Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 14: 1238120, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37885804

RESUMO

Metabolic disorders such as obesity and type 2 diabetes (T2D) are increasingly prevalent worldwide, necessitating a deeper comprehension of their underlying mechanisms. However, translating findings from animal research to human patients remains challenging. This study aimed to investigate the long-term effects of Streptozotocin (STZ) on metabolic, cardiac, and somatosensory function in mice fed a Western diet (WD) of high fat, sucrose, and cholesterol with low doses of STZ administration compared to mice fed WD alone. In our research, we thoroughly characterized energy balance and glucose homeostasis, as well as allodynia and cardiac function, all of which have been previously shown to be altered by WD feeding. Notably, our findings revealed that the treatment of WD-fed mice with STZ exacerbated dysfunction in glucose homeostasis via reduced insulin secretion in addition to impaired peripheral insulin signaling. Furthermore, both WD and WD + STZ mice exhibited the same degree of cardiac autonomic neuropathy, such as reduced heart rate variability and decreased protein levels of cardiac autonomic markers. Furthermore, both groups developed the same symptoms of neuropathic pain, accompanied by elevated levels of activating transcription factor 3 (Atf3) in the dorsal root ganglia. These discoveries enhance our understanding of metabolic activity, insulin resistance, neuropathy, and cardiac dysfunction of diet-induced models of obesity and diabetes. The exacerbation of impaired insulin signaling pathways by STZ did not lead to or worsen cardiac and somatosensory dysfunction. Additionally, they offer valuable insights into suitable diet induced translational mouse models, thereby advancing the development of potential interventions for associated conditions.

2.
Front Cardiovasc Med ; 10: 1105581, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36844730

RESUMO

More than 50% of patients with heart failure present with heart failure with preserved ejection fraction (HFpEF), and 80% of them are overweight or obese. In this study we developed an obesity associated pre-HFpEF mouse model and showed an improvement in both systolic and diastolic early dysfunction following fecal microbiome transplant (FMT). Our study suggests that the gut microbiome-derived short-chain fatty acid butyrate plays a significant role in this improvement. Cardiac RNAseq analysis showed butyrate to significantly upregulate ppm1k gene that encodes protein phosphatase 2Cm (PP2Cm) which dephosphorylates and activates branched-chain α-keto acid dehydrogenase (BCKDH) enzyme, and in turn increases the catabolism of branched chain amino acids (BCAAs). Following both FMT and butyrate treatment, the level of inactive p-BCKDH in the heart was reduced. These findings show that gut microbiome modulation can alleviate early cardiac mechanics dysfunction seen in the development of obesity associated HFpEF.

3.
Sci Rep ; 12(1): 10754, 2022 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-35750708

RESUMO

The prevalence of peripheral neuropathy is high in diabetic and overweight populations. Chronic neuropathic pain, a symptom of peripheral neuropathy, is a major disabling symptom that leads to a poor quality of life. Glucose management for diabetic and prediabetic individuals often fail to reduce or improve pain symptoms, therefore, exploring other mechanisms is necessary to identify effective treatments. A large body of evidence suggest that lipid signaling may be a viable target for management of peripheral neuropathy in obese individuals. The nuclear transcription factors, Liver X Receptors (LXR), are known regulators of lipid homeostasis, phospholipid remodeling, and inflammation. Notably, the activation of LXR using the synthetic agonist GW3965, delayed western diet (WD)-induced allodynia in rodents. To further understand the neurobiology underlying the effect of LXR, we used translating ribosome affinity purification and evaluated translatomic changes in the sensory neurons of WD-fed mice treated with the LXR agonist GW3965. We also observed that GW3965 decreased prostaglandin levels and decreased free fatty acid content, while increasing lysophosphatidylcholine, phosphatidylcholine, and cholesterol ester species in the sensory neurons of the dorsal root ganglia (DRG). These data suggest novel downstream interplaying mechanisms that modifies DRG neuronal lipid following GW3965 treatment.


Assuntos
Dieta Ocidental , Gânglios Espinais , Animais , Benzilaminas/farmacologia , Dieta Ocidental/efeitos adversos , Homeostase , Receptores X do Fígado/agonistas , Camundongos , Prostaglandina D2 , Qualidade de Vida
4.
J Neuroinflammation ; 19(1): 57, 2022 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-35219337

RESUMO

BACKGROUND: Peripheral neuropathy is a common and progressive disorder in the elderly that interferes with daily activities. It is of importance to find efficient treatments to treat or delay this age-related neurodegeneration. Silencing macrophages by reducing foamy macrophages showed significant improvement of age-related degenerative changes in peripheral nerves of aged mice. We previously demonstrated that activation of the cholesterol sensor Liver X receptor (LXR) with the potent agonist, GW3965, alleviates pain in a diet-induced obesity model. We sought to test whether LXR activation may improve neuropathy in aged mice. METHODS: 21-month-old mice were treated with GW3965 (25 mg/Kg body weight) for 3 months while testing for mechanical allodynia and thermal hyperalgesia. At termination, flow cytometry was used to profile dorsal root ganglia and sciatic nerve cells. Immune cells were sorted and analyzed for cholesterol and gene expression. Nerve fibers of the skin from the paws were analyzed. Some human sural nerves were also evaluated. Comparisons were made using either t test or one-way ANOVA. RESULTS: Treatment with GW3965 prevented the development of mechanical hypersensitivity and thermal hyperalgesia over time in aged mice. We also observed change in polarization and cholesterol content of sciatic nerve macrophages accompanied by a significant increase in nerve fibers of the skin. CONCLUSIONS: These results suggest that activation of the LXR may delay the PNS aging by modifying nerve-immune cell lipid content. Our study provides new potential targets to treat or delay neuropathy during aging.


Assuntos
Doenças do Sistema Nervoso Periférico , Animais , Gânglios Espinais/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Receptores X do Fígado/agonistas , Camundongos , Doenças do Sistema Nervoso Periférico/metabolismo , Nervo Isquiático/metabolismo
5.
Neuroendocrinology ; 112(4): 324-337, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34034255

RESUMO

INTRODUCTION: Mitochondria are essential organelles required for several cellular processes ranging from ATP production to cell maintenance. To provide energy, mitochondria are transported to specific cellular areas in need. Mitochondria also need to be recycled. These mechanisms rely heavily on trafficking events. While mechanisms are still unclear, hypothalamic mitochondria are linked to obesity. METHODS: We used C2 domain protein 5 (C2CD5, also called C2 domain-containing phosphoprotein [CDP138]) whole-body KO mice primary neuronal cultures and cell lines to perform electron microscopy, live cellular imaging, and oxygen consumption assay to better characterize mitochondrial alteration linked to C2CD5. RESULTS: This study identified that C2CD5 is necessary for proper mitochondrial trafficking, structure, and function in the hypothalamic neurons. We previously reported that mice lacking C2CD5 were obese and displayed reduced functional G-coupled receptor, melanocortin receptor 4 (MC4R) at the surface of hypothalamic neurons. Our data suggest that in neurons, normal MC4R endocytosis/trafficking necessities functional mitochondria. DISCUSSION: Our data show that C2CD5 is a new protein necessary for normal mitochondrial function in the hypothalamus. Its loss alters mitochondrial ultrastructure, localization, and activity within the hypothalamic neurons. C2CD5 may represent a new protein linking hypothalamic dysfunction, mitochondria, and obesity.


Assuntos
Domínios C2 , Hipotálamo , Animais , Hipotálamo/metabolismo , Camundongos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo
6.
Mol Metab ; 54: 101350, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34626852

RESUMO

OBJECTIVE: The vagus nerve provides a direct line of communication between the gut and the brain for proper regulation of energy balance and glucose homeostasis. Short-chain fatty acids (SCFAs) produced via gut microbiota fermentation of dietary fiber have been proposed to regulate host metabolism and feeding behavior via the vagus nerve, but the molecular mechanisms have not yet been elucidated. We sought to identify the G-protein-coupled receptors within vagal neurons that mediate the physiological and therapeutic benefits of SCFAs. METHODS: SCFA, particularly propionate, signaling occurs via free fatty acid receptor 3 (FFAR3), that we found expressed in vagal sensory neurons innervating throughout the gut. The lack of cell-specific animal models has impeded our understanding of gut/brain communication; therefore, we generated a mouse model for cre-recombinase-driven deletion of Ffar3. We comprehensively characterized the feeding behavior of control and vagal-FFAR3 knockout (KO) mice in response to various conditions including fasting/refeeding, western diet (WD) feeding, and propionate supplementation. We also utilized ex vivo organotypic vagal cultures to investigate the signaling pathways downstream of propionate FFAR3 activation. RESULTS: Vagal-FFAR3KO led to increased meal size in males and females, and increased food intake during fasting/refeeding and WD challenges. In addition, the anorectic effect of propionate supplementation was lost in vagal-FFAR3KO mice. Sequencing approaches combining ex vivo and in vivo experiments revealed that the cross-talk of FFAR3 signaling with cholecystokinin (CCK) and leptin receptor pathways leads to alterations in food intake. CONCLUSION: Altogether, our data demonstrate that FFAR3 expressed in vagal neurons regulates feeding behavior and mediates propionate-induced decrease in food intake.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Nervo Vago/metabolismo , Animais , Comportamento Alimentar , Microbioma Gastrointestinal , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/genética
7.
Proc Natl Acad Sci U S A ; 117(42): 26482-26493, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33020290

RESUMO

Obesity affects over 2 billion people worldwide and is accompanied by peripheral neuropathy (PN) and an associated poorer quality of life. Despite high prevalence, the molecular mechanisms underlying the painful manifestations of PN are poorly understood, and therapies are restricted to use of painkillers or other drugs that do not address the underlying disease. Studies have demonstrated that the gut microbiome is linked to metabolic health and its alteration is associated with many diseases, including obesity. Pathologic changes to the gut microbiome have recently been linked to somatosensory pain, but any relationships between gut microbiome and PN in obesity have yet to be explored. Our data show that mice fed a Western diet developed indices of PN that were attenuated by concurrent fecal microbiome transplantation (FMT). In addition, we observed changes in expression of genes involved in lipid metabolism and calcium handling in cells of the peripheral nerve system (PNS). FMT also induced changes in the immune cell populations of the PNS. There was a correlation between an increase in the circulating short-chain fatty acid butyrate and pain improvement following FMT. Additionally, butyrate modulated gene expression and immune cells in the PNS. Circulating butyrate was also negatively correlated with distal pain in 29 participants with varied body mass index. Our data suggest that the metabolite butyrate, secreted by the gut microbiome, underlies some of the effects of FMT. Targeting the gut microbiome, butyrate, and its consequences may represent novel viable approaches to prevent or relieve obesity-associated neuropathies.


Assuntos
Transplante de Microbiota Fecal/métodos , Obesidade/microbiologia , Doenças do Sistema Nervoso Periférico/terapia , Animais , Butiratos/metabolismo , Dieta Hiperlipídica , Dieta Ocidental , Ácidos Graxos Voláteis/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Expressão Gênica , Resistência à Insulina , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Microbiota , Neuralgia/metabolismo , Obesidade/fisiopatologia , Sistema Nervoso Periférico/metabolismo , Sistema Nervoso Periférico/fisiologia
8.
Sci Rep ; 10(1): 6396, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286429

RESUMO

Neuropathic pain caused by peripheral nerve injuries significantly affects sensory perception and quality of life. Accumulating evidence strongly link cholesterol with development and progression of Obesity and Diabetes associated-neuropathies. However, the exact mechanisms of how cholesterol/lipid metabolism in peripheral nervous system (PNS) contributes to the pathogenesis of neuropathy remains poorly understood. Dysregulation of LXR pathways have been identified in many neuropathic models. The cholesterol sensor, LXR α/ß, expressed in sensory neurons are necessary for proper peripheral nerve function. Deletion of LXR α/ß from sensory neurons lead to pain-like behaviors. In this study, we identified that LXR α/ß expressed in sensory neurons regulates neuronal Neuregulin 1 (Nrg1), protein involved in cell-cell communication. Using in vivo cell-specific approaches, we observed that loss of LXR from sensory neurons altered genes in non-neuronal cells located in the sciatic nerve (potentially representing Schwann cells (SC)). Our data suggest that neuronal LXRs may regulate non-neuronal cell function via a Nrg1-dependent mechanism. The decrease in Nrg1 expression in DRG neurons of WD-fed mice may suggest an altered Nrg1-dependent neuron-SC communication in Obesity. The communication between neurons and non-neuronal cells such as SC could be a new biological pathway to study and understand the molecular and cellular mechanism underlying Obesity-associated neuropathy and PNS dysfunction.


Assuntos
Dieta Ocidental , Receptores X do Fígado/metabolismo , Neuregulina-1/genética , Nervo Isquiático/metabolismo , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais , Animais , Orientação de Axônios , Receptores ErbB/metabolismo , Gânglios Espinais/metabolismo , Regulação da Expressão Gênica , Camundongos , Neuregulina-1/metabolismo , Células de Schwann/metabolismo , Transcrição Gênica
9.
J Exp Biol ; 223(Pt 8)2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32165434

RESUMO

Non-shivering thermogenesis can promote negative energy balance and weight loss. In this study, we identified a contextual stimulus that induces rapid and robust thermogenesis in skeletal muscle. Rats exposed to the odor of a natural predator (ferret) showed elevated skeletal muscle temperatures detectable as quickly as 2 min after exposure, reaching maximum thermogenesis of >1.5°C at 10-15 min. Mice exhibited a similar thermogenic response to the same odor. Ferret odor induced a significantly larger and qualitatively different response from that of novel or aversive odors, fox odor or moderate restraint stress. Exposure to predator odor increased energy expenditure, and both the thermogenic and energetic effects persisted when physical activity levels were controlled. Predator odor-induced muscle thermogenesis is subject to associative learning as exposure to a conditioned stimulus provoked a rise in muscle temperature in the absence of the odor. The ability of predator odor to induce thermogenesis is predominantly controlled by sympathetic nervous system activation of ß-adrenergic receptors, as unilateral sympathetic lumbar denervation and a peripherally acting ß-adrenergic antagonist significantly inhibited predator odor-induced muscle thermogenesis. The potential survival value of predator odor-induced changes in muscle physiology is reflected in an enhanced resistance to running fatigue. Lastly, predator odor-induced muscle thermogenesis imparts a meaningful impact on energy expenditure as daily predator odor exposure significantly enhanced weight loss with mild calorie restriction. This evidence signifies contextually provoked, centrally mediated muscle thermogenesis that meaningfully impacts energy balance.


Assuntos
Tecido Adiposo Marrom , Odorantes , Tecido Adiposo Marrom/metabolismo , Animais , Metabolismo Energético , Camundongos , Músculo Esquelético/metabolismo , Ratos , Termogênese
10.
Metabolism ; 102: 153990, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31666192

RESUMO

OBJECTIVE: Rates of overweight and obesity epidemic have risen significantly in the past few decades, and 34% of adults and 15-20% of children and adolescents in the United States are now obese. Melanocortin receptor 4 (MC4R), contributes to appetite control in hypothalamic neurons and is a target for future anti-obesity treatments (such as setmelanotide) or novel drug development effort. Proper MC4R trafficking regulation in hypothalamic neurons is crucial for normal neural control of homeostasis and is altered in obesity and in presence of lipids. The mechanisms underlying altered MC4R trafficking in the context of obesity is still unclear. Here, we discovered that C2CD5 expressed in the hypothalamus is involved in the regulation of MC4R endocytosis. This study unmasked a novel trafficking protein nutritionally regulated in the hypothalamus providing a novel target for MC4R dependent pathways involved in bodyweight homeostasis and Obesity. METHODS: To evaluate the expression of C2cd5, we first used in situ hybridization and RNAscope technology in combination with electronic microscopy. For in vivo, we characterized the energy balance of wild type (WT) and C2CD5 whole-body knockout (C2CD5KO) mice fed normal chow (NC) and/or western-diet (high-fat/high-sucrose/cholesterol) (WD). To this end, we performed comprehensive longitudinal assessment of bodyweight, energy balance (food intake, energy expenditure, locomotor activity using TSE metabolic cages), and glucose homeostasis. In addition, we evaluated the consequence of loss of C2CD5 on feeding behavior changes normally induced by MC4R agonist (Melanotan, MTII) injection in the paraventricular hypothalamus (PVH). For in vitro approach, we tease out the role of C2CD5 and its calcium sensing domain C2 in MC4R trafficking. We focused on endocytosis of MC4R using an antibody feeding experiment (in a neuronal cell line - Neuro2A (N2A) stably expressing HA-MC4R-GFP; against HA-tag and analyzed by flux cytometry). RESULTS: We found that 1) the expression of hypothalamic C2CD5 is decreased in diet-induced obesity models compared to controls, 2) mice lacking C2CD5 exhibit an increase in food intake compared to WT mice, 3) C2CD5 interacts with endocytosis machinery in hypothalamus, 4) loss of functional C2CD5 (lacking C2 domain) blunts MC4R endocytosis in vitro and increases MC4R at the surface that fails to respond to MC4R ligand, and, 5) C2CD5KO mice exhibit decreased acute responses to MTII injection into the PVH. CONCLUSIONS: Based on these, we conclude that hypothalamic C2CD5 is involved in MC4R endocytosis and regulate bodyweight homeostasis. These studies suggest that C2CD5 represents a new protein regulated by metabolic cues and involved in metabolic receptor endocytosis. C2CD5 represent a new target and pathway that could be targeted in Obesity.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Metabolismo Energético/genética , Hipotálamo/metabolismo , Proteínas de Membrana/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , Animais , Peso Corporal/genética , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas , Comportamento Alimentar/fisiologia , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Obesidade/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Transporte Proteico/genética
11.
Cell Rep ; 25(2): 271-277.e4, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30304667

RESUMO

Obesity is associated with many complications, including type 2 diabetes and painful neuropathy. There is no cure or prevention for obesity-induced pain, and the neurobiology underlying the onset of the disease is still obscure. In this study, we observe that western diet (WD)-fed mice developed early allodynia with an increase of ER stress markers in the sensory neurons of the dorsal root ganglia (DRG). Using cell-specific approaches, we demonstrate that neuronal liver X receptor (LXR) activation delays ER stress and allodynia in WD-fed mice. Our findings suggest that lipid-binding nuclear receptors expressed in the sensory neurons of the DRG play a role in the onset of obesity-induced hypersensitivity. The LXR and lipid-sensor pathways represent a research avenue to identify targets to prevent debilitating complications affecting the peripheral nerve system in obesity.


Assuntos
Estresse do Retículo Endoplasmático , Gânglios Espinais/efeitos dos fármacos , Hiperalgesia/etiologia , Receptores X do Fígado/fisiologia , Obesidade/complicações , Células Receptoras Sensoriais/efeitos dos fármacos , 1-Acilglicerofosfocolina O-Aciltransferase/genética , 1-Acilglicerofosfocolina O-Aciltransferase/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Dieta Ocidental/efeitos adversos , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/patologia , Receptores X do Fígado/agonistas , Masculino , Camundongos , Camundongos Knockout , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo
12.
Obesity (Silver Spring) ; 26(5): 885-894, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29566460

RESUMO

OBJECTIVE: Activity thermogenesis and energy expenditure (EE) are elevated in intrinsically lean rats (high-capacity runners [HCR]) and are also stimulated by melanocortin receptor activation in the ventromedial hypothalamus (VMH). This study determined whether HCR are more responsive to central modulation of activity EE compared with low-capacity runners (LCR). METHODS: HCR and LCR rats received intra-VMH microinjections of melanotan II (MTII), a mixed melanocortin receptor agonist. Changes in EE, respiratory exchange ratio, activity EE, muscle heat, norepinephrine turnover, and muscle energetic modulators were compared. RESULTS: HCR were significantly more responsive to intra-VMH MTII-induced changes in EE, activity EE, norepinephrine turnover to some muscle subgroups, and muscle mRNA expression of some energetic modulators. Though HCR had high muscle activity thermogenesis, limited MTII-induced modulation of muscle thermogenesis during activity was seen in LCR only. CONCLUSIONS: An inherently lean, high-capacity rat phenotype showed elevated response to central melanocortin stimulation of activity EE and use of fat as fuel. This may be driven by sympathetic outflow to skeletal muscle, which was elevated after MTII. Central melanocortin receptor activation also altered skeletal muscle energetic modulators in a manner consistent with elevated EE and lowered respiratory exchange ratio.


Assuntos
Músculo Esquelético/metabolismo , Receptores de Melanocortina/metabolismo , Termogênese/fisiologia , Animais , Masculino , Ratos
13.
Physiol Rep ; 5(4)2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28242830

RESUMO

During weight loss, adaptive thermogenesis occurs where energy expenditure (EE) is suppressed beyond that predicted for the smaller body size. Here, we investigated the contributions of resting and nonresting EE to the reduced total EE seen after 3 weeks of 50% calorie restriction (CR) in rats, focusing on activity-associated EE, muscle thermogenesis, and sympathetic outflow. Prolonged food restriction resulted in a 42% reduction in daily EE, through a 40% decrease in resting EE, and a 48% decline in nonresting EE These decreases in EE were significant even when the reductions in body weight and lean mass were taken into account. Along with a decreased caloric need for low-to-moderate-intensity treadmill activity with 50% CR, baseline and activity-related muscle thermogenesis were also suppressed, though the ability to increase muscle thermogenesis above baseline levels was not compromised. When sympathetic drive was measured by assessing norepinephrine turnover (NETO), 50% CR was found to decrease NETO in three of the four muscle groups examined, whereas elevated NETO was found in white adipose tissue of food-restricted rats. Central activation of melanocortin 4 receptors in the ventromedial hypothalamus stimulated this pathway, enhancing activity EE; this was not compromised by 50% CR These data suggest that suppressed activity EE contributes to adaptive thermogenesis during energy restriction. This may stem from decreased sympathetic drive to skeletal muscle, increasing locomotor efficiency and reducing skeletal muscle thermogenesis. The capacity to increase activity EE in response to central stimuli is retained, however, presenting a potential target for preventing weight regain.


Assuntos
Restrição Calórica , Metabolismo Energético/fisiologia , Músculo Esquelético/fisiologia , Condicionamento Físico Animal/fisiologia , Sistema Nervoso Simpático/fisiologia , Termogênese/fisiologia , Animais , Peso Corporal/fisiologia , Masculino , Músculo Esquelético/inervação , Ratos , Ratos Sprague-Dawley
14.
J Physiol ; 594(18): 5285-301, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27126579

RESUMO

KEY POINTS: The ventromedial hypothalamus (VMH) and the central melanocortin system both play vital roles in regulating energy balance by modulating energy intake and utilization. Recent evidence suggests that activation of the VMH alters skeletal muscle metabolism. We show that intra-VMH melanocortin receptor activation increases energy expenditure and physical activity, switches fuel utilization to fats, and lowers work efficiency such that excess calories are dissipated by skeletal muscle as heat. We also show that intra-VMH melanocortin receptor activation increases sympathetic nervous system outflow to skeletal muscle. Intra-VMH melanocortin receptor activation also induced significant changes in the expression of mediators of energy expenditure in muscle. These results support the role of melanocortin receptors in the VMH in the modulation of skeletal muscle metabolism. ABSTRACT: The ventromedial hypothalamus (VMH) and the brain melanocortin system both play vital roles in increasing energy expenditure (EE) and physical activity, decreasing appetite and modulating sympathetic nervous system (SNS) outflow. Because of recent evidence showing that VMH activation modulates skeletal muscle metabolism, we propose the existence of an axis between the VMH and skeletal muscle, modulated by brain melanocortins, modelled on the brain control of brown adipose tissue. Activation of melanocortin receptors in the VMH of rats using a non-specific agonist melanotan II (MTII), compared to vehicle, increased oxygen consumption and EE and decreased the respiratory exchange ratio. Intra-VMH MTII enhanced activity-related EE even when activity levels were held constant. MTII treatment increased gastrocnemius muscle heat dissipation during controlled activity, as well as in the home cage. Compared to vehicle-treated rats, rats with intra-VMH melanocortin receptor activation had higher skeletal muscle norepinephrine turnover, indicating an increased SNS drive to muscle. Lastly, intra-VMH MTII induced mRNA expression of muscle energetic mediators, whereas short-term changes at the protein level were primarily limited to phosphorylation events. These results support the hypothesis that melanocortin peptides act in the VMH to increase EE by lowering the economy of activity via the enhanced expression of mediators of EE in the periphery including skeletal muscle. The data are consistent with the role of melanocortins in the VMH in the modulation of skeletal muscle metabolism.


Assuntos
Metabolismo Energético , Hipotálamo/fisiologia , Músculo Esquelético/fisiologia , Receptores de Melanocortina/fisiologia , Termogênese , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/fisiologia , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/fisiologia , Animais , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/fisiologia , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Norepinefrina/metabolismo , Peptídeos Cíclicos/farmacologia , Condicionamento Físico Animal , Ratos Sprague-Dawley , Receptores de Melanocortina/agonistas , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
15.
Am J Physiol Endocrinol Metab ; 306(6): E635-47, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24398400

RESUMO

A high-calorie diet accompanied by low levels of physical activity (PA) accounts for the widespread prevalence of obesity today, and yet some people remain lean even in this obesogenic environment. Here, we investigate the cause for this exception. A key trait that predicts high PA in both humans and laboratory rodents is intrinsic aerobic capacity. Rats artificially selected as high-capacity runners (HCR) are lean and consistently more physically active than their low-capacity runner (LCR) counterparts; this applies to both males and females. Here, we demonstrate that HCR show heightened total energy expenditure (TEE) and hypothesize that this is due to higher nonresting energy expenditure (NREE; includes activity EE). After matching for body weight and lean mass, female HCR consistently had heightened nonresting EE, but not resting EE, compared with female LCR. Because of the dominant role of skeletal muscle in nonresting EE, we examined muscle energy use. We found that lean female HCR had higher muscle heat dissipation during activity, explaining their low economy of activity and high activity EE. This may be due to the amplified skeletal muscle expression levels of proteins involved in EE and reduced expression levels of proteins involved in energy conservation in HCR relative to LCR. This is also associated with an increased sympathetic drive to skeletal muscle in HCR compared with LCR. We find little support for the hypothesis that resting metabolic rate is correlated with maximal aerobic capacity if body size and composition are fully considered; rather, the critical factor appears to be activity thermogenesis.


Assuntos
Metabolismo Energético , Modelos Biológicos , Músculo Esquelético/metabolismo , Sistema Nervoso Simpático/metabolismo , Termogênese , Magreza/metabolismo , Regulação para Cima , Animais , Composição Corporal , Regulação da Temperatura Corporal , Peso Corporal , Tolerância ao Exercício , Feminino , Regulação da Expressão Gênica , Atividade Motora , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/inervação , Ratos
16.
Physiol Behav ; 125: 21-9, 2014 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-24291381

RESUMO

Brown adipose tissue (BAT) burns calories to produce heat, and is thus relevant to energy balance. Interscapular BAT (IBAT) of donor mice was transplanted into recipient mice (transBATation). To test whether transBATation counteracts high-fat diet (HFD)-induced obesity, some sham-operated and recipient mice were fed a HFD (HFD-sham, HFD-trans) while others remained on a standard chow (chow-sham, chow-trans). HFD-trans mice had lower body weight and fat and greater energy expenditure, but similar caloric intake compared with HFD-sham mice. We hypothesized that HFD-trans mice had elevated sympathetic activity compared with HFD-sham mice, contributing to increased energy expenditure and fuel mobilization. This was supported by findings that HFD-trans mice had greater energy expenditure during a norepinephrine challenge test and higher core temperatures after cold exposure than did HFD-sham mice, implicating enhanced whole-body metabolic response and elevated sympathetic activity. Additionally, transBATation selectively increased sympathetic drive to some, but not all, white adipose tissue depots and skeletal muscles, as well as the endogenous IBAT, heart, and liver. Collectively, transBATation confers resistance to HFD-induced obesity via increase in whole-body sympathetic activity, and differential activation of sympathetic drive to some of the tissues involved in energy expenditure and fuel mobilization.


Assuntos
Tecido Adiposo Marrom/transplante , Obesidade/fisiopatologia , Sistema Nervoso Simpático/fisiologia , Tecido Adiposo Marrom/inervação , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/inervação , Tecido Adiposo Branco/metabolismo , Animais , Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Resposta ao Choque Frio/fisiologia , Dieta Hiperlipídica/efeitos adversos , Ingestão de Energia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Coração/inervação , Fígado/inervação , Fígado/metabolismo , Masculino , Camundongos , Músculo Esquelético/inervação , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Obesidade/metabolismo , Obesidade/cirurgia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA