Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 134(3): 534-45, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18692475

RESUMO

Many protein-protein interactions are mediated through independently folding modular domains. Proteome-wide efforts to model protein-protein interaction or "interactome" networks have largely ignored this modular organization of proteins. We developed an experimental strategy to efficiently identify interaction domains and generated a domain-based interactome network for proteins involved in C. elegans early-embryonic cell divisions. Minimal interacting regions were identified for over 200 proteins, providing important information on their domain organization. Furthermore, our approach increased the sensitivity of the two-hybrid system, resulting in a more complete interactome network. This interactome modeling strategy revealed insights into C. elegans centrosome function and is applicable to other biological processes in this and other organisms.


Assuntos
Caenorhabditis elegans/embriologia , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário , Mapeamento de Interação de Proteínas , Animais , Divisão Celular , Domínios e Motivos de Interação entre Proteínas , Proteoma , Técnicas do Sistema de Duplo-Híbrido
2.
Nutr Neurosci ; 25(3): 621-630, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32654659

RESUMO

Introduction: Neuropeptide Y (NPY) signaling in the brain plays an important role in energy regulation, and is altered during diet-induced obesity. Yet, NPY function during the consumption of specific diet components remains to be fully determined. We have previously demonstrated that consumption of a saturated fat component (free-choice high-fat; fcHF), a sucrose solution (high-sugar; fcHS), or both (fcHFHS) combined with a standard diet (chow and water) has diverse effects on Npy expression in the arcuate nucleus and the sensitivity to intraventricular NPY administration. Arcuate NPY neurons project to the lateral hypothalamus (LHA), and NPY administration in the LHA potently promotes chow intake in rats on a standard diet. However, it is currently unclear if short-term consumption of a palatable free-choice diet alters NPY function in the LHA. Therefore, we assessed the effects of intra-LHA NPY administration on intake in rats following one-week consumption of a fcHF, fcHS, or fcHFHS diet.Methods: Male Wistar rats consumed a fcHF, fcHS, fcHFHS, or control (CHOW) diet for one week before NPY (0.3 µg / 0.3 µL) or phosphate-buffered saline (0.3 µL) was administered into the LHA. Intake was measured 2h later. fcHFHS-fed rats were divided into high-fat (fcHFHS-hf) and low-fat (fcHFHS-lf) groups based on differences in basal fat intake.Results: Intra-LHA NPY administration increased chow intake in fcHFHS- (irrespective of basal fat intake), fcHF- and CHOW-fed rats. Intra-LHA NPY infusion increased fat intake in fcHF-, fcHFHS-hf, but not fcHFHS-lf, rats. Intra-LHA NPY infusion did not increase caloric intake in fcHS-fed rats.Discussion: Our data demonstrate that the effects of intra-LHA NPY on caloric intake differ depending on the consumption of a fat or sugar component, or both, in a free-choice diet. Our data also indicate that baseline preference for the fat diet component modulates the effects of intra-LHA NPY in fcHFHS-fed rats.


Assuntos
Região Hipotalâmica Lateral , Neuropeptídeo Y , Animais , Dieta Hiperlipídica , Região Hipotalâmica Lateral/metabolismo , Hipotálamo/metabolismo , Masculino , Neuropeptídeo Y/metabolismo , Ratos , Ratos Wistar , Sacarose
3.
Appetite ; 167: 105597, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34273421

RESUMO

Central dopamine signaling regulates reward-related aspects of feeding behavior, and during diet-induced obesity dopamine receptor signaling is altered. Yet, the influence of dopamine signaling on the consumption of specific dietary components remains to be elucidated. We have previously shown that 6-hydroxydopamine-mediated lesions of dopamine neuron terminals in the lateral shell of the nucleus accumbens promotes fat intake in rats fed a multi-component free-choice high-fat high-sugar (fcHFHS) diet. It is however not yet determined which dopamine receptors are responsible for this shift towards fat preference. In this study, we assess the effects of D1-or D2 receptor acute inhibition in the lateral shell of the nucleus accumbens on fcHFHS diet consumption. We report that infusion of the D1 receptor antagonist SCH2 3390, but not the D2 receptor antagonist raclopride, promotes dietary fat consumption in male Sprague Dawley rats on a fcHFHS diet during 2 h after infusion. Furthermore, anatomical analysis of infusion sites revealed that the rostral region, but not the caudal region, of the lateral shell of the nucleus accumbens is sensitive to the D1 receptor inhibition effects on fat consumption. Our data highlight a role for D1 receptors in the rostral region of the lateral shell of the nucleus accumbens to control dietary fat consumption.


Assuntos
Núcleo Accumbens , Receptores de Dopamina D1 , Animais , Gorduras na Dieta , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D2
4.
Stress ; 22(5): 571-580, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31184537

RESUMO

The melanocortin-4 receptor (MC4R) facilitates hypothalamic-pituitary-adrenocortical (HPA) axis responses to acute stress in male rodents and is a well known to regulator of energy balance. Mutations in the MC4R is the most common monogenic cause of obesity in humans and has been associated with sex-specific effects, but whether stress regulation by the MC4R is sex-dependent, and whether the MC4R facilitates HPA responses to chronic stress, is unknown. We hypothesized that MC4R-signaling contributes to HPA axis dysregulation and metabolic pathophysiology following chronic stress exposure. We measured changes in energy balance, HPA axis tone, and vascular remodeling during chronic variable stress (CVS) in male and female rats with MC4R loss-of-function. Rats were placed into three groups (n = 9-18/genotype/sex) and half of each group was subjected to CVS for 30 days or were non-stressed littermate controls. All rats underwent an acute restraint stress challenge on Day 30. Rats were euthanized on Day 31, adrenals collected for weight, and descending aortas fixed for morphological indices of vascular pathophysiology. We observed a marked interaction between Mc4r genotype and sex for basal HPA axis tone and acute stress responsivity. MC4R loss-of-function blunted both endpoints in males but exaggerated them in females. Contrary to our hypothesis, Mc4r genotype had no effect on either HPA axis responses or metabolic responses to chronic stress. Heightened stress reactivity of females with MC4R mutations suggests a possible mechanism for the sex-dependent effects associated with this mutation in humans and highlights how stress may differentially regulate metabolism in males and females. Lay summary The hypothalamic melanocortin system is an important regulator of energy balance and stress responses. Here, we report a sex-difference in the stress reactivity of rats with a mutation in this system. Our findings highlight how stress may regulate metabolism differently in males and females and may provide insight into sex-differences associated with this mutation in humans.


Assuntos
Doenças Cardiovasculares/etiologia , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptor Tipo 4 de Melanocortina/genética , Estresse Psicológico/complicações , Estresse Psicológico/metabolismo , Glândulas Suprarrenais/metabolismo , Animais , Corticosterona/metabolismo , Feminino , Genótipo , Humanos , Hipotálamo/metabolismo , Masculino , Ratos , Restrição Física , Fatores Sexuais
5.
Appetite ; 120: 527-535, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-28988760

RESUMO

Consumption of fat and sugar induces hyperphagia and increases the prevalence of obesity and diabetes type 2. Low-grade inflammation in the hypothalamus, a key brain area involved in the regulation of energy homeostasis is shown to blunt signals of satiety after long term high fat diet. The fact that this mechanism can be activated after a few days of hyperphagia before apparent obesity is present led to our hypothesis that hypothalamic inflammation is induced with fat and sugar consumption. Here, we used a free-choice high-fat high-sugar (fcHFHS) diet-induced obesity model and tested the effects of differential overnight nutrient intake during the final experimental night on markers of hypothalamic inflammation. Male Wistar rats were fed a control diet or fcHFHS diet for one week, and assigned to three different feeding conditions during the final experimental night: 1) fcHFHS-fed, 2) fed a controlled amount of chow diet, or 3) fasted. RT-qPCR and Western blot were utilized to measure hypothalamic gene and protein expression, of cytokines and intermediates of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. Lastly, we investigated the effects of acute fat intake on markers of hypothalamic inflammation in fat-naïve rats. fcHFHS-fed rats consumed more calories, increased adipose tissue, and showed elevated expression of hypothalamic inflammation markers (increased phosphorylation of NF-κB protein, Nfkbia and Il6 gene expression) compared to chow-fed rats. These effects were evident in rats consuming relative high amounts of fat. Removal of the fat and sugar, or fasting, during the final experimental night ameliorated hypothalamic inflammation. Finally, a positive correlation was observed between overnight acute fat consumption and hypothalamic NF-κB phosphorylation in fat-naïve rats. Our data indicate that one week of fcHFHS diet, and especially the fat component, promotes hypothalamic inflammation, and removal of the fat and sugar component reverses these detrimental effects.


Assuntos
Ingestão de Alimentos , Hipotálamo/fisiopatologia , Inflamação/fisiopatologia , Obesidade/fisiopatologia , Adiposidade , Animais , Citocinas/sangue , Citocinas/genética , Dieta Hiperlipídica , Gorduras na Dieta/administração & dosagem , Açúcares da Dieta/administração & dosagem , Modelos Animais de Doenças , Privação de Alimentos , Hiperfagia/dietoterapia , Hiperfagia/etiologia , Leptina/sangue , Masculino , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação , Ratos , Ratos Wistar
6.
Am J Physiol Endocrinol Metab ; 307(11): E1065-72, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25315695

RESUMO

Vertical sleeve gastrectomy (VSG) is currently one of the most effective treatments for obesity. Despite recent developments, the underlying mechanisms that contribute to the metabolic improvements following bariatric surgery remain unresolved. VSG reduces postprandial intestinal triglyceride (TG) production, but whether the effects of VSG on intestinal metabolism are related to metabolic outcomes has yet to be established. The lipid synthesis enzyme acyl CoA:monoacylglycerol acyltransferase-2 (Mogat2; MGAT2) plays a crucial role in the assimilation of dietary fat in the intestine and in regulation of adiposity stores as well. Given the phenotypic similarities between VSG-operated and MGAT2-deficient animals, we reasoned that this enzyme could also have a key role in mediating the metabolic benefits of VSG. However, VSG reduced body weight and fat mass and improved glucose metabolism similarly in whole body MGAT2-deficient (Mogat2(-/-)) mice and wild-type littermates. Furthermore, along with an increase in energy expenditure, surgically naive Mogat2(-/-) mice had altered macronutrient preference, shifting preference away from fat and toward carbohydrates, and increased locomotor activity. Collectively, these data suggest that the beneficial effects of VSG on body weight and glucose metabolism are independent of MGAT2 activity and rather that they are separate from the effects of MGAT2 deficiency. Because MGAT2 inhibitors are proposed as a pharmacotherapeutic option for obesity, our data suggest that, in addition to increasing energy expenditure, shifting macronutrient preference away from fat could be another important mechanism by which these compounds could contribute to weight loss.


Assuntos
Gastrectomia , N-Acetilglucosaminiltransferases/deficiência , Animais , Composição Corporal , Peso Corporal , Dieta com Restrição de Gorduras , Ingestão de Alimentos , Preferências Alimentares , Masculino , Camundongos , Camundongos Knockout , Obesidade/genética
7.
Brain Plast ; 9(1-2): 43-73, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38993577

RESUMO

In our ageing global population, the cognitive decline associated with dementia and neurodegenerative diseases represents a major healthcare problem. To date, there are no effective treatments for age-related cognitive impairment, thus preventative strategies are urgently required. Physical exercise is gaining traction as a non-pharmacological approach to promote brain health. Adult hippocampal neurogenesis (AHN), a unique form of brain plasticity which is necessary for certain cognitive functions declines with age and is enhanced in response to exercise. Accumulating evidence from research in rodents suggests that physical exercise has beneficial effects on cognition through its proneurogenic capabilities. Given ethical and technical limitations in human studies, preclinical research in rodents is crucial for a better understanding of such exercise-induced brain and behavioural changes. In this review, exercise paradigms used in preclinical research are compared. We provide an overview of the effects of different exercise paradigms on age-related cognitive decline from middle-age until older-age. We discuss the relationship between the age-related decrease in AHN and the potential impact of exercise on mitigating this decline. We highlight the emerging literature on the impact of exercise on gut microbiota during ageing and consider the role of the gut-brain axis as a future possible strategy to optimize exercise-enhanced cognitive function. Finally, we propose a guideline for designing optimal exercise protocols in rodent studies, which would inform clinical research and contribute to developing preventative strategies for age-related cognitive decline.

8.
Am J Physiol Regul Integr Comp Physiol ; 305(1): R68-77, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23616105

RESUMO

Overconsumption of a high-fat diet promotes weight gain that can result in obesity and associated comorbidities, including Type 2 diabetes mellitus. Consumption of a high-fat diet also alters gut-brain communication. Glucagon-like peptide 1 (GLP-1) is an important gastrointestinal signal that modulates both short- and long-term energy balance and is integral in maintenance of glucose homeostasis. In the current study, we investigated whether high-fat diets (40% or 81% kcal from fat) modulated the ability of the GLP-1 receptor (GLP-1r) agonists exendin-4 (Ex4) and liraglutide to reduce food intake and body weight. We observed that rats maintained on high-fat diets had a delayed acute anorexic response to peripheral administration of Ex4 or liraglutide compared with low-fat diet-fed rats (17% kcal from fat). However, once suppression of food intake in response to Ex4 or liraglutide started, the effect persisted for a longer time in the high-fat diet-fed rats compared with low-fat diet-fed rats. In contrast, centrally administered Ex4 suppressed food intake similarly between high-fat diet-fed and low-fat diet-fed rats. Chronic consumption of a high-fat diet did not change the pharmacokinetics of Ex4 but increased intestinal Glp1r expression and decreased hindbrain Glp1r expression. Taken together, these findings demonstrate that dietary composition alters the temporal profile of the anorectic response to exogenous GLP-1r agonists.


Assuntos
Peso Corporal/fisiologia , Dieta Hiperlipídica , Ingestão de Alimentos/fisiologia , Desnutrição/fisiopatologia , Receptores de Glucagon/agonistas , Receptores de Glucagon/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Dieta com Restrição de Gorduras , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Exenatida , Peptídeo 1 Semelhante ao Glucagon/efeitos adversos , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Glucose/metabolismo , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/farmacologia , Liraglutida , Masculino , Desnutrição/induzido quimicamente , Modelos Animais , Peptídeos/efeitos adversos , Peptídeos/farmacologia , Ratos , Ratos Long-Evans , Peçonhas/efeitos adversos , Peçonhas/farmacologia
9.
Brain Connect ; 13(9): 541-552, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37578129

RESUMO

Introduction: The habenula, a brain region involved in aversion, might negatively modulate caloric intake. Functional magnetic resonance imaging (fMRI) studies reported associations between weight loss and habenula functional connectivity. However, whether habenula resting-state functional connectivity (rsFC) and reward-related activity are altered in obesity is yet unknown. Methods: Using data from the Human Connectome Project, we included 300 subjects with various body mass indexes (BMIs) and a healthy long-term blood glucose (hemoglobin A1c [HbA1c]). In addition, we investigated a potential BMI × HbA1c interaction in a separate cohort including subjects with prediabetes (n = 72). Habenula rsFC was assessed using a region of interest (ROI)-to-ROI analysis. Furthermore, a separate analysis using gambling task fMRI data focused on reward-related habenula activity. Results: We did not find an association between BMI and habenula rsFC for any of the ROIs. For the exploratory analysis of the BMI × HbA1c effect, a significant interaction effect was found for the habenula-ventral tegmental area (VTA) connection, but this did not survive multiple comparisons correction. Monetary punishment compared with reward activated the bilateral habenula in the BMI sample, but this activity was not associated with BMI. Discussion: In conclusion, we did not find evidence for an association between BMI and habenula rsFC or reward-related activity. However, there might be an interaction between BMI and HbA1c for the habenula-VTA rsFC, suggestive of a role of the habenula in glucose regulation. Future studies should focus on metabolic parameters in their experimental design to confirm our findings and explore the precise role of the habenula in metabolism.


Assuntos
Conectoma , Habenula , Humanos , Conectoma/métodos , Habenula/diagnóstico por imagem , Habenula/fisiologia , Hemoglobinas Glicadas , Imageamento por Ressonância Magnética , Obesidade/diagnóstico por imagem , Recompensa
10.
Physiol Behav ; 268: 114239, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37196819

RESUMO

Physical exercise training has been positioned as a behavioral strategy to prevent or alleviate obesity via promotion of energy expenditure as well as modulation of energy intake resulting from changes in dietary preference. Brain adaptations underlying the latter process are incompletely understood. Voluntary wheel running (VWR) is a self-reinforcing rodent paradigm that mimics aspects of human physical exercise training. Behavioral and mechanistic insight from such fundamental studies can help optimize therapies that improve body weight and metabolic health based on physical exercise training in humans. To assess the effects of VWR on dietary self-selection, male Wistar rats were given access to a two-component "no-choice" control diet (CD; consisting of prefabricated nutritionally complete pellets and a bottle with tap water) or a four-component free-choice high-fat high-sucrose diet (fc-HFHSD; consisting of a container with prefabricated nutritionally complete pellets, a dish with beef tallow, a bottle with tap water, and a bottle with 30% sucrose solution). Metabolic parameters and baseline dietary self-selection behavior during sedentary (SED) housing were measured for 21 days, after which half of the animals were allowed to run on a vertical running wheel (VWR) for another 30 days. This resulted in four experimental groups (SEDCD, SEDfc-HFHSD, VWRCD, and VWRfc-HFHSD). Gene expression of opioid and dopamine neurotransmission components, which are associated with dietary self-selection, was assessed in the lateral hypothalamus (LH) and nucleus accumbens (NAc), two brain regions involved in reward-related behavior, following 51 and 30 days of diet consumption and VWR, respectively. Compared to CD controls, consumption of fc-HFHSD before and during VWR did not alter total running distances. VWR and fc-HFHSD had opposite effects on body weight gain and terminal fat mass. VWR transiently lowered caloric intake and increased and decreased terminal adrenal and thymus mass, respectively, independent of diet. VWR during fc-HFHSD consumption consistently increased CD self-selection, had an acute negative effect on fat self-selection, and a delayed negative effect on sucrose solution self-selection compared to SED controls. Gene expression of opioid and dopamine neurotransmission components in LH and NAc were unaltered by fc-HFHSD or VWR. We conclude that VWR modulates fc-HFHSD component self-selection in a time-dependent manner in male Wistar rats.


Assuntos
Analgésicos Opioides , Atividade Motora , Ratos , Animais , Bovinos , Masculino , Humanos , Ratos Wistar , Analgésicos Opioides/farmacologia , Dopamina/farmacologia , Dieta Hiperlipídica , Peso Corporal , Sacarose/farmacologia
11.
Temperature (Austin) ; 10(3): 379-393, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37554387

RESUMO

We have previously identified predator odor as a potent stimulus activating thermogenesis in skeletal muscle in rats. As this may prove relevant for energy balance and weight loss, the current study investigated whether skeletal muscle thermogenesis was altered with negative energy balance, obesity propensity seen in association with low intrinsic aerobic fitness, and monogenic obesity. First, weight loss subsequent to 3 wk of 50% calorie restriction suppressed the muscle thermogenic response to predator odor. Next, we compared rats bred based on artificial selection for intrinsic aerobic fitness - high- and low-capacity runners (HCR, LCR) - that display robust leanness and obesity propensity, respectively. Aerobically fit HCR showed enhanced predator odor-induced muscle thermogenesis relative to the less-fit LCR. This contrasted with the profound monogenic obesity displayed by rats homozygous for a loss of function mutation in Melanocortin 4 receptor (Mc4rK3a,4X/K314X rats), which showed no discernable deficit in thermogenesis. Taken together, these data imply that body size or obesity per se are not associated with deficient muscle thermogenesis. Rather, the physiological phenotype associated with polygenic obesity propensity may encompass pleiotropic mechanisms in the thermogenic pathway. Adaptive thermogenesis associated with weight loss also likely alters muscle thermogenic mechanisms.

12.
J Biol Chem ; 286(30): 26781-93, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21715287

RESUMO

The Lpin1 gene encodes the phosphatidate phosphatase (PAP1) enzyme Lipin 1, which plays a critical role in lipid metabolism. In this study we describe the identification and characterization of a rat model with a mutated Lpin1 gene (Lpin1(1Hubr)), generated by N-ethyl-N-nitrosourea mutagenesis. Lpin1(1Hubr) rats are characterized by hindlimb paralysis and mild lipodystrophy that are detectable from the second postnatal week. Sequencing of Lpin1 identified a point mutation in the 5'-end splice site of intron 18 resulting in mis-splicing, a reading frameshift, and a premature stop codon. As this mutation does not induce nonsense-mediated decay, it allows the production of a truncated Lipin 1 protein lacking PAP1 activity. Lpin1(1Hubr) rats developed hypomyelination and mild lipodystrophy rather than the pronounced demyelination and adipocyte defects characteristic of Lpin1(fld/fld) mice, which carry a null allele for Lpin1. Furthermore, biochemical, histological, and molecular analyses revealed that these lesions improve in older Lpin1(1Hubr) rats as compared with young Lpin1(1Hubr) rats and Lpin1(fld/fld) mice. We observed activation of compensatory biochemical pathways substituting for missing PAP1 activity that, in combination with a possible non-enzymatic Lipin 1 function residing outside of its PAP1 domain, may contribute to the less severe phenotypes observed in Lpin1(1Hubr) rats as compared with Lpin1(fld/fld) mice. Although we are cautious in making a direct parallel between the presented rodent model and human disease, our data may provide new insight into the pathogenicity of recently identified human LPIN1 mutations.


Assuntos
Doenças Desmielinizantes/enzimologia , Íntrons , Lipodistrofia/enzimologia , Mutação , Fosfatidato Fosfatase/metabolismo , Alquilantes/farmacologia , Animais , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/patologia , Etilnitrosoureia/farmacologia , Células HEK293 , Humanos , Lipodistrofia/genética , Lipodistrofia/patologia , Camundongos , Mutagênese , Proteínas Associadas a Pancreatite , Fosfatidato Fosfatase/genética , Estrutura Terciária de Proteína , Sítios de Splice de RNA , Ratos , Ratos Mutantes
13.
Am J Physiol Endocrinol Metab ; 303(1): E103-10, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22535749

