates the production of additional bone mass to cope with the increase in bodyweight and processes need to be in place to communicate this new weight bearing demand. Here, we investigate the interaction between leptin and NPY, two factors critically involved in the regulation of both energy metabolism and bone mass, in this process. METHODS: We assessed the co-localization of leptin receptors on NPY neurons using RNAScope followed by a systematic examination of body composition and energy metabolism profiling in male and female mice lacking leptin receptors specifically in NPY neurons (Leprlox/lox;NPYCre/+). The effect of short-term switching between chow and high-fat diet was also examined in these mice. RESULTS: We uncovered that leptin receptor expression is greater on a subpopulation of NPY neurons in the arcuate that do not express AgRP. We further show that Leprlox/lox;NPYCre/+ mice exhibit significantly increased adiposity while bone mass is diminished. These body composition changes occur in the absence of alterations in food intake or energy expenditure, demonstrating a prominent role for leptin signaling in NPY neurons in the control of energy partitioning. Importantly however, when fed a high-fat diet, these mice display a switch in energy partitioning whereby they exhibit a significantly enhanced ability to increase their bone mass to match the increased bodyweight caused by higher caloric intake concurrent with attenuated adiposity. CONCLUSIONS: Taken together, these results demonstrate that leptin signaling in NPY neurons is critical for coordinating energy partitioning between fat and bone mass especially during situations of changes in energy balance.


Asunto(s)
Tejido Adiposo/metabolismo , Huesos/metabolismo , Metabolismo Energético , Hipotálamo/metabolismo , Leptina/metabolismo , Neuronas/metabolismo , Adiposidad , Animales , Composición Corporal , Dieta Alta en Grasa , Ingestión de Energía , Femenino , Masculino , Ratones , Receptores de Leptina
3.
Curr Osteoporos Rep ; 14(1): 26-31, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26872458

RESUMEN

Neural pathways are now a well-appreciated factor in the regulatory milieu controlling the maintenance of bone mass. A number of neural pathways from the brain to bone have been identified. These pathways often involve elements of the energy homeostatic apparatus, indicating links between the regulation of bone metabolism and energy balance. Neuropeptide Y is one such factor that co-regulates these two processes. Initial studies outlined the skeletal actions of NPY from within the brain and the interactions with energy homeostatic processes. However, in recent years, an appreciation for the actions of NPY within bone cells has expanded. Cells of the osteoblastic lineage express both NPY ligand and a cognate receptor NPY, Y1R. Murine studies have demonstrated that both ligand and receptor actively control bone mass and osteoblast activity and interact with mechanical signals to integrate with the local loading environment. Local NPY signalling regulates osteoprogenitor production and differentiation, to cover the entire osteoblastic lineage. In addition, several recent studies have demonstrated extra-skeletal actions of osteoblastic NPY signalling, to regulate energy expenditure and with it adiposity, and in a separate study, to control release of a factor-controlling beta cell mass and insulin production/release and with it glucose tolerance. Thus, osteoblastic neuropeptide production and signalling illustrates the rapidly widening sphere of influence of skeletal tissue, and suggests a far more complex and interconnected physiology then is currently appreciated.


Asunto(s)
Huesos/metabolismo , Metabolismo Energético , Neuropéptido Y/metabolismo , Osteoblastos/metabolismo , Animales , Densidad Ósea , Proliferación Celular , Homeostasis , Hipotálamo/metabolismo , Ratones , Osteocitos/metabolismo , Receptores de Neuropéptido Y/metabolismo
4.
Sci Rep ; 6: 18614, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26726071

RESUMEN

Prader-Willi syndrome (PWS) is the predominant genetic cause of obesity in humans. Recent clinical reports have suggested that micro-deletion of the Snord116 gene cluster can lead to PWS, however, the extent of the contributions of the encoded snoRNAs is unknown. Here we show that mice lacking Snord116 globally have low birth weight, increased body weight gain, energy expenditure and hyperphagia. Consistent with this, microarray analysis of hypothalamic gene expression revealed a significant alteration in feeding related pathways that was also confirmed by in situ hybridisation. Importantly, selective deletion of Snord116 only from NPY expressing neurons mimics almost exactly the global deletion phenotype including the persistent low birth weight, increased body weight gain in early adulthood, increased energy expenditure and hyperphagia. Mechanistically, the lack of Snord116 in NPY neurons leads to the upregulation of NPY mRNA consistent with the hyperphagic phenotype and suggests a critical role of Snord116 in the control of NPY neuronal functions that might be dysregulated in PWS.


Asunto(s)
Regulación del Apetito , ARN Nucleolar Pequeño/fisiología , Animales , Composición Corporal , Peso Corporal , Metabolismo de los Hidratos de Carbono , Dieta Alta en Grasa/efectos adversos , Ingestión de Alimentos , Metabolismo Energético , Femenino , Expresión Génica , Hipotálamo/metabolismo , Masculino , Ratones Noqueados , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Obesidad/etiología , Obesidad/genética
5.
Bone ; 82: 56-63, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26545334

RESUMEN

Obesity and osteoporosis have become major public health challenges worldwide. The brain is well established as a pivotal regulator of energy homeostasis, appetite and fuel metabolism. However, there is now clear evidence for regulation between the brain and bone. Similarly, evidence also indicates that the involvement of the brain in bone and adipose regulation is both related and interdependent. The hypothalamus, with its semi-permeable blood brain barrier, is one of the most powerful regulatory regions within the body, integrating and relaying signals not only from peripheral tissues but also from within the brain itself. Two main neuronal populations within the arcuate nucleus of the hypothalamus regulate energy homeostasis: The orexigenic, appetite-stimulating neurons that co-express neuropeptide Y and agouti-related peptide and the anorexigenic, appetite-suppressing neurons that co-express proopiomelanocortin and cocaine- and amphetamine related transcript. From within the arcuate, these four neuropeptides encompass some of the most powerful control of energy homeostasis in the entire body. Moreover, they also regulate skeletal homeostasis, identifying a co-ordination network linking the processes of bone and energy homeostasis. Excitingly, the number of central neuropeptides and neural factors known to regulate bone and energy homeostasis continues to grow, with cannabinoid receptors and semaphorins also involved in bone homeostasis. These neuronal pathways represent a growing area of research that is identifying novel regulatory axes between the brain and the bone, and links with other homeostatic networks; thereby revealing a far more complex, and interdependent bone biology than previously envisioned. This review examines the current understanding of the central regulation of bone and energy metabolism.


Asunto(s)
Remodelación Ósea/fisiología , Encéfalo/metabolismo , Metabolismo Energético/fisiología , Homeostasis/fisiología , Adipocitos/metabolismo , Animales , Calcificación Fisiológica/fisiología , Humanos , Hipotálamo/metabolismo , Obesidad/metabolismo , Osteoblastos/metabolismo
6.
Bone ; 80: 95-100, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26453499

RESUMEN

The functional interplay between bone and muscle that enables locomotion is a fundamental aspect of daily life. However, other interactions between bone and muscle continue to attract attention as our understanding of the breath and importance of this inter-relationship continues to expand. Of particular interest is the regulatory connection between bone and muscle, which adds a new insight to the coordination of the bone/muscle unit. We have appreciated the importance of neuronal signaling to the control of bone turnover and muscle contraction, but recent data indicate that neuronal inputs control a far wider range of bone and muscle physiology than previously appreciated. This review outlines the role of the sympathetic nervous system and neuronal/neuropeptide inputs upon the regulation of bone and muscle tissue, and the potential for co-regulatory actions, particularly involving the sympathetic nervous system. This article is part of a Special Issue entitled "Muscle Bone Interactions".


Asunto(s)
Hipotálamo/citología , Neuronas/citología , Animales , Huesos , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Células Musculares/citología , Células Musculares/metabolismo , Neuronas/fisiología , Neuropéptido Y/metabolismo
7.
Neuropeptides ; 46(6): 383-94, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23062312

RESUMEN

Chronic opiate usage, whether prescribed or illicit, has been associated with changes in bone mass and is a recognized risk factor for the development of osteoporosis; however, the mechanism behind this effect is unknown. Here we show that lack of dynorphin, an endogenous opioid, in mice (Dyn-/-), resulted in a significantly elevated cancellous bone volume associated with greater mineral apposition rate and increased resorption indices. A similar anabolic phenotype was evident in bone of mice lacking dynorphin's cognate receptor, the kappa opioid receptor. Lack of opioid receptor expression in primary osteoblastic cultures and no change in bone cell function after dynorphin agonist treatment in vitro indicates an indirect mode of action. Consistent with a hypothalamic action, central dynorphin signaling induces extracellular signal-regulated kinase (ERK) phosphorylation and c-fos activation of neurons in the arcuate nucleus of the hypothalamus (Arc). Importantly, this signaling also leads to an increase in Arc NPY mRNA expression, a change known to decrease bone formation. Further implicating NPY in the skeletal effects of dynorphin, Dyn-/-/NPY-/- double mutant mice showed comparable increases in bone formation to single mutant mice, suggesting that dynorphin acts upstream of NPY signaling to control bone formation. Thus the dynorphin system, acting via NPY, may represent a pathway by which higher processes including stress, reward/addiction and depression influence skeletal metabolism. Moreover, understanding of these unique interactions may enable modulation of the adverse effects of exogenous opioid treatment without directly affecting analgesic responses.


Asunto(s)
Huesos/fisiología , Dinorfinas/fisiología , Homeostasis/fisiología , Animales , Western Blotting , Composición Corporal/genética , Composición Corporal/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , ADN Complementario/biosíntesis , ADN Complementario/aislamiento & purificación , Dinorfinas/genética , Femenino , Homeostasis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Neuropéptido Y/fisiología , Osteoblastos/fisiología , Embarazo , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología , Células del Estroma/fisiología , Tomografía Computarizada por Rayos X
8.
PLoS One ; 7(6): e40191, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768253

RESUMEN

Neuropeptide Y (NPY) acting in the hypothalamus is one of the most powerful orexigenic agents known. Of the five known Y receptors, hypothalamic Y1 and Y5 have been most strongly implicated in mediating hyperphagic effects. However, knockout of individual Y1 or Y5 receptors induces late-onset obesity--and Y5 receptor knockout also induces hyperphagia, possibly due to redundancy in functions of these genes. Here we show that food intake in mice requires the combined actions of both Y1 and Y5 receptors. Germline Y1Y5 ablation in Y1Y5(-/-) mice results in hypophagia, an effect that is at least partially mediated by the hypothalamus, since mice with adult-onset Y1Y5 receptor dual ablation targeted to the paraventricular nucleus (PVN) of the hypothalamus (Y1Y5(Hyp/Hyp)) also exhibit reduced spontaneous or fasting-induced food intake when fed a high fat diet. Interestingly, despite hypophagia, mice with germline or hypothalamus-specific Y1Y5 deficiency exhibited increased body weight and/or increased adiposity, possibly due to compensatory responses to gene deletion, such as the decreased energy expenditure observed in male Y1Y5(-/-) animals relative to wildtype values. While Y1 and Y5 receptors expressed in other hypothalamic areas besides the PVN--such as the dorsomedial nucleus and the ventromedial hypothalamus--cannot be excluded from having a role in the regulation of food intake, these studies demonstrate the pivotal, combined role of both Y1 and Y5 receptors in the mediation of food intake.


Asunto(s)
Metabolismo Energético , Conducta Alimentaria , Homeostasis , Receptores de Neuropéptido Y/metabolismo , Adiposidad/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Animales , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Ayuno , Conducta Alimentaria/efectos de los fármacos , Eliminación de Gen , Células Germinativas/efectos de los fármacos , Células Germinativas/metabolismo , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Insulina/farmacología , Ratones , Ratones Noqueados , Neuropéptido Y/metabolismo , Obesidad/patología , Especificidad de Órganos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
9.
J Mol Neurosci ; 43(2): 123-31, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20635164

RESUMEN

Both the neuropeptide Y (NPY) and the leptin systems have been shown to be important central mediators of bone metabolism. However, the interaction between these two systems is complex and not fully understood. Here, we show that a unique interaction exists between Y2 and Y4 receptors in the regulation of bone homeostasis that is not evident when combined with lack of Y1 signalling. Despite the hypoleptinaemia shown in male Y2/Y4 double knockout (Y2⁻/⁻ Y4⁻/⁻) mice, when on the leptin-deficient ob/ob background, these mice display reduced cancellous bone mass. However, combined Y2/Y4 deletion enhances the effect of leptin deficiency on the cortical bone compartment. By replicating the enhanced central NPY expression evident in ob/ob mice using virally mediated overexpression of NPY in the hypothalamus of Y receptor knockout mice, we demonstrate that Y2⁻/⁻ Y4⁻/⁻ mice have an exaggerated response to the anti-osteogenic effects of elevated hypothalamic NPY in both cancellous and cortical bone and that this effect appears to be dependent on Y1 receptor signalling. This study highlights the complex interaction between Y receptors in the control of bone mass. Moreover, it suggests that the reduction in cortical bone observed in the absence of leptin is due to the anti-osteogenic effect of elevated hypothalamic NPY levels.


Asunto(s)
Huesos/metabolismo , Neuropéptido Y/metabolismo , Receptores de Neuropéptido Y/metabolismo , Transducción de Señal/fisiología , Animales , Femenino , Homeostasis , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , Receptores de Neuropéptido Y/genética
10.
Eur Cell Mater ; 20: 431-41; discussion 441-2, 2010 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-21181649

RESUMEN

The effects of bone anabolic agents such as bone morphogenetic proteins (BMPs) have the potential to be augmented by co-treatment with an anti-catabolic such as a bisphosphonate. We hypothesised that the effects of bisphosphonates on BMP-induced bone anabolism would be dose dependent, and we aimed to test this in a small animal model. Agents were delivered locally using a biodegradable poly-D, L-lactic-acid (PDLLA) polymer delivery system. Recombinant human BMP-7 (25 µg) was tested with a range of doses of the bisphosphonate pamidronate (0.02 mg, 0.2 mg and 2 mg local PAM; 0.3 mg/kg and 3 mg/kg thrice-weekly systemic PAM) versus BMP-7 alone. Polymer pellets were surgically implanted in the hind limbs of female C57BL6/J mice (8-10 week) and ectopic bone nodules were harvested at 3 and 8 weeks post-operatively. At 3 weeks, local low dose PAM (0.02 mg) induced a 102% increase in rhBMP-7 induced bone volume (p<0.01) as measured by miroCT, and this was comparable to systemic PAM (0.3 mg/kg thrice-weekly). In contrast, local high dose PAM (2 mg) resulted in a 97% decrease in bone volume (p<0.01). Radiography and histology indicated that the polymer vehicle was still largely present at 8 weeks indicating inefficient biodegradation. This is the first study to validate the utility of local co-delivery of BMP/bisphosphonate via biodegradable polymer and supports the continued refinement of more advanced bioresorbable delivery systems for clinical applications.


Asunto(s)
Conservadores de la Densidad Ósea/administración & dosificación , Proteína Morfogenética Ósea 7/administración & dosificación , Huesos/efectos de los fármacos , Difosfonatos/administración & dosificación , Ácido Láctico , Polímeros , Proteínas Recombinantes/administración & dosificación , Animales , Conservadores de la Densidad Ósea/farmacología , Proteína Morfogenética Ósea 7/farmacología , Huesos/fisiología , Portadores de Fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Osteogénesis , Pamidronato , Poliésteres , Músculo Cuádriceps , Proteínas Recombinantes/farmacología , Ingeniería de Tejidos
11.
J Mol Endocrinol ; 45(4): 175-81, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660619

RESUMEN

On initial inspection, bone remodeling, the process whereby the skeleton adapts through time, appears to be relatively simple. Two cell types, the bone-forming osteoblasts and the bone-resorbing osteoclasts, interact to keep bone mass relatively stable throughout adult life. However, the complexity of the regulatory influences on these cells is continuing to expand our understanding of the intricacy of skeletal physiology and also the interactions between other organ systems and bone. One such example of the broadening of understanding in this field has occurred in the last decade with study of the central, neural regulation of bone mass. Initial studies of an adipose-derived hormone, leptin, helped define a direct, sympathetic pathway involving efferent neural signals from the hypothalamus to receptors on the osteoblast. Since the leptin-mediated pathway has been continuously modified to reveal a complex system involving neuromedin U, cocaine- and amphetamine-related transcript and serotonin interacting within the hypothalamus and brainstem to regulate both bone formation and resorption in cancellous bone, a number of other systems have also been identified. Neuropeptide Y, acting through hypothalamic Y2 receptors, is capable of skeleton-wide modulation of osteoblast activity, with important coordination between body weight and bone mass. Cannabinoids, acting through central cannabinoid receptor 1 and bone cell cannabinoid receptor 2 receptors, modulate osteoclast activity, thereby identifying pathways active on both aspects of the bone remodeling process. This review explores the key central pathways to bone and explores the complexity of the interactions being revealed by this emergent field of research.


Asunto(s)
Huesos/metabolismo , Hipotálamo/metabolismo , Animales , Peso Corporal , Humanos , Leptina/metabolismo , Osteogénesis/fisiología , Serotonina/metabolismo
12.
J Biol Chem ; 285(36): 28164-73, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20558734

RESUMEN

PTH stimulates osteoblastic cells to form new bone and to produce osteoblast-osteoclast coupling factors such as RANKL. Whether osteoclasts or their activity are needed for PTH anabolism remains uncertain. We treated ovariectomized huRANKL knock-in mice with a human RANKL inhibitor denosumab (DMAb), alendronate (Aln), or vehicle for 4 weeks, followed by co-treatment with intermittent PTH for 4 weeks. Loss of bone mass and microarchitecture was prevented by Aln and further significantly improved by DMAb. PTH improved bone mass, microstructure, and strength, and was additive to Aln but not to DMAb. Aln inhibited biochemical and histomorphometrical indices of bone turnover,--i.e. osteocalcin and bone formation rate (BFR) on cancellous bone surfaces-, and Dmab inhibited them further. However Aln increased whereas Dmab suppressed osteoclast number and surfaces. PTH significantly increased osteocalcin and bone formation indices, in the absence or presence of either antiresorptive, although BFR remained lower in presence of Dmab. To further evaluate PTH effects in the complete absence of osteoclasts, high dose PTH was administered to RANK(-/-) mice. PTH increased osteocalcin similarly in RANK(-/-) and WT mice. It also increased BMD in RANK(-/-) mice, although less than in WT. These results further indicate that osteoclasts are not strictly required for PTH anabolism, which presumably still occurs via stimulation of modeling-based bone formation. However the magnitude of PTH anabolic effects on the skeleton, in particular its additive effects with antiresorptives, depends on the extent of the remodeling space, as determined by the number and activity of osteoclasts on bone surfaces.


Asunto(s)
Alendronato/farmacología , Anticuerpos Monoclonales/farmacología , Huesos/metabolismo , Técnicas de Sustitución del Gen , Osteoclastos/efectos de los fármacos , Hormona Paratiroidea/farmacología , Ligando RANK/farmacología , Receptor Activador del Factor Nuclear kappa-B/genética , Alendronato/administración & dosificación , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Biomarcadores/metabolismo , Densidad Ósea/efectos de los fármacos , Resorción Ósea/tratamiento farmacológico , Huesos/citología , Huesos/efectos de los fármacos , Huesos/fisiología , Denosumab , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Ovariectomía , Hormona Paratiroidea/administración & dosificación , Ligando RANK/administración & dosificación , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/deficiencia , Receptor Activador del Factor Nuclear kappa-B/metabolismo
13.
Trends Endocrinol Metab ; 21(7): 411-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20202858

RESUMEN

The hypothalamus regulates the skeleton and adipose tissue via endocrine mechanisms. Changes in sex steroid levels in menopause and aging are central to the associated changes in bone mass and adiposity. Whereas many of these effects occur via direct actions on osteoblasts or adipocytes, sex hormones can also mediate effects on bone and adipose tissue via interaction with neuronal pathways. A key hypothalamic regulator of bone and adipose tissue is neuropeptide Y (NPY), which coordinately influences these tissues via effects on neuroendocrine and sympathetic nervous output. Better understanding of the interaction between NPY and sex steroids in regulating skeletal and energy homeostasis could lead to more effective treatments for osteoporosis and obesity.


Asunto(s)
Tejido Adiposo/fisiología , Huesos/fisiología , Hormonas Esteroides Gonadales/fisiología , Hipotálamo/fisiología , Neuropéptido Y/fisiología , Animales , Humanos
14.
PLoS One ; 4(12): e8415, 2009 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-20027231

RESUMEN

Changes in whole body energy levels are closely linked to alterations in body weight and bone mass. Here, we show that hypothalamic signals contribute to the regulation of bone mass in a manner consistent with the central perception of energy status. Mice lacking neuropeptide Y (NPY), a well-known orexigenic factor whose hypothalamic expression is increased in fasting, have significantly increased bone mass in association with enhanced osteoblast activity and elevated expression of bone osteogenic transcription factors, Runx2 and Osterix. In contrast, wild type and NPY knockout (NPY (-/-)) mice in which NPY is specifically over expressed in the hypothalamus (AAV-NPY+) show a significant reduction in bone mass despite developing an obese phenotype. The AAV-NPY+ induced loss of bone mass is consistent with models known to mimic the central effects of fasting, which also show increased hypothalamic NPY levels. Thus these data indicate that, in addition to well characterized responses to body mass, skeletal tissue also responds to the perception of nutritional status by the hypothalamus independently of body weight. In addition, the reduction in bone mass by AAV NPY+ administration does not completely correct the high bone mass phenotype of NPY (-/-) mice, indicating the possibility that peripheral NPY may also be an important regulator of bone mass. Indeed, we demonstrate the expression of NPY specifically in osteoblasts. In conclusion, these data identifies NPY as a critical integrator of bone homeostatic signals; increasing bone mass during times of obesity when hypothalamic NPY expression levels are low and reducing bone formation to conserve energy under 'starving' conditions, when hypothalamic NPY expression levels are high.


Asunto(s)
Peso Corporal/fisiología , Huesos/anatomía & histología , Neuropéptido Y/deficiencia , Adiposidad , Animales , Huesos/citología , Huesos/metabolismo , Femenino , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Neuropéptido Y/metabolismo , Tamaño de los Órganos , Osteogénesis , Fenotipo , Transducción de Señal
15.
Semin Cell Dev Biol ; 19(5): 452-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18761098

RESUMEN

The traditional view of skeletal homeostasis as a primarily endocrine activity has been expanded in recent years following the identification of direct neural pathways controlling bone homeostasis via central relays. Powerful control over both anabolic and catabolic activities have been isolated to neurons of the hypothalamus, enabling large changes in bone mass to be achieved by minute changes in the levels of these central neural signals. Initiated by studies of leptin and expanding rapidly, the breadth and complexity of this regulatory axis to bone is sure to increase. Critically though, the translation of these findings into therapeutic interventions is likely to present a greater challenge. However, the contribution to our understanding that these initial studies are making indicates an exciting potential to help to alleviate the growing challenge presented by musculoskeletal disease.


Asunto(s)
Fenómenos Biológicos , Hipotálamo/fisiología , Leptina/fisiología , Animales , Densidad Ósea , Desarrollo Óseo , Huesos/fisiología , Humanos , Hipotálamo/metabolismo , Leptina/metabolismo , Leptina/farmacología , Melanocortinas/metabolismo , Neuronas/fisiología , Neuropéptido Y/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Receptores de Cannabinoides/metabolismo , Receptores de Neuropéptido Y/metabolismo , Sistema Nervioso Simpático/fisiología
16.
J Biol Chem ; 282(26): 19092-102, 2007 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-17491016

RESUMEN

The importance of neuropeptide Y (NPY) and Y2 receptors in the regulation of bone and energy homeostasis has recently been demonstrated. However, the contributions of the other Y receptors are less clear. Here we show that Y1 receptors are expressed on osteoblastic cells. Moreover, bone and adipose tissue mass are elevated in Y1(-/-) mice with a generalized increase in bone formation on cortical and cancellous surfaces. Importantly, the inhibitory effects of NPY on bone marrow stromal cells in vitro are absent in cells derived from Y1(-/-) mice, indicating a direct action of NPY on bone cells via this Y receptor. Interestingly, in contrast to Y2 receptor or germ line Y1 receptor deletion, conditional deletion of hypothalamic Y1 receptors in adult mice did not alter bone homeostasis, food intake, or adiposity. Furthermore, deletion of both Y1 and Y2 receptors did not produce additive effects in bone or adiposity. Thus Y1 receptor pathways act powerfully to inhibit bone production and adiposity by nonhypothalamic pathways, with potentially direct effects on bone tissue through a single pathway with Y2 receptors.


Asunto(s)
Huesos/metabolismo , Metabolismo Energético/fisiología , Homeostasis/fisiología , Receptores de Neuropéptido Y/genética , Receptores de Neuropéptido Y/metabolismo , Factores de Edad , Animales , Conducta Animal/fisiología , Densidad Ósea/fisiología , Huesos/citología , Células Cultivadas , Femenino , Hipotálamo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/metabolismo , Fenotipo , Células del Estroma/metabolismo
17.
J Bone Miner Res ; 21(10): 1600-7, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16995815

RESUMEN

UNLABELLED: NeuropeptideY-, Y2 receptor (Y2)-, and leptin-deficient mice show similar anabolic action in cancellous bone but have not been assessed in cortical bone. Cortical bone mass is elevated in Y2(-/-) mice through greater osteoblast activity. In contrast, leptin deficiency results in reduced bone mass. We show opposing central regulation of cortical bone. INTRODUCTION: Treatment of osteoporosis is confounded by a lack of agents capable of stimulating the formation of bone by osteoblasts. Recently, the brain has been identified as a potent anabolic regulator of bone formation. Hypothalamic leptin or Y2 receptor signaling are known to regulate osteoblast activity in cancellous bone. However, assessment of these pathways in the structural cortical bone is critical to understanding their role in skeletal health and their potential clinical relevance to osteoporosis and its treatment. MATERIALS AND METHODS: Long bones of 16-week male ob/ob and germline and hypothalamic Y2(-/-) mice were assessed by QCT. Cortical osteoblast activity was assessed histologically. RESULTS: The femora of skeletally mature Y2(-/-) mice and of leptin-deficient ob/ob and Y2(-/-)ob/ob mice were assessed for changes in cortical osteoblast activity and bone mass. Ablation of Y2 receptors increased osteoblast activity on both endosteal and periosteal surfaces, independent of leptin, resulting in increased cortical bone mass and density in Y2(-/-) mice along the entire femur. Importantly, these changes were evident after deletion of hypothalamic Y2 receptors in adult mice, with a 5-fold elevation in periosteal bone formation. This is in marked contrast to leptin-deficient models that displayed reduced cortical mass and density. These changes were associated with substantial differences in calculated strength between the Y2(-/-) and leptin-deficient mice. CONCLUSIONS: These results indicate that the Y2-mediated anabolic pathway stimulates cortical and cancellous bone formation, whereas the leptin-mediated pathway has opposing effects in cortical and cancellous bone, diminishing the production of cortical bone. The findings from conditional hypothalamic Y2 knockout show a novel, inducible control mechanism for cortical bone formation and a potential new pathway for anabolic treatment of osteoporosis.


Asunto(s)
Hipotálamo/metabolismo , Leptina/metabolismo , Osteogénesis/fisiología , Receptores de Neuropéptido Y/metabolismo , Animales , Fémur/metabolismo , Fémur/patología , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Osteoblastos/metabolismo , Osteoblastos/patología , Receptores de Leptina , Receptores de Neuropéptido Y/deficiencia , Receptores de Neuropéptido Y/genética , Transducción de Señal
18.
J Bone Miner Res ; 20(10): 1851-7, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16160743

RESUMEN

UNLABELLED: Leptin and Y2 receptors on hypothalamic NPY neurons mediate leptin effects on energy homeostasis; however, their interaction in modulating osteoblast activity is not established. Here, direct testing of this possibility indicates distinct mechanisms of action for leptin anti-osteogenic and Y2-/- anabolic pathways in modulating bone formation. INTRODUCTION: Central enhancement of bone formation by hypothalamic neurons is observed in leptin-deficient ob/ob and Y2 receptor null mice. Similar elevation in central neuropeptide Y (NPY) expression and effects on osteoblast activity in these two models suggest a shared pathway between leptin and Y2 receptors in the central control of bone physiology. The aim of this study was to test whether the leptin and Y2 receptor pathways regulate bone by the same or distinct mechanisms. MATERIALS AND METHODS: The interaction of concomitant leptin and Y2 receptor deficiency in controlling bone was examined in Y2-/- ob/ob double mutant mice, to determine whether leptin and Y2 receptor deficiency have additive effects. Interaction between leptin excess and Y2 receptor deletion was examined using recombinant adeno-associated viral vector overproduction of NPY (AAV-NPY) to produce weight gain and thus leptin excess in adult Y2-/- mice. Cancellous bone volume and bone cell function were assessed. RESULTS: Osteoblast activity was comparably elevated in ob/ob, Y2-/-, and Y2-/- ob/ob mice. However, greater bone resorption in ob/ob and Y2-/- ob/ob mice reduced cancellous bone volume compared with Y2-/-. Both wildtype and Y2-/- AAV-NPY mice exhibited marked elevation of white adipose tissue accumulation and hence leptin expression, thereby reducing osteoblast activity. Despite this anti-osteogenic leptin effect in the obese AAV-NPY model, osteoblast activity in Y2-/- AAV-NPY mice remained significantly greater than in wildtype AAV-NPY mice. CONCLUSIONS: This study suggests that NPY is not a key regulator of the leptin-dependent osteoblast activity, because both the leptin-deficient stimulation of bone formation and the excess leptin inhibition of bone formation can occur in the presence of high hypothalamic NPY. The Y2-/- pathway acts consistently to stimulate bone formation; in contrast, leptin continues to suppress bone formation as circulating levels increase. As a result, they act increasingly in opposition as obesity becomes more marked. Thus, in the absence of leptin, the cancellous bone response to loss of Y2 receptor and leptin activity can not be distinguished. However, as leptin levels increase to physiological levels, distinct signaling pathways are revealed.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Osteogénesis/fisiología , Receptores de Neuropéptido Y/metabolismo , Transducción de Señal/fisiología , Animales , Leptina/deficiencia , Ratones , Ratones Noqueados , Neuropéptido Y/metabolismo , Osteoblastos/metabolismo , Receptores de Leptina , Receptores de Neuropéptido Y/genética
19.
J Clin Invest ; 109(7): 915-21, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11927618

RESUMEN

Neuropeptide Y (NPY) is a downstream modulator of leptin action, possibly at the level of the arcuate nucleus where NPY neurons are known to express both leptin receptors and Y2 receptors. In addition to the well-described role of NPY and leptin in energy balance and obesity, intracerebroventricular administration of NPY or leptin also causes bone loss. Here we show that Y2 receptor-deficient mice have a twofold increase in trabecular bone volume as well as greater trabecular number and thickness compared with control mice. We also demonstrate that central Y2 receptors are crucial for this process, since selective deletion of hypothalamic Y2 receptors in mature conditional Y2 knockout mice results in an identical increase in trabecular bone volume within 5 weeks. This hypothalamus-specific Y2 receptor deletion stimulates osteoblast activity and increases the rate of bone mineralization and formation, with no effect on osteoblast or osteoclast surface measurements. The lack of any changes in plasma total calcium, leptinemia, or hypothalamo-pituitary-corticotropic, -thyrotropic, -somatotropic, or -gonadotropic output suggests that Y2 receptors do not modulate bone formation by humoral mechanisms, and that alteration of autonomic function through hypothalamic Y2 receptors may play a key role in a major central regulatory circuit of bone formation.


Asunto(s)
Desarrollo Óseo , Hipotálamo/metabolismo , Receptores de Neuropéptido Y/fisiología , Animales , Huesos/fisiología , Femenino , Fémur/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Neuropéptido Y/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA