Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
JCI Insight ; 2(8)2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28422749

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), characterized by an excess accumulation of hepatic triglycerides, is a growing health epidemic. While ER stress in the liver has been implicated in the development of NAFLD, the role of brain ER stress - which is emerging as a key contributor to a number of chronic diseases including obesity - in NAFLD remains unclear. These studies reveal that chemical induction of ER stress in the brain caused hepatomegaly and hepatic steatosis in mice. Conversely, pharmacological reductions in brain ER stress in diet-induced obese mice rescued NAFLD independent of body weight, food intake, and adiposity. Evaluation of brain regions involved revealed robust activation of ER stress biomarkers and ER ultrastructural abnormalities in the circumventricular subfornical organ (SFO), a nucleus situated outside of the blood-brain-barrier, in response to high-fat diet. Targeted reductions in SFO-ER stress in obese mice via SFO-specific supplementation of the ER chaperone 78-kDa glucose-regulated protein ameliorated hepatomegaly and hepatic steatosis without altering body weight, food intake, adiposity, or obesity-induced hypertension. Overall, these findings indicate a novel role for brain ER stress, notably within the SFO, in the pathogenesis of NAFLD.

2.
JCI Insight ; 2(2): e87094, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28138551

ABSTRACT

A role for oxidative stress in the brain has been suggested in the pathogenesis of diet-induced obesity (DIO), although the underlying neural regions and mechanisms remain incompletely defined. We tested the hypothesis that NADPH oxidase-dependent oxidative stress in the paraventricular nucleus (PVN), a hypothalamic energy homeostasis center, contributes to the development of DIO. Cre/LoxP technology was coupled with selective PVN adenoviral microinjection to ablate p22phox , the obligatory subunit for NADPH oxidase activity, in mice harboring a conditional p22phox allele. Selective deletion of p22phox in the PVN protected mice from high-fat DIO independent of changes in food intake or locomotor activity. This was accompanied by ß3-adrenoceptor-dependent increases in energy expenditure, elevations in brown adipose tissue thermogenesis, and browning of white adipose tissue. These data reveal a potentially novel role for brain oxidative stress in the development of DIO by modulating ß3-adrenoceptor mechanisms and point to the PVN as an underlying neural site.


Subject(s)
Cytochrome b Group/genetics , Diet, High-Fat , Energy Metabolism/genetics , NADPH Oxidases/genetics , Obesity/genetics , Oxidative Stress , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Adrenergic, beta-3/metabolism , Thermogenesis/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Brain/metabolism , Cytochrome b Group/metabolism , Eating , Hypothalamus/metabolism , Locomotion , Mice , NADPH Oxidases/metabolism , Obesity/metabolism
4.
Mol Metab ; 4(4): 337-43, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25830096

ABSTRACT

OBJECTIVE: Elevations in brain angiotensin-II cause increased energy expenditure and a lean phenotype. Interestingly, the metabolic effects of increased brain angiotensin-II mimic the actions of leptin, suggesting an interaction between the two systems. Here we demonstrate that angiotensin-type 1a receptors (AT1aR) in the subfornical organ (SFO), a forebrain structure emerging as an integrative metabolic center, play a key role in the body weight-reducing effects of leptin via brown adipose tissue (BAT) thermogenesis. METHODS: Cre/LoxP technology coupled with targeted viral delivery to the SFO in a mouse line bearing a conditional allele of the Agtr1a gene was utilized to determine the interaction between leptin and SFO AT1aR in metabolic regulation. RESULTS: Selective deletion of AT1aR in the SFO attenuated leptin-induced weight loss independent of changes in food intake or locomotor activity. This was associated with diminished leptin-induced increases in core body temperature, blunted upregulation of BAT thermogenic markers, and abolishment of leptin-mediated sympathetic activation to BAT. CONCLUSIONS: These data identify a novel interaction between angiotensin-II and leptin in the control of BAT thermogenesis and body weight, and highlight a previously unrecognized role for the forebrain SFO in metabolic regulation.

6.
Am J Physiol Regul Integr Comp Physiol ; 305(6): R566-81, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23883674

ABSTRACT

In addition to effects on appetite and metabolism, leptin influences many neuroendocrine and physiological systems, including the sympathetic nervous system. Building on my Carl Ludwig Lecture of the American Physiological Society, I review the sympathetic and cardiovascular actions of leptin. The review focuses on a critical analysis of the concept of selective leptin resistance (SLR) and the role of leptin in the pathogenesis of obesity-induced hypertension in both experimental animals and humans. We introduced the concept of SLR in 2002 to explain how leptin might increase blood pressure (BP) in obese states, such as diet-induced obesity (DIO), that are accompanied by partial leptin resistance. This concept, analogous to selective insulin resistance in the metabolic syndrome, holds that in several genetic and acquired models of obesity, there is preservation of the renal sympathetic and pressor actions of leptin despite attenuation of the appetite and weight-reducing actions. Two potential overlapping mechanisms of SLR are reviewed: 1) differential leptin molecular signaling pathways that mediate selective as opposed to universal leptin action and 2) brain site-specific leptin action and resistance. Although the phenomenon of SLR in DIO has so far focused on preservation of sympathetic and BP actions of leptin, consideration should be given to the possibility that this concept may extend to preservation of other actions of leptin. Finally, I review perplexing data on the effects of leptin on sympathetic activity and BP in humans and its role in human obesity-induced hypertension.


Subject(s)
Brain/metabolism , Hypertension/metabolism , Leptin/metabolism , Myocardium/metabolism , Obesity/metabolism , Sympathetic Nervous System/metabolism , Animals , Humans , Hypertension/etiology , Obesity/complications
7.
8.
Hypertension ; 61(3): 737-44, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23357182

ABSTRACT

The adipocyte-derived hormone leptin acts within the central nervous system to decrease food intake and body weight and to increase renal and thermogenic brown adipose tissue sympathetic nerve activity (SNA). Previous studies have focused on hypothalamic brain regions, although recent findings have identified leptin receptors (ObR) in a distributed brain network, including the circumventricular subfornical organ (SFO), a forebrain region devoid of a blood-brain barrier. We tested the hypothesis that ObR in the SFO are functionally linked to leptin-induced decreases in food intake and body weight and increases in SNA. SFO-targeted microinjections of an adenovirus encoding Cre-recombinase in ObR(flox/flox) mice resulted in selective ablation of ObR in the SFO. Interestingly, deletion of ObR in the SFO did not influence the decreases in either food intake or body weight in response to daily systemic or cerebroventricular administration of leptin. In line with these findings, reduction in SFO ObR did not attenuate leptin-mediated increases in thermogenic brown adipose tissue SNA. In contrast, increases in renal SNA induced by systemic or cerebroventricular administration of leptin were abolished in mice with SFO-targeted deletion of ObR. These results demonstrate that ObR in the SFO play an important role in leptin-induced renal sympathoexcitation, but not in the body weight, food intake, or brown adipose tissue SNA thermogenic effects of leptin. These findings highlight the concept of a distributed brain network of leptin action and illustrate that brain regions, including the SFO, can mediate distinct cardiovascular and metabolic responses to leptin.


Subject(s)
Kidney/drug effects , Leptin/administration & dosage , Subfornical Organ/drug effects , Sympathetic Nervous System/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Animals , Body Weight/drug effects , Brain/drug effects , Eating/drug effects , Gene Silencing , Injections, Intraventricular , Kidney/innervation , Male , Mice , Receptors, Leptin/genetics , Thermogenesis/drug effects
9.
Hypertension ; 61(3): 716-22, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23266541

ABSTRACT

Although elevated renin-angiotensin system activity and angiotensinergic signaling within the brain are required for hypertension, polydipsia, and increased metabolic rate induced by deoxycorticosterone acetate (DOCA)-salt, the contribution of specific receptor subtypes and brain nuclei mediating these responses remains poorly defined. We hypothesized that angiotensin type 1a receptors (AT(1a)R) within the subfornical organ (SFO) mediate these responses. Transgenic mice carrying a conditional allele of the endogenous AT(1a)R (AT(1a)R(flox)) were administered an adenovirus encoding Cre-recombinase and enhanced green fluorescent protein (eGFP) or adenovirus encoding eGFP alone into the lateral cerebral ventricle. Adenovirus encoding Cre-recombinase reduced AT(1a)R mRNA and induced recombination in AT(1a)R(flox) genomic DNA specifically in the SFO, without significant effect in the paraventricular or arcuate nuclei, and also induced SFO-specific recombination in ROSA(TdTomato) reporter mice. The effect of SFO-targeted ablation of endogenous AT(1a)R was evaluated in AT(1a)R(flox) mice at 3 time points: (1) baseline, (2) 1 week after virus injection but before DOCA-salt, and (3) after 3 weeks of DOCA-salt. DOCA-salt-treated mice with deletion of AT(1a)R in SFO exhibited a blunted increase in arterial pressure. Increased sympathetic cardiac modulation and urine copeptin, a marker of vasopressin release, were both significantly reduced in DOCA-salt mice when AT(1a)R was deleted in the SFO. Additionally, deletion of AT(1a)R in the SFO significantly attenuated the polydipsia, polyuria, and sodium intake in response to DOCA-salt. Together, these data highlight the contribution of AT(1a)R in the SFO to arterial pressure regulation potentially through changes on sympathetic cardiac modulation, vasopressin release, and hydromineral balance in the DOCA-salt model of hypertension.


Subject(s)
Desoxycorticosterone/adverse effects , Hypertension/chemically induced , Mineralocorticoids/adverse effects , Receptor, Angiotensin, Type 1/physiology , Subfornical Organ/drug effects , Subfornical Organ/physiopathology , Animals , Arterial Pressure/drug effects , Biomarkers/urine , Glycopeptides/urine , Heart/drug effects , Heart/innervation , Male , Mice , Mice, Transgenic , Polydipsia/chemically induced , Polyuria/chemically induced , Receptor, Angiotensin, Type 1/genetics , Recombination, Genetic , Sodium/metabolism , Sympathetic Nervous System/drug effects
10.
J Clin Invest ; 122(11): 3960-4, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064361

ABSTRACT

Although endoplasmic reticulum (ER) stress is a pathologic mechanism in a variety of chronic diseases, it is unclear what role it plays in chronic hypertension (HTN). Dysregulation of brain mechanisms controlling arterial pressure is strongly implicated in HTN, particularly in models involving angiotensin II (Ang II). We tested the hypothesis that ER stress in the brain is causally linked to Ang II-dependent HTN. Chronic systemic infusion of low-dose Ang II in C57BL/6 mice induced slowly developing HTN, which was abolished by co-infusion of the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) into the lateral cerebroventricle. Investigations of the brain regions involved revealed robust increases in ER stress biomarkers and profound ER morphological abnormalities in the circumventricular subfornical organ (SFO), a region outside the blood-brain barrier and replete with Ang II receptors. Ang II-induced HTN could be prevented in this model by selective genetic supplementation of the ER chaperone 78-kDa glucose-regulated protein (GRP78) in the SFO. These data demonstrate that Ang II-dependent HTN is mediated by ER stress in the brain, particularly the SFO. To our knowledge, this is the first report that ER stress, notably brain ER stress, plays a key role in chronic HTN. Taken together, these findings may have broad implications for the pathophysiology of this disease.


Subject(s)
Angiotensin II/adverse effects , Brain/metabolism , Endoplasmic Reticulum Stress , Hypertension/metabolism , Vasoconstrictor Agents/adverse effects , Angiotensin II/pharmacology , Animals , Brain/pathology , Cholagogues and Choleretics/pharmacology , Chronic Disease , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Hypertension/chemically induced , Hypertension/pathology , Mice , Subfornical Organ/metabolism , Subfornical Organ/pathology , Taurochenodeoxycholic Acid/pharmacology , Vasoconstrictor Agents/pharmacology
11.
Am J Physiol Heart Circ Physiol ; 303(2): H197-206, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22610169

ABSTRACT

The sympathetic nervous system, leptin, and renin-angiotensin system (RAS) have been implicated in obesity-associated hypertension. There is increasing evidence for the presence of both leptin and angiotensin II receptors in several key brain cardiovascular and metabolic control regions. We tested the hypothesis that the brain RAS plays a facilitatory role in the sympathetic nerve responses to leptin. In rats, intracerebroventricular (ICV) administration of losartan (5 µg) selectively inhibited increases in renal and brown adipose tissue (BAT) sympathetic nerve activity (SNA) produced by leptin (10 µg ICV) but did not reduce the SNA responses to corticotrophin-releasing factor (CRF) or the melanocortin receptor agonist MTII. In mice with deletion of angiotensin II type-1a receptors (AT(1a)R(-/-)), increases in renal and BAT SNA induced by leptin (2 µg ICV) were impaired whereas SNA responses to MTII were preserved. Decreases in food intake and body weight with ICV leptin did not differ in AT(1a)R(-/-) vs. AT(1a)R(+/+) mice. ICV leptin in rats increased AT(1a)R and angiotensin-converting enzyme (ACE) mRNA in the subfornical organ and AT(1a)R mRNA in the arcuate nucleus, suggesting leptin-induced upregulation of the brain RAS in specific brain regions. To evaluate the role of de novo production of brain angiotensin II in SNA responses to leptin, we treated rats with captopril (12.5 µg ICV). Captopril attenuated leptin effects on renal and BAT SNA. In conclusion, these studies provide evidence that the brain RAS selectively facilitates renal and BAT sympathetic nerve responses to leptin while sparing effects on food intake.


Subject(s)
Brain/physiology , Leptin/physiology , Renin-Angiotensin System/physiology , Sympathetic Nervous System/physiology , Adipose Tissue/drug effects , Adipose Tissue/innervation , Angiotensin II/biosynthesis , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Body Weight/drug effects , Body Weight/physiology , Captopril/pharmacology , Corticotropin-Releasing Hormone/pharmacology , Eating/drug effects , Eating/psychology , Gene Deletion , Losartan/pharmacology , Male , Mice , Mice, Inbred C57BL , Peptidyl-Dipeptidase A/biosynthesis , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/physiology , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
12.
PLoS One ; 6(8): e23364, 2011.
Article in English | MEDLINE | ID: mdl-21853117

ABSTRACT

Obese, leptin deficient obob mice have profoundly decreased activity and increased food seeking behavior. The decreased activity has been attributed to obesity. In mice, we tested the hypothesis that leptin increases total locomotor activity but inhibits food anticipatory activity. We also sought to determine if leptin induced increases in total locomotor activity are independent of changes in body weight and obesity. We studied obob mice and also created a novel transgenic mouse where leptin is over-expressed in a tetracycline-off system and can be abruptly and non-invasively suppressed by doxycycline within few hours. The studies were performed using two independent behavioral assays: home cage activity (HCA) and running wheel activity (RWA). Systemic administration of leptin (150 ng/hr) to obob mice produced a 122%±30% (mean ± SEM) increase (p≤0.01) in locomotor activity within 2 days In addition, cerebroventricular administration of leptin (5 ng/hr) also produced an early and progressive increase in total locomotor activity beginning on the 1st day (+28±8%; p≤0.05) and increasing to +69±23% on day 3 without a decrease in body weight during this time. The increase in activity was restricted to the dark phase. Conversely, in a tet-off transgenic obob mouse line, acute leptin suppression reduced spontaneous locomotor activity. To further define activities that are leptin regulated, we assayed food anticipatory activity (FAA) and found that it was markedly augmented in obob mice compared to wild type mice (+38±6.7 in obob vs +20±6.3% in wild type at peak; mean ± SEM; p≤0.001) and abolished by leptin. Although melanocortin-3 receptors (MC3R) reportedly mediate FAA, we found augmented FAA and preserved inhibitory effects of leptin on FAA in MC3R-/-obob mice. In summary, this study demonstrates that total activity and FAA are regulated independently by leptin. Leptin, acting in the central nervous system and at physiologic levels, produces early increases in locomotor activity before substantial weight loss. In contrast, leptin suppresses augmented food anticipatory activity in obob mice.


Subject(s)
Anticipation, Psychological/drug effects , Feeding Behavior/drug effects , Leptin/pharmacology , Motor Activity/drug effects , Animals , Injections, Intraventricular , Leptin/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Nervous System/drug effects , Physical Conditioning, Animal , Receptor, Melanocortin, Type 3/metabolism , Tetracycline/pharmacology , Time Factors , Weight Loss/drug effects
13.
Circ Res ; 108(7): 808-12, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21311043

ABSTRACT

RATIONALE: The hypothalamic arcuate nucleus (ARC) is considered a major site for leptin signaling that regulates several physiological processes. OBJECTIVE: To test the hypothesis that leptin receptor in the ARC is required to mediate leptin-induced sympathetic activation. METHODS AND RESULTS: First, we used the ROSA Cre-reporter mice to establish the feasibility of driving Cre expression in the ARC in a controlled manner with bilateral microinjection of adenovirus-expressing Cre-recombinase (Ad-Cre). Ad-Cre microinjection into the ARC of ObR(flox/flox) mice robustly reduced ObR expression and leptin-induced Stat3 activation in the ARC but not in the adjacent nuclei, confirming the efficacy and selectivity of the ARC deletion of ObR. Critically, deletion of ObR in the ARC attenuated brown adipose tissue and renal sympathetic nerve responses to leptin. We also examined whether ObR in the ARC is required for the preserved leptin-induced increase in renal sympathetic activity in dietary obesity. We found that deletion of ARC ObR abrogated leptin-induced increases in renal sympathetic discharge and resolved arterial pressure elevation in diet-induced obese ObR(flox/flox) mice. CONCLUSIONS: These data demonstrate a critical role for ObR in the ARC in mediating the sympathetic nerve responses to leptin and in the adverse sympathoexcitatory effects of leptin in obesity.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Gene Deletion , Leptin/pharmacology , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Sympathetic Nervous System/drug effects , Adenoviridae/genetics , Animals , Disease Models, Animal , Green Fluorescent Proteins , Homozygote , Integrases/genetics , Integrases/metabolism , Mice , Mice, Inbred C57BL , Obesity/physiopathology , Sympathetic Nervous System/physiopathology
14.
Hypertension ; 57(3): 627-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21263121

ABSTRACT

Leptin acts in the brain to regulate food intake and energy expenditure. Leptin also increases renal sympathetic nerve activity and arterial pressure. The divergent signaling capacities of the leptin receptor (ObRb) mediate the stimulation of various intracellular pathways that are important for leptin control of physiological processes. We evaluated the cardiovascular and sympathetic consequences of disrupting the signal emanating from tyrosine985 of ObRb. For this, we used Lepr(L985) (l/l) mice, which carry a loss of function mutation replacing tyrosine985 of ObRb with leucine. Body weight of l/l mice was not significantly different from wild-type controls. In contrast, radiotelemetry measurements revealed that the l/l mice had higher arterial pressure and heart rate as compared with controls. Ganglionic blockade caused a greater arterial pressure fall in the l/l mice relative to controls. In addition, leptin treatment induced a larger increase in arterial pressure and heart rate in the l/l versus wild-type mice. Finally, we compared the response of renal and brown adipose tissue sympathetic nerve activity to intracerebroventricular injection of leptin (2 µg) between l/l and control mice. Leptin-induced increase in renal sympathetic nerve activity was greater in l/l mice relative to controls. In contrast, the brown adipose tissue sympathetic nerve activity response to leptin was attenuated in the l/l mice relative to controls. These data indicate that selective loss of leptin receptor signaling emanating from tyrosine985 enhances the cardiovascular and renal sympathetic effects of leptin. These findings provide important insight into the molecular mechanisms underlying leptin's effects on the sympathetic cardiovascular function and arterial pressure.


Subject(s)
Blood Pressure/genetics , Heart Rate/genetics , Mutation , Receptors, Leptin/genetics , Sympathetic Nervous System/physiology , Adipose Tissue, Brown/innervation , Adipose Tissue, Brown/metabolism , Amino Acid Substitution , Analysis of Variance , Animals , Blood Pressure/drug effects , Heart Rate/drug effects , Kidney/innervation , Kidney/metabolism , Leptin/metabolism , Leptin/pharmacology , Mice , Mice, Transgenic , Receptors, Leptin/metabolism , Sympathetic Nervous System/drug effects
15.
Cell Metab ; 12(5): 431-42, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21035755

ABSTRACT

The renin-angiotensin system (RAS), in addition to its endocrine functions, plays a role within individual tissues such as the brain. The brain RAS is thought to control blood pressure through effects on fluid intake, vasopressin release, and sympathetic nerve activity (SNA), and may regulate metabolism through mechanisms which remain undefined. We used a double-transgenic mouse model that exhibits brain-specific RAS activity to examine mechanisms contributing to fluid and energy homeostasis. The mice exhibit high fluid turnover through increased adrenal steroids, which is corrected by adrenalectomy and attenuated by mineralocorticoid receptor blockade. They are also hyperphagic but lean because of a marked increase in body temperature and metabolic rate, mediated by increased SNA and suppression of the circulating RAS. ß-adrenergic blockade or restoration of circulating angiotensin-II, but not adrenalectomy, normalized metabolic rate. Our data point to contrasting mechanisms by which the brain RAS regulates fluid intake and energy expenditure.


Subject(s)
Adrenal Glands/metabolism , Angiotensinogen/metabolism , Brain/metabolism , Energy Metabolism , Renin-Angiotensin System , Renin/metabolism , Synapsins/metabolism , Angiotensin II/metabolism , Angiotensinogen/genetics , Animals , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Polyuria/etiology , Polyuria/genetics , Polyuria/metabolism , Promoter Regions, Genetic , Renin/genetics , Steroids/metabolism , Sympathetic Nervous System/physiology , Synapsins/genetics , Thermogenesis
17.
Diabetes ; 58(3): 536-42, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19066310

ABSTRACT

OBJECTIVE: Leptin is an adipocyte hormone that plays a major role in energy balance. Leptin receptors in the hypothalamus are known to signal via distinct mechanisms, including signal transducer and activator of transcription-3 (STAT3) and phosphoinositol-3 kinase (PI 3-kinase). Here, we tested the hypothesis that extracellular signal-regulated kinase (ERK) is mediating leptin action in the hypothalamus. RESEARCH DESIGN AND METHODS: Biochemical, pharmacological, and physiological approaches were combined to characterize leptin activation of ERK in the hypothalamus in rats. RESULTS: Leptin activates ERK1/2 in a receptor-mediated manner that involves JAK2. Leptin-induced ERK1/2 activation was restricted to the hypothalamic arcuate nucleus. Pharmacological blockade of hypothalamic ERK1/2 reverses the anorectic and weight-reducing effects of leptin. The pharmacological antagonists of ERK1/2 did not attenuate leptin-induced activation of STAT3 or PI 3-kinase. Blockade of ERK1/2 abolishes leptin-induced increases in sympathetic nerve traffic to thermogenic brown adipose tissue (BAT) but does not alter the stimulatory effects of leptin on sympathetic nerve activity to kidney, hindlimb, or adrenal gland. In contrast, blockade of PI 3-kinase prevents leptin-induced sympathetic activation to kidney but not to BAT, hindlimb, or adrenal gland. CONCLUSIONS: Our findings indicate that hypothalamic ERK plays a key role in the control of food intake, body weight, and thermogenic sympathetic outflow by leptin but does not participate in the cardiovascular and renal sympathetic actions of leptin.


Subject(s)
Adipose Tissue, Brown/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypothalamus/physiology , Leptin/pharmacology , Receptors, Leptin/physiology , Adipose Tissue, Brown/drug effects , Animals , Body Weight/drug effects , Energy Intake , Hypothalamus/drug effects , Injections, Intraperitoneal , Injections, Intraventricular , Leptin/administration & dosage , Male , Mice , Neurons/drug effects , Neurons/physiology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiology , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptors, Leptin/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology
18.
Hypertension ; 53(2): 375-80, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19103999

ABSTRACT

The hypothalamic arcuate nucleus was initially regarded as the principal site of leptin action, but there is increasing evidence for functional leptin receptors in extrahypothalamic sites, including the nucleus tractus solitarii (NTS). We demonstrated previously that arcuate injection of leptin increases sympathetic nerve activity (SNA) to brown adipose tissue and kidney. In this study, we tested the hypothesis that leptin signaling in the NTS affects sympathetic neural outflow. Using a stereotaxic device in anesthetized rats, we microinjected leptin (0.25 to 1.00 microg) or saline into the NTS while recording SNA to kidney and brown adipose tissue. Microinjection of leptin into the commissural and medial subnuclei of the caudal NTS at the level of the area postrema in Sprague-Dawley rats produced a dose-related increase in renal SNA (+112+/-15% with leptin 1 microg; n=7; P<0.001) but did not increase SNA to brown adipose tissue (-15+/-12%; P value not significant). This effect depended on intact functional leptin receptors, because it was not observed in Zucker obese rats that have a missense mutation in the leptin receptor. Rostral NTS injection of leptin failed to increase SNA, indicating that leptin signaling in the NTS is probably confined to the caudal NTS at the level of the area postrema. In summary, this study demonstrates that leptin signaling in the caudal NTS increases SNA to the kidney but not to the brown adipose tissue. The study strengthens the concept of a distributed brain network of leptin action and demonstrates that these distributed brain sites can mediate contrasting sympathetic responses to leptin.


Subject(s)
Kidney/innervation , Leptin/physiology , Signal Transduction/physiology , Solitary Nucleus/metabolism , Sympathetic Nervous System/physiology , Adipose Tissue, Brown/innervation , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Kidney/physiology , Leptin/administration & dosage , Microinjections , Mutation, Missense , Obesity/metabolism , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Solitary Nucleus/drug effects
20.
Curr Hypertens Rep ; 9(5): 368-72, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18177582

ABSTRACT

Weight reduction is generally recommended as the first line of treatment for the increasing problem of obesity-associated hypertension. At first glance, this recommendation seems compelling, but evidence suggests that weight loss for obesity-associated hypertension is neither simple nor consistently effective as antihypertensive therapy. First, dietary and behavioral therapy is accompanied by an extremely high rate of weight regain after loss. Mounting evidence shows that this recidivism reflects neurobiologic and not simply psychologic adaptations to dietary restriction. Second, chronic blood pressure-lowering effects of weight loss produced by diet, weight-reducing drugs, or bariatric surgery may not be as pronounced as commonly thought. Third, there is evidence that dietary restriction, independent of weight loss, reduces sympathetic nervous system activity and might thereby contribute to reducing blood pressure. This phenomenon deserves more consideration in designing and interpreting studies of blood pressure changes during diet-induced weight loss. This article reviews these issues and highlights the nuances and challenges in the effectiveness of weight loss for treatment of obesity-induced hypertension.


Subject(s)
Hypertension/therapy , Obesity/therapy , Weight Loss/physiology , Appetite Stimulants/therapeutic use , Bariatric Surgery , Behavior Therapy , Diet Therapy , Humans , Hypertension/etiology , Hypertension/physiopathology , Obesity/complications , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL
...