RESUMO

Bariatric surgery is currently the most effective treatment for obesity. Vertical sleeve gastrectomy (VSG), a commonly applied bariatric procedure, involves surgically incising most of the volume of the stomach. In humans, partial loss of melanocortin receptor-4 (MC4R) activity is the most common monogenic correlate of obesity regardless of lifestyle. At present it is unclear whether genetic alteration of MC4R signaling modulates the beneficial effects of VSG. Following VSG, we analyzed body weight, food intake, glucose sensitivity, and macronutrient preference of wild-type and MC4R-deficient (Mc4r(+/-) and Mc4r(-/-)) rats compared with sham-operated controls. VSG reduced body weight and fat mass and improved glucose metabolism and also shifted preference toward carbohydrates and away from fat. All of this occurred independently of MC4R activity. In addition, MC4R was resequenced in 46 human subjects who underwent VSG. We observed common genetic variations in the coding sequence of MC4R in five subjects. However, none of those variations appeared to affect the outcome of VSG. Taken together, these data suggest that the beneficial effect of VSG on body weight and glucose metabolism is not mediated by alterations in MC4R activity.


Assuntos
Modelos Animais de Doenças , Gastrectomia , Gastroplastia , Obesidade/metabolismo , Obesidade/cirurgia , Receptor Tipo 4 de Melanocortina/metabolismo , Adiposidade , Animais , Comportamento Animal , Ingestão de Energia , Preferências Alimentares , Gastrectomia/métodos , Gastroplastia/métodos , Estudos de Associação Genética , Variação Genética , Glucose/metabolismo , Intolerância à Glucose/etiologia , Intolerância à Glucose/prevenção & controle , Heterozigoto , Homozigoto , Humanos , Masculino , Mutação , Obesidade/genética , Obesidade/fisiopatologia , Ratos , Receptor Tipo 4 de Melanocortina/química , Receptor Tipo 4 de Melanocortina/genética , Redução de Peso
14.
Neurobiol Stress ; 15: 100410, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34926732

RESUMO

BACKGROUND: Social stress is an important environmental risk factor for the development of psychiatric disorders, including depression and anxiety disorders. Social stress paradigms are commonly used in rats and mice to gain insight into the pathogenesis of these disorders. The social instability stress (SIS) paradigm entails frequent (up to several times a week) introduction of one or multiple unfamiliar same-sex home-cage partners. The subsequent recurring formation of a new social hierarchy results in chronic and unpredictable physical and social stress. PURPOSE: We compare and discuss the stress-related behavioral and physiological impact of SIS protocols in rat and mouse, and address limitations due to protocol variability. We further provide practical recommendations to optimize reproducibility of SIS protocols. METHODS: We conducted a systematic review in accordance with the PRISMA statement in the following three databases: PubMed, Web of Science and Scopus. Our search strategy was not restricted to year of publication but was limited to articles in English that were published in peer-reviewed journals. Search terms included "social* instab*" AND ("animal" OR "rodent" OR "rat*" OR "mice" OR "mouse"). RESULTS: Thirty-three studies met our inclusion criteria. Fifteen articles used a SIS protocol in which the composition of two cage mates is altered daily for sixteen days (SIS16D). Eleven articles used a SIS protocol in which the composition of four cage mates is altered twice per week for 49 days (SIS49D). The remaining seven studies used SIS protocols that differed from these two protocols in experiment duration or cage mate quantity. Behavioral impact of SIS was primarily assessed by quantifying depressive-like, anxiety-like, social-, and cognitive behavior. Physiological impact of SIS was primarily assessed using metabolic parameters, hypothalamus-pituitary-adrenal axis activity, and the assessment of neurobiological parameters such as neuroplasticity and neurogenesis. CONCLUSION: Both shorter and longer SIS protocols induce a wide range of stress-related behavioral and physiological impairments that are relevant for the pathophysiology of depression and anxiety disorders. To date, SIS16D has only been reported in rats, whereas SIS49D has only been reported in mice. Given this species-specific application as well as variability in reported SIS protocols, additional studies should determine whether SIS effects are protocol duration- or species-specific. We address several issues, including a lack of consistency in the used SIS protocols, and suggest practical, concrete improvements in design and reporting of SIS protocols to increase standardization and reproducibility of this etiologically relevant preclinical model of social stress.

16.
Am J Physiol Endocrinol Metab ; 298(3): E477-88, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19934402

RESUMO

Postnatal development and puberty are times of strong physical maturation and require large quantities of energy. The hypothalamic neuropeptide melanin-concentrating hormone (MCH) regulates nutrient intake and energy homeostasis, but the underlying mechanisms are not completely understood. Here we use a novel rat knockout model in which the MCH precursor Pmch has been inactivated to study the effects of loss of MCH on energy regulation in more detail. Pmch(-/-) rats are lean, hypophagic, osteoporotic, and although endocrine parameters were changed in pmch(-/-) rats, endocrine dynamics were normal, indicating an adaptation to new homeostatic levels rather than disturbed metabolic mechanisms. Detailed body weight growth and feeding behavior analysis revealed that Pmch expression is particularly important during early rat development and puberty, i.e., the first 8 postnatal weeks. Loss of Pmch resulted in a 20% lower set point for body weight that was determined solely during this period and remained unchanged during adulthood. Although the final body weight is diet dependent, the Pmch-deficiency effect was similar for all diets tested in this study. Loss of Pmch affected energy expenditure in both young and adult rats, although these effects seem secondary to the observed hypophagia. Our findings show an important role for Pmch in energy homeostasis determination during early development and indicate that the MCH receptor 1 system is a plausible target for childhood obesity treatment, currently a major health issue in first world countries.


Assuntos
Peso Corporal/fisiologia , Ingestão de Energia/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Homeostase/fisiologia , Hormônios Hipotalâmicos/metabolismo , Hormônios Hipotalâmicos/fisiologia , Melaninas/metabolismo , Hormônios Hipofisários/metabolismo , Precursores de Proteínas/fisiologia , Animais , Ratos
17.
Artigo em Inglês | MEDLINE | ID: mdl-32111717

RESUMO

OBJECTIVE: Poor maternal and paternal environments increase the risk for obesity and diabetes in offspring, whereas maternal and paternal exercise in mice can improve offspring metabolic health. We determined the effects of combined maternal and paternal exercise on offspring health and the effects of parental exercise on offspring pancreas phenotype, a major tissue regulating glucose homeostasis. RESEARCH DESIGN AND METHODS: Breeders were high fat fed and housed±running wheels before breeding (males) and before and during gestation (females). Offspring groups were: both parents sedentary (Sed); maternal exercise only (Mat Ex); paternal exercise only (Pat Ex); and maternal+paternal exercise (Mat+Pat Ex). Offspring were sedentary, chow fed, and studied at weaning, 12, 20 and 52 weeks. RESULTS: While there was no effect of parental exercise on glucose tolerance at younger ages, at 52 weeks, offspring of Mat Ex, Pat Ex and Mat+Pat Ex displayed lower glycemia and improved glucose tolerance. The greatest effects were in offspring from parents that both exercised (Mat+Pat Ex). Offspring from Mat Ex, Pat Ex, and Mat+Pat Ex had decreased beta cell size, whereas islet size and beta cell mass only decreased in Mat+Pat Ex offspring. CONCLUSIONS: Maternal and paternal exercise have additive effects to improve glucose tolerance in offspring as they age, accompanied by changes in the offspring endocrine pancreas. These findings have important implications for the prevention and treatment of type 2 diabetes.


Assuntos
Glicemia/análise , Pai , Homeostase/fisiologia , Células Secretoras de Insulina/metabolismo , Mães , Fenótipo , Condicionamento Físico Animal/métodos , Animais , Diabetes Mellitus Tipo 2/prevenção & controle , Dieta Hiperlipídica , Feminino , Teste de Tolerância a Glucose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/prevenção & controle , Gravidez , Comportamento Sedentário , Desmame
18.
J Comp Neurol ; 527(16): 2659-2674, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30950054

RESUMO

The hypothalamic neuropeptide Y (NPY) circuitry is a key regulator of feeding behavior. NPY also acts in the mesolimbic dopaminergic circuitry, where it can increase motivational aspects of feeding behavior through effects on dopamine output in the nucleus accumbens (NAc) and on neurotransmission in the ventral tegmental area (VTA). Endogenous NPY in the NAc originates from local interneurons and afferent projections from the hypothalamic arcuate nucleus (Arc). However, the origin of endogenous NPY in the VTA is unknown. We determined, in normal-weight male Wistar rats, if the source of VTA NPY is local, and/or whether it is derived from VTA-projecting neurons. Immunocytochemistry, in situ hybridization and RT-qPCR were utilized, when appropriate in combination with colchicine treatment or 24 hr fasting, to assess NPY/Npy expression locally in the VTA. Retrograde tracing using cholera toxin beta (CTB) in the VTA, fluorescent immunocytochemistry and confocal microscopy were used to determine NPY-immunoreactive afferents to the VTA. NPY in the VTA was observed in fibers, but not following colchicine pretreatment. No NPY- or Npy-expressing cell bodies were observed in the VTA. Fasting for 24 hr, which increased Npy expression in the Arc, failed to induce Npy expression in the VTA. Double-labeling with CTB and NPY was observed in the Arc and in the ventrolateral medulla. Thus, VTA NPY originates from the hypothalamic Arc and the ventrolateral medulla of the brainstem in normal-weight male Wistar rats. These afferent connections link hypothalamic and brainstem processing of physiologic state to VTA-driven motivational behavior.


Assuntos
Neurônios Aferentes/citologia , Neurônios Aferentes/metabolismo , Neuropeptídeo Y/metabolismo , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo , Vias Aferentes/citologia , Vias Aferentes/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/metabolismo , Imuno-Histoquímica , Masculino , Bulbo/citologia , Bulbo/metabolismo , Microscopia Confocal , Técnicas de Rastreamento Neuroanatômico , Pró-Opiomelanocortina/metabolismo , Ratos Wistar
19.
J Neuroendocrinol ; 31(5): e12718, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30958590

RESUMO

Humans have engineered a dietary environment that has driven the global prevalence of obesity and several other chronic metabolic diseases to pandemic levels. To prevent or treat obesity and associated comorbidities, it is crucial that we understand how our dietary environment, especially in combination with a sedentary lifestyle and/or daily-life stress, can dysregulate energy balance and promote the development of an obese state. Substantial mechanistic insight into the maladaptive adaptations underlying caloric overconsumption and excessive weight gain has been gained by analysing brains from rodents that were eating prefabricated nutritionally-complete pellets of high-fat diet (HFD). Although long-term consumption of HFDs induces chronic metabolic diseases, including obesity, they do not model several important characteristics of the modern-day human diet. For example, prefabricated HFDs ignore the (effects of) caloric consumption from a fluid source, do not appear to model the complex interplay in humans between stress and preference for palatable foods, and, importantly, lack any aspect of choice. Therefore, our laboratory uses an obesogenic free-choice high-fat high-sucrose (fc-HFHS) diet paradigm that provides rodents with the opportunity to choose from several diet components, varying in palatability, fluidity, texture, form and nutritive content. Here, we review recent advances in our understanding how the fc-HFHS diet disrupts peripheral metabolic processes and produces adaptations in brain circuitries that govern homeostatic and hedonic components of energy balance. Current insight suggests that the fc-HFHS diet has good construct and face validity to model human diet-induced chronic metabolic diseases, including obesity, because it combines the effects of food palatability and energy density with the stimulating effects of variety and choice. We also highlight how behavioural, physiological and molecular adaptations might differ from those induced by prefabricated HFDs that lack an element of choice. Finally, the advantages and disadvantages of using the fc-HFHS diet for preclinical studies are discussed.


Assuntos
Dieta Hiperlipídica , Modelos Animais de Doenças , Ingestão de Energia , Doenças Metabólicas/fisiopatologia , Obesidade/fisiopatologia , Animais , Comportamento de Escolha , Açúcares da Dieta/administração & dosagem , Metabolismo Energético , Humanos , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Estresse Psicológico
20.
Nat Metab ; 1(2): 291-303, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-31032475

RESUMO

Exercise improves health and well-being across diverse organ systems, and elucidating mechanisms underlying the beneficial effects of exercise can lead to new therapies. Here, we show that transforming growth factor-ß2 (TGF-ß2) is secreted from adipose tissue in response to exercise and improves glucose tolerance in mice. We identify TGF-ß2 as an exercise-induced adipokine in a gene expression analysis of human subcutaneous adipose tissue biopsies after exercise training. In mice, exercise training increases TGF-ß2 in scWAT, serum, and its secretion from fat explants. Transplanting scWAT from exercise-trained wild type mice, but not from adipose tissue-specific Tgfb2-/- mice, into sedentary mice improves glucose tolerance. TGF-ß2 treatment reverses the detrimental metabolic effects of high fat feeding in mice. Lactate, a metabolite released from muscle during exercise, stimulates TGF-ß2 expression in human adipocytes. Administration of the lactate-lowering agent dichloroacetate during exercise training in mice decreases circulating TGF-ß2 levels and reduces exercise-stimulated improvements in glucose tolerance. Thus, exercise training improves systemic metabolism through inter-organ communication with fat via a lactate-TGF-ß2-signaling cycle.


Assuntos
Adipocinas/metabolismo , Ácidos Graxos/metabolismo , Glucose/metabolismo , Condicionamento Físico Animal , Fator de Crescimento Transformador beta2/metabolismo , Tecido Adiposo/metabolismo , Animais , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA