Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell Metab ; 35(9): 1613-1629.e8, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37572666

ABSTRACT

Hypothalamic gliosis associated with high-fat diet (HFD) feeding increases susceptibility to hyperphagia and weight gain. However, the body-weight-independent contribution of microglia to glucose regulation has not been determined. Here, we show that reducing microglial nuclear factor κB (NF-κB) signaling via cell-specific IKKß deletion exacerbates HFD-induced glucose intolerance despite reducing body weight and adiposity. Conversely, two genetic approaches to increase microglial pro-inflammatory signaling (deletion of an NF-κB pathway inhibitor and chemogenetic activation through a modified Gq-coupled muscarinic receptor) improved glucose tolerance independently of diet in both lean and obese rodents. Microglial regulation of glucose homeostasis involves a tumor necrosis factor alpha (TNF-α)-dependent mechanism that increases activation of pro-opiomelanocortin (POMC) and other hypothalamic glucose-sensing neurons, ultimately leading to a marked amplification of first-phase insulin secretion via a parasympathetic pathway. Overall, these data indicate that microglia regulate glucose homeostasis in a body-weight-independent manner, an unexpected mechanism that limits the deterioration of glucose tolerance associated with obesity.


Subject(s)
Microglia , NF-kappa B , Humans , Microglia/metabolism , NF-kappa B/metabolism , Obesity/metabolism , Body Weight/physiology , Glucose/metabolism , Hypothalamus/metabolism , Diet, High-Fat
2.
Am J Physiol Endocrinol Metab ; 324(5): E461-E475, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37053049

ABSTRACT

Hypogonadism in males confers elevated cardiovascular disease (CVD) risk by unknown mechanisms. Recent radiological evidence suggests that low testosterone (T) is associated with mediobasal hypothalamic (MBH) gliosis, a central nervous system (CNS) cellular response linked to metabolic dysfunction. To address mechanisms linking CNS androgen action to CVD risk, we generated a hypogonadal, hyperlipidemic mouse model with orchiectomy (ORX) combined with hepatic PCSK9 overexpression. After 4 wk of high-fat, high-sucrose diet (HFHS) consumption, despite equal body weights and glucose tolerance, androgen-deficient ORX mice had a more atherogenic lipid profile and increased liver and leukocyte inflammatory signaling compared with sham-operated control mice. Along with these early CVD risk indicators, ORX markedly amplified HFHS-induced astrogliosis in the MBH. Transcriptomic analysis further revealed that ORX and high-fat diet feeding induced upregulation of inflammatory pathways and downregulation of metabolic pathways in hypothalamic astrocytes. To interrogate the role of sex steroid signaling in the CNS in cardiometabolic risk and MBH inflammation, central infusion of T and dihydrotestosterone (DHT) was performed on ORX mice. Central DHT prevented MBH astrogliosis and reduced the liver inflammatory signaling and monocytosis induced by HFHS and ORX; T had a partial protective effect. Finally, a cross-sectional study in 41 adult men demonstrated a positive correlation between radiological evidence of MBH gliosis and plasma lipids. These findings demonstrate that T deficiency in combination with a Western-style diet promotes hypothalamic gliosis concomitant with increased atherogenic risk factors and provide supportive evidence for regulation of lipid metabolism and cardiometabolic risk determinants by the CNS action of sex steroids.NEW & NOTEWORTHY This study provides evidence that hypothalamic gliosis is a key early event through which androgen deficiency in combination with a Western-style diet might lead to cardiometabolic dysregulation in males. Furthermore, this work provides the first evidence in humans of a positive association between hypothalamic gliosis and LDL-cholesterol, advancing our knowledge of CNS influences on CVD risk progression.


Subject(s)
Androgens , Cardiovascular Diseases , Humans , Mice , Male , Animals , Proprotein Convertase 9 , Diet, High-Fat/adverse effects , Gliosis , Orchiectomy , Cross-Sectional Studies , Risk Factors , Dihydrotestosterone
3.
Diabetes ; 72(2): 233-244, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36318114

ABSTRACT

In rodents, susceptibility to diet-induced obesity requires microglial activation, but the molecular components of this pathway remain incompletely defined. Prostaglandin PGE2 levels increase in the mediobasal hypothalamus during high-fat-diet (HFD) feeding, and the PGE2 receptor EP4 regulates microglial activation state and phagocytic activity, suggesting a potential role for microglial EP4 signaling in obesity pathogenesis. To test the role of microglial EP4 in energy balance regulation, we analyzed the metabolic phenotype in a microglia-specific EP4 knockout (MG-EP4 KO) mouse model. Microglial EP4 deletion markedly reduced weight gain and food intake in response to HFD feeding. Corresponding with this lean phenotype, insulin sensitivity was also improved in HFD-fed MG-EP4 KO mice, though glucose tolerance remained surprisingly unaffected. Mechanistically, EP4-deficient microglia showed an attenuated phagocytic state marked by reduced CD68 expression and fewer contacts with pro-opiomelanocortin (POMC) neuron processes. These cellular changes observed in the MG-EP4 KO mice corresponded with an increased density of POMC neurites extending into the paraventricular nucleus (PVN). These findings reveal that microglial EP4 signaling promotes body weight gain and insulin resistance during HFD feeding. Furthermore, the data suggest that curbing microglial phagocytic function may preserve POMC cytoarchitecture and PVN input to limit overconsumption during diet-induced obesity.


Subject(s)
Dinoprostone , Microglia , Obesity , Animals , Mice , Diet, High-Fat/adverse effects , Dinoprostone/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Obesity/genetics , Obesity/metabolism , Phagocytosis , Pro-Opiomelanocortin/metabolism , Weight Gain
4.
Int J Mol Sci ; 23(12)2022 Jun 07.
Article in English | MEDLINE | ID: mdl-35742824

ABSTRACT

Both hypothalamic microglial inflammation and melanocortin pathway dysfunction contribute to diet-induced obesity (DIO) pathogenesis. Previous studies involving models of altered microglial signaling demonstrate altered DIO susceptibility with corresponding POMC neuron cytological changes, suggesting a link between microglia and the melanocortin system. We addressed this hypothesis using the specific microglial silencing molecule, CX3CL1 (fractalkine), to determine whether reducing hypothalamic microglial activation can restore POMC/melanocortin signaling to protect against DIO. We performed metabolic analyses in high fat diet (HFD)-fed mice with targeted viral overexpression of CX3CL1 in the hypothalamus. Electrophysiologic recording in hypothalamic slices from POMC-MAPT-GFP mice was used to determine the effects of HFD feeding and microglial silencing via minocycline or CX3CL1 on GFP-labeled POMC neurons. Finally, mice with hypothalamic overexpression of CX3CL1 received central treatment with the melanocortin receptor antagonist SHU9119 to determine whether melanocortin signaling is required for the metabolic benefits of CX3CL1. Hypothalamic overexpression of CX3CL1 increased leptin sensitivity and POMC gene expression, while reducing weight gain in animals fed an HFD. In electrophysiological recordings from hypothalamic slice preparations, HFD feeding was associated with reduced POMC neuron excitability and increased amplitude of inhibitory postsynaptic currents. Microglial silencing using minocycline or CX3CL1 treatment reversed these HFD-induced changes in POMC neuron electrophysiologic properties. Correspondingly, blockade of melanocortin receptor signaling in vivo prevented both the acute and chronic reduction in food intake and body weight mediated by CX3CL1. Our results show that suppressing microglial activation during HFD feeding reduces DIO susceptibility via a mechanism involving increased POMC neuron excitability and melanocortin signaling.


Subject(s)
Diet, High-Fat , Melanocortins , Animals , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Melanocortins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Minocycline/pharmacology , Neurons/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism
6.
Cell Metab ; 26(1): 185-197.e3, 2017 Jul 05.
Article in English | MEDLINE | ID: mdl-28683286

ABSTRACT

Dietary excess triggers accumulation of pro-inflammatory microglia in the mediobasal hypothalamus (MBH), but the components of this microgliosis and its metabolic consequences remain uncertain. Here, we show that microglial inflammatory signaling determines the immunologic response of the MBH to dietary excess and regulates hypothalamic control of energy homeostasis in mice. Either pharmacologically depleting microglia or selectively restraining microglial NF-κB-dependent signaling sharply reduced microgliosis, an effect that includes prevention of MBH entry by bone-marrow-derived myeloid cells, and greatly limited diet-induced hyperphagia and weight gain. Conversely, forcing microglial activation through cell-specific deletion of the negative NF-κB regulator A20 induced spontaneous MBH microgliosis and cellular infiltration, reduced energy expenditure, and increased both food intake and weight gain even in absence of a dietary challenge. Thus, microglial inflammatory activation, stimulated by dietary excess, orchestrates a multicellular hypothalamic response that mediates obesity susceptibility, providing a mechanistic rationale for non-neuronal approaches to treat metabolic diseases.


Subject(s)
Appetite Regulation , Energy Metabolism , Hypothalamus/immunology , Inflammation/immunology , Microglia/immunology , Obesity/immunology , Animals , Hyperphagia/immunology , Hyperphagia/metabolism , Hyperphagia/physiopathology , Hypothalamus/metabolism , Hypothalamus/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , NF-kappa B/immunology , NF-kappa B/metabolism , Obesity/metabolism , Obesity/physiopathology , Signal Transduction
7.
Nat Commun ; 8: 14556, 2017 02 22.
Article in English | MEDLINE | ID: mdl-28223698

ABSTRACT

Female mice are less susceptible to the negative metabolic consequences of high-fat diet feeding than male mice, for reasons that are incompletely understood. Here we identify sex-specific differences in hypothalamic microglial activation via the CX3CL1-CX3CR1 pathway that mediate the resistance of female mice to diet-induced obesity. Female mice fed a high-fat diet maintain CX3CL1-CX3CR1 levels while male mice show reductions in both ligand and receptor expression. Female Cx3cr1 knockout mice develop 'male-like' hypothalamic microglial accumulation and activation, accompanied by a marked increase in their susceptibility to diet-induced obesity. Conversely, increasing brain CX3CL1 levels in male mice through central pharmacological administration or virally mediated hypothalamic overexpression converts them to a 'female-like' metabolic phenotype with reduced microglial activation and body-weight gain. These data implicate sex differences in microglial activation in the modulation of energy homeostasis and identify CX3CR1 signalling as a potential therapeutic target for the treatment of obesity.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Microglia/metabolism , Microglia/pathology , Obesity/metabolism , Obesity/pathology , Sex Characteristics , Signal Transduction , Animals , CX3C Chemokine Receptor 1/deficiency , Calcium-Binding Proteins/metabolism , Diet, High-Fat , Disease Susceptibility , Estrogens/pharmacology , Feeding Behavior/drug effects , Female , Hypothalamus/pathology , Inflammation/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Microglia/drug effects , Phenotype , Weight Gain
8.
Diabetes ; 66(4): 920-934, 2017 04.
Article in English | MEDLINE | ID: mdl-28073831

ABSTRACT

Effectors of the phosphoinositide 3-kinase (PI3K) signal transduction pathway contribute to the hypothalamic regulation of energy and glucose homeostasis in divergent ways. Here we show that central nervous system (CNS) action of the PI3K signaling intermediate atypical protein kinase C (aPKC) constrains food intake, weight gain, and glucose intolerance in both rats and mice. Pharmacological inhibition of CNS aPKC activity acutely increases food intake and worsens glucose tolerance in chow-fed rodents and causes excess weight gain during high-fat diet (HFD) feeding. Similarly, selective deletion of the aPKC isoform Pkc-λ in proopiomelanocortin (POMC) neurons disrupts leptin action, reduces melanocortin content in the paraventricular nucleus, and markedly increases susceptibility to obesity, glucose intolerance, and insulin resistance specifically in HFD-fed male mice. These data implicate aPKC as a novel regulator of energy and glucose homeostasis downstream of the leptin-PI3K pathway in POMC neurons.


Subject(s)
Eating/genetics , Glucose Intolerance/genetics , Glucose/metabolism , Isoenzymes/genetics , Neurons/metabolism , Obesity/genetics , Protein Kinase C/genetics , Weight Gain/genetics , Animals , Diet, High-Fat , Eating/drug effects , Energy Metabolism/drug effects , Energy Metabolism/genetics , Glucose Intolerance/metabolism , Hypothalamus/metabolism , Insulin Resistance , Leptin/metabolism , Male , Melanocortins/metabolism , Mice , Obesity/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pro-Opiomelanocortin/drug effects , Pro-Opiomelanocortin/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Signal Transduction , Weight Gain/drug effects
9.
Diabetologia ; 60(2): 226-236, 2017 02.
Article in English | MEDLINE | ID: mdl-27986987

ABSTRACT

Body weight stability requires homeostatic regulation to balance energy intake and energy expenditure. Research on this system and how it is affected by obesity has largely focused on the role of hypothalamic neurons as integrators of information about long-term fuel storage, short-term nutrient availability and metabolic demand. Recent studies have uncovered glial cells as additional contributors to energy balance regulation and obesity pathogenesis. Beginning with early work on leptin signalling in astrocytes, this area of research rapidly emerged after the discovery of hypothalamic inflammation and gliosis in obese rodents and humans. Current studies have revealed the involvement of a wide variety of glial cell types in the modulation of neuronal activity, regulation of hormone and nutrient availability, and participation in the physiological regulation of feeding behaviour. In addition, one glial type, microglia, has recently been implicated in susceptibility to diet-induced obesity. Together, these exciting new findings deepen our understanding of energy homeostasis regulation and raise the possibility of identifying novel mechanisms that contribute to the pathogenesis of obesity.


Subject(s)
Homeostasis/physiology , Neuroglia/metabolism , Obesity/metabolism , Animals , Body Weight/genetics , Body Weight/physiology , Central Nervous System/metabolism , Homeostasis/genetics , Humans , Hypothalamus/metabolism , Obesity/genetics
10.
Nat Med ; 22(7): 800-6, 2016 07.
Article in English | MEDLINE | ID: mdl-27213816

ABSTRACT

Type 2 diabetes (T2D) is among the most common and costly disorders worldwide. The goal of current medical management for T2D is to transiently ameliorate hyperglycemia through daily dosing of one or more antidiabetic drugs. Hypoglycemia and weight gain are common side effects of therapy, and sustained disease remission is not obtainable with nonsurgical approaches. On the basis of the potent glucose-lowering response elicited by activation of brain fibroblast growth factor (FGF) receptors, we explored the antidiabetic efficacy of centrally administered FGF1, which, unlike other FGF peptides, activates all FGF receptor subtypes. We report that a single intracerebroventricular injection of FGF1 at a dose one-tenth of that needed for antidiabetic efficacy following peripheral injection induces sustained diabetes remission in both mouse and rat models of T2D. This antidiabetic effect is not secondary to weight loss, does not increase the risk of hypoglycemia, and involves a novel and incompletely understood mechanism for increasing glucose clearance from the bloodstream. We conclude that the brain has an inherent potential to induce diabetes remission and that brain FGF receptors are potential pharmacological targets for achieving this goal.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Fibroblast Growth Factor 1/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Body Composition , Brain/drug effects , Brain/metabolism , Carbon Radioisotopes , Deoxyglucose , Diet, High-Fat , Disease Models, Animal , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Forkhead Box Protein O1/genetics , Glucose Tolerance Test , Heart/drug effects , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/metabolism , Hyperglycemia/metabolism , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Liver/metabolism , Male , Mice , Mice, Knockout , Mice, Obese , Molecular Chaperones , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myocardium/metabolism , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Zucker , Real-Time Polymerase Chain Reaction , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/genetics , Remission Induction
11.
Proc Natl Acad Sci U S A ; 113(14): E2073-82, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-27001850

ABSTRACT

Previous studies implicate the hypothalamic ventromedial nucleus (VMN) in glycemic control. Here, we report that selective inhibition of the subset of VMN neurons that express the transcription factor steroidogenic-factor 1 (VMN(SF1) neurons) blocks recovery from insulin-induced hypoglycemia whereas, conversely, activation of VMN(SF1) neurons causes diabetes-range hyperglycemia. Moreover, this hyperglycemic response is reproduced by selective activation of VMN(SF1) fibers projecting to the anterior bed nucleus of the stria terminalis (aBNST), but not to other brain areas innervated by VMN(SF1) neurons. We also report that neurons in the lateral parabrachial nucleus (LPBN), a brain area that is also implicated in the response to hypoglycemia, make synaptic connections with the specific subset of glucoregulatory VMN(SF1) neurons that project to the aBNST. These results collectively establish a physiological role in glucose homeostasis for VMN(SF1) neurons and suggest that these neurons are part of an ascending glucoregulatory LPBN→VMN(SF1)→aBNST neurocircuit.


Subject(s)
Blood Glucose/metabolism , Neurons, Afferent/physiology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Insulin/administration & dosage , Mice , Ventromedial Hypothalamic Nucleus/cytology
12.
Curr Opin Endocrinol Diabetes Obes ; 22(5): 325-30, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26192704

ABSTRACT

PURPOSE OF REVIEW: Hypothalamic inflammation and gliosis are recently discovered mechanisms that may contribute to obesity pathogenesis. Current research in this area suggests that investigation of these central nervous system responses may provide opportunities to develop new weight loss treatments. RECENT FINDINGS: In rodents, hypothalamic inflammation and gliosis occur rapidly with high-fat diet consumption prior to significant weight gain. In addition, sensitivity or resistance to diet-induced obesity in rodents generally correlates with the presence or absence of hypothalamic inflammation and reactive gliosis (brain response to injury). Moreover, functional interventions that increase or decrease inflammation in neurons and glia correspondingly alter diet-associated weight gain. However, some conflicting data have recently emerged that question the contribution of hypothalamic inflammation to obesity pathogenesis. Nevertheless, several studies have detected gliosis and disrupted connectivity in obese humans, highlighting the potential translational importance of this mechanism. SUMMARY: There is growing evidence that obesity is associated with brain inflammation in humans, particularly in the hypothalamus where its presence may disrupt body weight control and glucose homeostasis. More work is needed to determine whether this response is common in human obesity and to what extent it can be manipulated for therapeutic benefit.


Subject(s)
Encephalitis/pathology , Gliosis/pathology , Hypothalamus/pathology , Obesity/pathology , Animals , Diet, High-Fat/adverse effects , Encephalitis/etiology , Gliosis/etiology , Glucose/metabolism , Humans
13.
Diabetes ; 64(7): 2376-87, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25633417

ABSTRACT

Several lines of evidence implicate excess glucagon secretion in the elevated rates of hepatic glucose production (HGP), hyperglycemia, and ketosis characteristic of uncontrolled insulin-deficient diabetes (uDM), but whether hyperglucagonemia is required for hyperglycemia in this setting is unknown. To address this question, adult male Wistar rats received either streptozotocin (STZ) to induce uDM (STZ-DM) or vehicle and remained nondiabetic. Four days later, animals received daily subcutaneous injections of either the synthetic GLP-1 receptor agonist liraglutide in a dose-escalating regimen to reverse hyperglucagonemia or its vehicle for 10 days. As expected, plasma glucagon levels were elevated in STZ-DM rats, and although liraglutide treatment lowered glucagon levels to those of nondiabetic controls, it failed to attenuate diabetic hyperglycemia, elevated rates of glucose appearance (Ra), or increased hepatic gluconeogenic gene expression. In contrast, it markedly reduced levels of both plasma ketone bodies and hepatic expression of the rate-limiting enzyme involved in ketone body production. To independently confirm this finding, in a separate study, treatment of STZ-DM rats with a glucagon-neutralizing antibody was sufficient to potently lower plasma ketone bodies but failed to normalize elevated levels of either blood glucose or Ra. These data suggest that in rats with uDM, hyperglucagonemia is required for ketosis but not for increased HGP or hyperglycemia.


Subject(s)
Diabetes Mellitus, Experimental/complications , Glucagon/blood , Glucose/metabolism , Hyperglycemia/etiology , Ketosis/etiology , Liver/metabolism , Animals , Forkhead Box Protein O1 , Forkhead Transcription Factors/physiology , Glucagon/physiology , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Hyperglycemia/blood , Insulin/pharmacology , Ketone Bodies/blood , Ketosis/blood , Liraglutide , Male , Rats, Wistar , Receptors, Glucagon/physiology , Streptozocin
14.
Endocrinology ; 155(8): 3098-111, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24877631

ABSTRACT

Neurotrophins (NTs), once believed to be neural-specific trophic factors, are now known to also provide developmental cues to non-neural cells. In the ovary, NTs contribute to both the formation and development of follicles. Here we show that oocyte-specific deletion of the Ntrk2 gene that encodes the NTRK2 receptor (NTRK2) for neurotrophin-4/5 and brain-derived neurotrophic factor (BDNF) results in post-pubertal oocyte death, loss of follicular organization, and early adulthood infertility. Oocytes lacking NTRK2 do not respond to gonadotropins with activation of phosphatidylinositol 3-kinase (PI3K)-AKT-mediated signaling. Before puberty, oocytes only express a truncated NTRK2 form (NTRK2.T1), but at puberty full-length (NTRK2.FL) receptors are rapidly induced by the preovulatory gonadotropin surge. A cell line expressing both NTRK2.T1 and the kisspeptin receptor (KISS1R) responds to BDNF stimulation with activation of Ntrk2 expression only if kisspeptin is present. This suggests that BDNF and kisspeptin that are produced by granulosa cells (GCs) of periovulatory follicles act in concert to mediate the effect of gonadotropins on Ntrk2 expression in oocytes. In keeping with this finding, the oocytes of NTRK2-intact mice fail to respond to gonadotropins with increased Ntrk2 expression in the absence of KISS1R. Our results demonstrate that the preovulatory gonadotropin surge promotes oocyte survival at the onset of reproductive cyclicity by inducing oocyte expression of NTRK2.FL receptors that set in motion an AKT-mediated survival pathway. They also suggest that gonadotropins activate NTRK2.FL expression via a dual communication pathway involving BDNF and kisspeptin produced in GCs and their respective receptors NTRK2.T1 and KISS1R expressed in oocytes.


Subject(s)
Membrane Glycoproteins/metabolism , Oocytes/metabolism , Ovary/metabolism , Primary Ovarian Insufficiency/etiology , Protein-Tyrosine Kinases/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Female , Gonadotropins/physiology , Infertility, Female/genetics , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Kisspeptin-1
15.
Endocrinology ; 155(8): 3088-97, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24885574

ABSTRACT

Premature ovarian failure (POF) affects 1% of women in reproductive age, but its etiology remains uncertain. Whereas kisspeptins, the products of Kiss1 that act via Kiss1r (aka, Gpr54), are known to operate at the hypothalamus to control GnRH/gonadotropin secretion, additional actions at other reproductive organs, including the ovary, have been proposed. Yet, their physiological relevance is still unclear. We present here a series of studies in Kiss1r haplo-insufficient and null mice suggesting a direct role of kisspeptin signaling in the ovary, the defect of which precipitates a state of primary POF. Kiss1r hypomorph mice displayed a premature decline in ovulatory rate, followed by progressive loss of antral follicles, oocyte loss, and a reduction in all categories of preantral follicles. These alterations were accompanied by reduced fertility. Because of this precocious ovarian ageing, mice more than 48 weeks of age showed atrophic ovaries, lacking growing follicles and corpora lutea. This phenomenon was associated with a drop in ovarian Kiss1r mRNA expression, but took place in the absence of a decrease in circulating gonadotropins. In fact, FSH levels increased in aged hypomorph animals, reflecting loss of follicular function. In turn, Kiss1r-null mice, which do not spontaneously ovulate and have arrested follicular development, failed to show normal ovulatory responses to standard gonadotropin priming and required GnRH prestimulation during 1 week in order to display gonadotropin-induced ovulation. Yet, the magnitude of such ovulatory responses was approximately half of that seen in control immature wild-type animals. Altogether, our data are the first to demonstrate that Kiss1r haplo-insufficiency induces a state of POF, which is not attributable to defective gonadotropin secretion. We also show that the failure of follicular development and ovulation linked to the absence of Kiss1r cannot be fully rescued by (even extended) gonadotropin replacement. These findings suggest a direct ovarian role of kisspeptin signaling, the perturbation of which may contribute to the pathogenesis of POF.


Subject(s)
Kisspeptins/metabolism , Ovary/physiology , Ovulation , Primary Ovarian Insufficiency/etiology , Receptors, G-Protein-Coupled/metabolism , Animals , Female , Gonadotropins/metabolism , Hypogonadism/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, Kisspeptin-1
17.
Endocrinology ; 154(9): 3067-76, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23782941

ABSTRACT

In rodent models of type 1 diabetes, leptin administration into brain ventricles normalizes blood glucose at doses that have no effect when given peripherally. The ventromedial nucleus of the hypothalamus (VMN) is a potential target for leptin's antidiabetic effects because leptin-sensitive neurons in this brain area are implicated in glucose homeostasis. To test this hypothesis, we injected leptin directly into the bilateral VMN of rats with streptozotocin-induced uncontrolled diabetes mellitus. This intervention completely normalized both hyperglycemia and the elevated rates of hepatic glucose production and plasma glucagon levels but had no effect on tissue glucose uptake in the skeletal muscle or brown adipose tissue as measured using tracer dilution techniques during a basal clamp. To determine whether VMN leptin signaling is required for leptin-mediated normalization of diabetic hyperglycemia, we studied mice in which the leptin receptor gene was deleted in VMN steroidogenic factor 1 neurons using cre-loxP technology. Our findings indicate leptin action within these neurons is not required for the correction of diabetic hyperglycemia by central leptin infusion. We conclude that leptin signaling in the VMN is sufficient to mediate leptin's antidiabetic action but may not be necessary for this effect. Leptin action within a distributed neuronal network may mediate its effects on glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Hyperglycemia/prevention & control , Leptin/metabolism , Neurons/metabolism , Receptors, Leptin/metabolism , Signal Transduction , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/pathology , Glucagon/blood , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Gluconeogenesis/drug effects , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Infusions, Intraventricular , Injections, Intraventricular , Leptin/administration & dosage , Leptin/genetics , Leptin/therapeutic use , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Rats , Rats, Wistar , Receptors, Leptin/agonists , Receptors, Leptin/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/pathology
18.
Endocrinology ; 152(12): 5005-16, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22028443

ABSTRACT

Tropomyosin-related kinase (TRK) receptor B (TRKB) mediates the supportive actions of neurotrophin 4/5 and brain-derived neurotrophic factor on early ovarian follicle development. Absence of TRKB receptors reduces granulosa cell (GC) proliferation and delays follicle growth. In the present study, we offer mechanistic insights into this phenomenon. DNA array and quantitative PCR analysis of ovaries from TrkB-null mice revealed that by the end of the first week of postnatal life, Jagged1, Hes1, and Hey2 mRNA abundance is reduced in the absence of TRKB receptors. Although Jagged1 encodes a NOTCH receptor ligand, Hes1 and Hey2 are downstream targets of the JAGGED1-NOTCH2 signaling system. Jagged1 is predominantly expressed in oocytes, and the abundance of JAGGED1 is decreased in TrkB(-/-) oocytes. Lack of TRKB receptors also resulted in reduced expression of c-Myc, a NOTCH target gene that promotes entry into the cell cycle, but did not alter the expression of genes encoding core regulators of cell-cycle progression. Selective restoration of JAGGED1 synthesis in oocytes of TrkB(-/-) ovaries via lentiviral-mediated transfer of the Jagged1 gene under the control of the growth differentiation factor 9 (Gdf9) promoter rescued c-Myc expression, GC proliferation, and follicle growth. These results suggest that neurotrophins acting via TRKB receptors facilitate early follicle growth by supporting a JAGGED1-NOTCH2 oocyte-to-GC communication pathway, which promotes GC proliferation via a c-MYC-dependent mechanism.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Communication/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Growth Factors/physiology , Ovary/growth & development , Receptor, Notch2/metabolism , Receptor, trkB/physiology , Animals , Cell Cycle/genetics , Cell Proliferation , Female , Gene Expression Regulation/physiology , Granulosa Cells/cytology , Jagged-1 Protein , Mice , Mice, Knockout , Nerve Growth Factors/metabolism , Ovarian Follicle/cytology , Receptor, Notch2/genetics , Receptor, trkB/metabolism , Serrate-Jagged Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...