Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
9.
Brain Res ; 1512: 97-105, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23548599

ABSTRACT

Astrocytes respond to multiple forms of central nervous system (CNS) injury by entering a reactive state characterized by morphological changes and a specific pattern of altered protein expression. Termed astrogliosis, this response has been shown to strongly influence the injury response and functional recovery of CNS tissues. This pattern of CNS inflammation and injury associated with astrogliosis has recently been found to occur in the energy homeostasis centers of the hypothalamus during diet-induced obesity (DIO) in rodent models, but the characterization of the astrocyte response remains incomplete. Here, we report that astrocytes in the mediobasal hypothalamus respond robustly and rapidly to purified high-fat diet (HFD) feeding by cleaving caspase-3, a protease whose cleavage is often associated with apoptosis. Although obesity develops in HFD-fed rats by day 14, caspase-3 cleavage occurs by day 3, prior to the development of obesity, suggesting the possibility that it could play a causal role in the hypothalamic neuropathology and fat gain observed in DIO. Caspase-3 cleavage is not associated with an increase in the rate of apoptosis, as determined by TUNEL staining, suggesting it plays a non-apoptotic role analogous to the response to excitotoxic neuron injury. Our results indicate that astrocytes in the mediobasal hypothalamus respond rapidly and robustly to HFD feeding, activating caspase-3 in the absence of apoptosis, a process that has the potential to influence the course of DIO.


Subject(s)
Astrocytes/metabolism , Caspase 3/metabolism , Diet, High-Fat/adverse effects , Hypothalamus/pathology , Obesity/chemically induced , Obesity/pathology , Analysis of Variance , Animals , Apoptosis/physiology , Body Composition/physiology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , In Situ Nick-End Labeling , Male , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar
10.
Am J Physiol Endocrinol Metab ; 304(11): E1245-50, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23548614

ABSTRACT

A hallmark of brain injury from infection, vascular, neurodegenerative, and other disorders is the development of gliosis, which can be detected by magnetic resonance imaging (MRI). In rodent models of diet-induced obesity (DIO), high-fat diet (HFD) consumption rapidly induces inflammation and gliosis in energy-regulating regions of the mediobasal hypothalamus (MBH), and recently we reported MRI findings suggestive of MBH gliosis in obese humans. Thus, noninvasive imaging may obviate the need to assess MBH gliosis using histopathological end points, an obvious limitation to human studies. To investigate whether quantitative MRI is a valid tool with which to measure MBH gliosis, we performed analyses, including measurement of T(2) relaxation time from high-field MR brain imaging of mice fed HFD and chow-fed controls. Mean bilateral T(2) relaxation time was prolonged significantly in the MBH, but not in the thalamus or cortex, of HFD-fed mice compared with chow-fed controls. Histological analysis confirmed evidence of increased astrocytosis and microglial accumulation in the MBH of HFD-fed mice compared with controls, and T(2) relaxation times in the right MBH correlated positively with mean intensity of glial fibrillary acidic protein staining (a marker of astrocytes) in HFD-fed animals. Our findings indicate that T(2) relaxation time obtained from high-field MRI is a useful noninvasive measurement of HFD-induced gliosis in the mouse hypothalamus with potential for translation to human studies.


Subject(s)
Gliosis/pathology , Hypothalamus/pathology , Magnetic Resonance Imaging/methods , Obesity/pathology , Animals , Body Composition/physiology , Diet, High-Fat , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Hypothalamus/metabolism , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism
11.
J Clin Invest ; 122(1): 153-62, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22201683

ABSTRACT

Rodent models of obesity induced by consuming high-fat diet (HFD) are characterized by inflammation both in peripheral tissues and in hypothalamic areas critical for energy homeostasis. Here we report that unlike inflammation in peripheral tissues, which develops as a consequence of obesity, hypothalamic inflammatory signaling was evident in both rats and mice within 1 to 3 days of HFD onset, prior to substantial weight gain. Furthermore, both reactive gliosis and markers suggestive of neuron injury were evident in the hypothalamic arcuate nucleus of rats and mice within the first week of HFD feeding. Although these responses temporarily subsided, suggesting that neuroprotective mechanisms may initially limit the damage, with continued HFD feeding, inflammation and gliosis returned permanently to the mediobasal hypothalamus. Consistent with these data in rodents, we found evidence of increased gliosis in the mediobasal hypothalamus of obese humans, as assessed by MRI. These findings collectively suggest that, in both humans and rodent models, obesity is associated with neuronal injury in a brain area crucial for body weight control.


Subject(s)
Hypothalamus/pathology , Obesity/pathology , Adolescent , Adult , Animals , Base Sequence , Cytokines/genetics , Diet, High-Fat/adverse effects , Female , Gliosis/etiology , Gliosis/pathology , Humans , Hypothalamus/injuries , Hypothalamus/metabolism , Inflammation/etiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , NF-kappa B/metabolism , Neurons/pathology , Obesity/genetics , Obesity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Signal Transduction , Time Factors , Young Adult
12.
Endocrinology ; 151(9): 4109-15, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20573720

ABSTRACT

Obesity induced by high-fat (HF) feeding is associated with low-grade inflammation in peripheral tissues that predisposes to insulin resistance. Recent evidence suggests the occurrence of a similar process in the hypothalamus, which favors weight gain through impairment of leptin and insulin signaling. In addition to its implications for obesity pathogenesis, this hypothesis suggests that centrally targeted antiinflammatory therapies may prove effective in prevention and treatment of this disorder. This article highlights molecular and cellular mechanisms by which hypothalamic inflammation predisposes to diet-induced obesity.


Subject(s)
Hypothalamus/physiopathology , Inflammation/physiopathology , Insulin Resistance , Obesity/physiopathology , Animals , Glucose Intolerance/metabolism , Glucose Intolerance/physiopathology , Humans , Inflammation/metabolism , Leptin/metabolism , Models, Biological , Obesity/metabolism , Weight Gain/physiology
13.
Am J Physiol Endocrinol Metab ; 299(1): E47-53, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20371733

ABSTRACT

In peripheral tissues, the link between obesity and insulin resistance involves low-grade inflammation induced by macrophage activation and proinflammatory cytokine signaling. Since proinflammatory cytokines are also induced in the hypothalamus of animals placed on a high-fat (HF) diet and can inhibit neuronal signal transduction pathways required for normal energy homeostasis, hypothalamic inflammation is hypothesized to contribute to the pathogenesis of diet-induced obesity (DIO). We addressed this hypothesis by perturbing the inflammatory milieu of the hypothalamus in adult male Wistar rats using intracerebroventricular (icv) administration of interleukin-4 (IL-4), a Th2 cytokine that promotes alternative activation (M2) of macrophages and microglia. During HF feeding, icv IL-4 administration increased hypothalamic proinflammatory cytokine gene expression and caused excess weight gain. Intracerebroventricular pretreatment with PS1145, an inhibitor of IKKbeta (a key intracellular mediator of inflammatory signaling), blocked both IL-4 effects, suggesting a causal relationship between IL-4-induced weight gain and hypothalamic inflammation. These observations add to growing evidence linking hypothalamic inflammation to obesity pathogenesis.


Subject(s)
Dietary Fats/administration & dosage , Energy Metabolism/physiology , Hypothalamus/physiopathology , Inflammation/physiopathology , Interleukin-4/pharmacology , Obesity/physiopathology , Animals , Blood Glucose/analysis , Body Weight/physiology , Dietary Fats/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Insulin/blood , Leptin/blood , Macrophage Activation/physiology , Male , Pyridines/pharmacology , Rats , Rats, Wistar , Specific Pathogen-Free Organisms
14.
Front Neuroendocrinol ; 31(1): 79-84, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19822168

ABSTRACT

Determining the effect of hypothalamic inflammatory signals on energy balance presents a paradox. On the one hand, a large body of work has identified inflammatory signaling in the hypothalamus as an essential mediator of the sickness response--the anorexia, cachexia, fever, inactivity, lethargy, anhedonia and adipsia that are triggered by systemic inflammatory stimuli and promote negative energy balance. On the other hand, numerous recent studies implicate inflammatory activation within the hypothalamus as a key factor whereby high-fat diets--and saturated fats in particular--cause central leptin and insulin resistance and thereby promote the defense of elevated body weight. This paradox will likely remain unresolved until several issues have been addressed. Firstly, the hypothalamus--unlike many peripheral inflamed tissues--is an extremely heterogeneous tissue comprised of astrocytes, oligodendrocytes, microglia, endothelial cells, ependymal cells as well as numerous neuronal subgroups. Determining exactly which cells activate defined inflammatory signals in response to a particular stimulus--i.e. sepsis vs. nutrient excess--may yield critical clues. Secondly, for the sake of simplicity many studies evaluate inflammation as an on/off phenomenon. More realistically, inflammatory signaling occurs as a cascade or cycle that changes and progresses over time. Accordingly, even within the same cell type, the low-grade, chronic signal induced by nutrient excess may invoke a different cascade of signals than a strong, acute signal such as sepsis. In addition, because tolerance can develop to certain inflammatory mediators, physiological outcomes may not correlate with early biochemical markers. Lastly, the neuroanatomical location, magnitude, and duration of the inflammatory stimulus can undoubtedly influence the net CNS response. Rigorously evaluating the progression of the inflammatory signaling cascade within specific hypothalamic cell types is a key next step towards resolving the paradox surrounding the effect of inflammatory signaling on energy homeostasis.


Subject(s)
Energy Metabolism/physiology , Homeostasis/physiology , Hypothalamus/physiopathology , Inflammation/physiopathology , Animals , Dietary Fats/administration & dosage , Eating/physiology , Humans , Insulin Resistance , Leptin , Melanocortins , Obesity , Signal Transduction , Weight Gain
15.
Endocrinology ; 150(12): 5362-72, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19819945

ABSTRACT

By activating the Toll-like receptor 4-nuclear factor-kappaB signal transduction pathway, the bacterial endotoxin lipopolysaccharide (LPS) induces anorexia, weight loss, fever, and other components of the sickness response. By comparison, the hormones leptin and insulin cause anorexia without sickness via a central mechanism involving the phosphatidylinositol-3 kinase signaling pathway. In the current study, we investigated whether a common Toll-like receptor 4 and phosphatidylinositol-3 kinase signaling intermediate, atypical protein kinase Czeta/lambda (aPKC), contributes to changes of energy balance induced by these stimuli. Immunohistochemistry analysis revealed that aPKC is expressed in the arcuate and paraventricular nuclei of the hypothalamus, key sites of leptin, insulin, and LPS action. Although administration of LPS, insulin, and leptin each acutely increased hypothalamic aPKC activity at doses that also reduce food intake, LPS treatment caused over 10-fold greater activation of hypothalamic a PKC signaling than that induced by leptin or insulin. Intracerebroventricular pretreatment with an aPKC inhibitor blocked anorexia induced by LPS but not insulin or leptin. Similarly, LPS-induced hypothalamic inflammation (as judged by induction of proinflammatory cytokine gene expression) and neuronal activation in the paraventricular nucleus (as judged by c-fos induction) were reduced by central aPKC inhibition. Although intracerebroventricular aPKC inhibitor administration also abolished LPS-induced fever, it had no effect on sickness-related hypoactivity or weight loss. We conclude that although hypothalamic aPKC signaling is not required for food intake inhibition by insulin or leptin, it plays a key role in inflammatory anorexia and fever induced by LPS.


Subject(s)
Anorexia/enzymology , Hypothalamus/drug effects , Lipopolysaccharides/toxicity , Protein Kinase C/metabolism , Animals , Anorexia/chemically induced , Cell Line , Cytokines/genetics , Eating/drug effects , Fever/chemically induced , Fever/enzymology , Gene Expression/drug effects , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Immunohistochemistry , Inflammation/enzymology , Inflammation/pathology , Insulin/pharmacology , Isoenzymes/metabolism , Leptin/pharmacology , Male , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL