Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
J Clin Invest ; 133(17)2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37463052

RESUMO

The liver has a high demand for phosphatidylcholine (PC), particularly in overnutrition, where reduced phospholipid levels have been implicated in the development of nonalcoholic fatty liver disease (NAFLD). Whether other pathways exist in addition to de novo PC synthesis that contribute to hepatic PC pools remains unknown. Here, we identified the lysophosphatidylcholine (LPC) transporter major facilitator superfamily domain containing 2A (Mfsd2a) as critical for maintaining hepatic phospholipid pools. Hepatic Mfsd2a expression was induced in patients having NAFLD and in mice in response to dietary fat via glucocorticoid receptor action. Mfsd2a liver-specific deficiency in mice (L2aKO) led to a robust nonalcoholic steatohepatitis-like (NASH-like) phenotype within just 2 weeks of dietary fat challenge associated with reduced hepatic phospholipids containing linoleic acid. Reducing dietary choline intake in L2aKO mice exacerbated liver pathology and deficiency of liver phospholipids containing polyunsaturated fatty acids (PUFAs). Treating hepatocytes with LPCs containing oleate and linoleate, two abundant blood-derived LPCs, specifically induced lipid droplet biogenesis and contributed to phospholipid pools, while LPC containing the omega-3 fatty acid docosahexaenoic acid (DHA) promoted lipid droplet formation and suppressed lipogenesis. This study revealed that PUFA-containing LPCs drive hepatic lipid droplet formation, suppress lipogenesis, and sustain hepatic phospholipid pools - processes that are critical for protecting the liver from excess dietary fat.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Hipernutrição , Animais , Camundongos , Fosfolipídeos/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Fígado/metabolismo , Lisofosfolipídeos/metabolismo , Fosfatidilcolinas/metabolismo , Gorduras na Dieta , Hipernutrição/patologia
2.
Immunity ; 56(1): 58-77.e11, 2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-36521495

RESUMO

Obesity-induced chronic liver inflammation is a hallmark of nonalcoholic steatohepatitis (NASH)-an aggressive form of nonalcoholic fatty liver disease. However, it remains unclear how such a low-grade, yet persistent, inflammation is sustained in the liver. Here, we show that the macrophage phagocytic receptor TREM2, induced by hepatocyte-derived sphingosine-1-phosphate, was required for efferocytosis of lipid-laden apoptotic hepatocytes and thereby maintained liver immune homeostasis. However, prolonged hypernutrition led to the production of proinflammatory cytokines TNF and IL-1ß in the liver to induce TREM2 shedding through ADAM17-dependent proteolytic cleavage. Loss of TREM2 resulted in aberrant accumulation of dying hepatocytes, thereby further augmenting proinflammatory cytokine production. This ultimately precipitated a vicious cycle that licensed chronic inflammation to drive simple steatosis transition to NASH. Therefore, impaired macrophage efferocytosis is a previously unrecognized key pathogenic event that enables chronic liver inflammation in obesity. Blocking TREM2 cleavage to restore efferocytosis may represent an effective strategy to treat NASH.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Hipernutrição , Humanos , Hepatopatia Gordurosa não Alcoólica/patologia , Hipernutrição/patologia , Fígado/patologia , Inflamação/patologia , Obesidade/patologia , Glicoproteínas de Membrana , Receptores Imunológicos
3.
Proc Nutr Soc ; 81(2): 146-161, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35934688

RESUMO

In recent years, a wealth of factors are associated with increased risk of developing non-alcoholic fatty liver disease (NAFLD) and NAFLD is now thought to increase the risk of multiple extra-hepatic diseases. The aim of this review is first to focus on the role of ageing and sex as key, poorly understood risk factors in the development and progression of NAFLD. Secondly, we aim to discuss the roles of white adipose tissue (WAT) and intestinal dysfunction, as producers of extra-hepatic factors known to further contribute to the pathogenesis of NAFLD. Finally, we aim to summarise the role of NAFLD as a multi-system disease affecting other organ systems beyond the liver. Both increased age and male sex increase the risk of NAFLD and this may be partly driven by alterations in the distribution and function of WAT. Similarly, changes in gut microbiota composition and intestinal function with ageing and chronic overnutrition are likely to contribute to the development of NAFLD both directly (i.e. by affecting hepatic function) and indirectly via exacerbating WAT dysfunction. Consequently, the presence of NAFLD significantly increases the risk of various extra-hepatic diseases including CVD, type 2 diabetes mellitus, chronic kidney disease and certain extra-hepatic cancers. Thus changes in WAT and intestinal function with ageing and chronic overnutrition contribute to the development of NAFLD - a multi-system disease that subsequently contributes to the development of other chronic cardiometabolic diseases.


Assuntos
Diabetes Mellitus Tipo 2 , Hepatopatia Gordurosa não Alcoólica , Hipernutrição , Tecido Adiposo , Envelhecimento , Diabetes Mellitus Tipo 2/etiologia , Humanos , Fígado , Masculino , Hepatopatia Gordurosa não Alcoólica/etiologia , Hipernutrição/complicações , Hipernutrição/patologia
4.
J Clin Invest ; 132(14)2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35700043

RESUMO

Hepatic inflammation is culpable for the evolution of asymptomatic steatosis to nonalcoholic steatohepatitis (NASH). Hepatic inflammation results from abnormal macrophage activation. We found that FoxO1 links overnutrition to hepatic inflammation by regulating macrophage polarization and activation. FoxO1 was upregulated in hepatic macrophages, correlating with hepatic inflammation, steatosis, and fibrosis in mice and patients with NASH. Myeloid cell conditional FoxO1 knockout skewed macrophage polarization from proinflammatory M1 to the antiinflammatory M2 phenotype, accompanied by a reduction in macrophage infiltration in liver. These effects mitigated overnutrition-induced hepatic inflammation and insulin resistance, contributing to improved hepatic metabolism and increased energy expenditure in myeloid cell FoxO1-knockout mice on a high-fat diet. When fed a NASH-inducing diet, myeloid cell FoxO1-knockout mice were protected from developing NASH, culminating in a reduction in hepatic inflammation, steatosis, and fibrosis. Mechanistically, FoxO1 counteracts Stat6 to skew macrophage polarization from M2 toward the M1 signature to perpetuate hepatic inflammation in NASH. FoxO1 appears to be a pivotal mediator of macrophage activation in response to overnutrition and a therapeutic target for ameliorating hepatic inflammation to stem the disease progression from benign steatosis to NASH.


Assuntos
Proteína Forkhead Box O1 , Hepatopatia Gordurosa não Alcoólica , Hipernutrição , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Fibrose , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Hipernutrição/patologia
5.
Am J Physiol Endocrinol Metab ; 321(5): E702-E713, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34632797

RESUMO

In chronic obesity, activated adipose tissue proinflammatory cascades are tightly linked to metabolic dysfunction. Yet, close temporal analyses of the responses to obesogenic environment such as high-fat feeding (HFF) in susceptible mouse strains question the causal relationship between inflammation and metabolic dysfunction, and/or raises the possibility that certain inflammatory cascades play adaptive/homeostatic, rather than pathogenic roles. Here, we hypothesized that CTRP6, a C1QTNF family member, may constitute an early responder to acute nutritional changes in adipose tissue, with potential physiological roles. Both 3-days high-fat feeding (3dHFF) and acute obesity reversal [2-wk switch to low-fat diet after 8-wk HFF (8wHFF)] already induced marked changes in whole body fuel utilization. Although adipose tissue expression of classical proinflammatory cytokines (Tnf-α, Ccl2, and Il1b) exhibited no, or only minor, change, C1qtnf6 uniquely increased, and decreased, in response to 3dHFF and acute obesity reversal, respectively. CTRP6 knockout (KO) mouse embryonic fibroblasts (MEFs) exhibited increased adipogenic gene expression (Pparg, Fabp4, and Adipoq) and markedly reduced inflammatory genes (Tnf-α, Ccl2, and Il6) compared with wild-type MEFs, and recombinant CTRP6 induced the opposite gene expression signature, as assessed by RNA sequencing. Consistently, 3dHFF of CTRP6-KO mice induced a greater whole body and adipose tissue weight gain compared with wild-type littermates. Collectively, we propose CTRP6 as a gene that rapidly responds to acute changes in caloric intake, acting in acute overnutrition to induce a "physiological inflammatory response" that limits adipose tissue expansion.NEW & NOTEWORTHY CTRP6 (C1qTNF6), a member of adiponectin gene family, regulates inflammation and metabolism in established obesity. Here, short-term high-fat feeding in mice is shown to increase adipose tissue expression of CTRP6 before changes in the expression of classical inflammatory genes occur. Conversely, CTRP6 expression in adipose tissue decreases early in the course of obesity reversal. Gain- and loss-of-function models suggest CTRP6 as a positive regulator of inflammatory cascades, and a negative regulator of adipogenesis and adipose tissue expansion.


Assuntos
Adipocinas/fisiologia , Tecido Adiposo/patologia , Inflamação/genética , Fenômenos Fisiológicos da Nutrição/genética , Adipogenia/genética , Adipocinas/genética , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Dieta Hiperlipídica , Embrião de Mamíferos , Feminino , Células HEK293 , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho do Órgão/genética , Hipernutrição/genética , Hipernutrição/metabolismo , Hipernutrição/patologia , Gravidez
6.
J Endocrinol ; 250(3): 81-91, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34101615

RESUMO

We tested whether chronic supplementation with soy isoflavones could modulate insulin secretion levels and subsequent recovery of pancreatic islet function as well as prevent metabolic dysfunction induced by early overfeeding in adult male rats. Wistar rats raised in small litters (SL, three pups/dam) and normal litters (NL, nine pups/dam) were used as models of early overfeeding and normal feeding, respectively. At 30 to 90 days old, animals in the SL and NL groups received either soy isoflavones extract (ISO) or water (W) gavage serving as controls. At 90 days old, body weight, visceral fat deposits, glycemia, insulinemia were evaluated. Glucose-insulin homeostasis and pancreatic-islet insulinotropic response were also determined. The early life overnutrition induced by small litter displayed metabolic dysfunction, glucose, and insulin homeostasis disruption in adult rats. However, adult SL rats treated with soy isoflavones showed improvement in glucose tolerance, insulin sensitivity, insulinemia, fat tissue accretion, and body weight gain, compared with the SL-W group. Pancreatic-islet response to cholinergic, adrenergic, and glucose stimuli was improved in both isoflavone-treated groups. In addition, different isoflavone concentrations increased glucose-stimulated insulin secretion in islets of all groups with higher magnitude in both NL and SL isoflavone-treated groups. These results indicate that long-term treatment with soy isoflavones inhibits early overfeeding-induced metabolic dysfunction in adult rats and modulated the process of insulin secretion in pancreatic islets.


Assuntos
Ilhotas Pancreáticas/efeitos dos fármacos , Isoflavonas/farmacologia , Doenças Metabólicas/prevenção & controle , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Isoflavonas/isolamento & purificação , Masculino , Doenças Metabólicas/etiologia , Doenças Metabólicas/patologia , Hipernutrição/complicações , Hipernutrição/metabolismo , Hipernutrição/patologia , Gravidez , Ratos , Ratos Wistar , Fatores Sexuais , /química
7.
Artigo em Inglês | MEDLINE | ID: mdl-33865538

RESUMO

Using a rabbit model, we investigated whether maternal intake of a high-fat and high-carbohydrate diet (HFCD) before and during pregnancy induces an increase in micronuclei frequency and oxidative stress in offspring during adulthood. Female rabbits received a standard diet (SD) or HFCD for two months before mating and during gestation. The offspring from both groups were nursed by foster mothers fed SD until postnatal day 35. After weaning, all the animals received SD until postnatal day 440. At postnatal day 370, the frequency of micronuclei in peripheral blood reticulocytes (MN-RETs) increased in the male offspring from HFCD-fed mothers compared with the male offspring from SD-fed mothers. Additionally, fasting serum glucose increased in the offspring from HFCD-fed mothers compared with the offspring from SD-fed mothers. At postnatal day 440, the offspring rabbits were challenged with HFCD or continued with SD for 30 days. There was an increase in MN-RET frequency in the male rabbits from HFCD-fed mothers, independent of the type of challenging diet consumed during adulthood. The challenge induced changes in serum cholesterol, LDL and HDL that were influenced by the maternal diet and offspring sex. We measured malondialdehyde in the liver of rabbits as an oxidative stress marker after diet challenge. Oxidative stress in the liver only increased in the female offspring from HFCD-fed mothers who were also challenged with this same diet. The data indicate that maternal overnutrition before and during pregnancy is able to promote different effects depending on the sex of the animals, with chromosomal instability in male offspring and oxidative stress and hypercholesterolemia in female offspring. Our data might be important in the understanding of chronic diseases that develop in adulthood due to in utero exposure to maternal diet.


Assuntos
Dano ao DNA , Hipernutrição/genética , Efeitos Tardios da Exposição Pré-Natal/genética , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Hipernutrição/complicações , Hipernutrição/patologia , Estresse Oxidativo/fisiologia , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Coelhos , Caracteres Sexuais
8.
Cell Metab ; 33(7): 1418-1432.e6, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-33761312

RESUMO

Associative learning allows animals to adapt their behavior in response to environmental cues. For example, sensory cues associated with food availability can trigger overconsumption even in sated animals. However, the neural mechanisms mediating cue-driven non-homeostatic feeding are poorly understood. To study this, we recently developed a behavioral task in which contextual cues increase feeding even in sated mice. Here, we show that an insular cortex to central amygdala circuit is necessary for conditioned overconsumption, but not for homeostatic feeding. This projection is marked by a population of glutamatergic nitric oxide synthase-1 (Nos1)-expressing neurons, which are specifically active during feeding bouts. Finally, we show that activation of insular cortex Nos1 neurons suppresses satiety signals in the central amygdala. The data, thus, indicate that the insular cortex provides top-down control of homeostatic circuits to promote overconsumption in response to learned cues.


Assuntos
Comportamento Alimentar/fisiologia , Córtex Insular/fisiologia , Neurônios/fisiologia , Óxido Nítrico Sintase Tipo I/genética , Hipernutrição/etiologia , Animais , Clozapina/análogos & derivados , Clozapina/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Sinais (Psicologia) , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Feminino , Córtex Insular/efeitos dos fármacos , Córtex Insular/metabolismo , Córtex Insular/patologia , Aprendizagem/efeitos dos fármacos , Aprendizagem/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Hipernutrição/genética , Hipernutrição/metabolismo , Hipernutrição/patologia
9.
FEBS J ; 288(12): 3784-3798, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32654397

RESUMO

The pervasion of three daily meals and snacks is a relatively new introduction to our shared experience and is coincident with an epidemic rise in obesity and cardiometabolic disorders of overnutrition. The past two decades have yielded convincing evidence regarding the adaptive, protective effects of calorie restriction (CR) and intermittent fasting (IF) against cardiometabolic, neurodegenerative, proteostatic, and inflammatory diseases. Yet, durable adherence to intensive lifestyle changes is rarely attainable. New evidence now demonstrates that restricting carbohydrate entry into the hepatocyte by itself mimics several key signaling responses and physiological outcomes of IF and CR. This discovery raises the intriguing proposition that targeting hepatocyte carbohydrate transport to mimic fasting and caloric restriction can abate cardiometabolic and perhaps other fasting-treatable diseases. Here, we review the metabolic and signaling fates of a hepatocyte carbohydrate, identify evidence to target the key mediators within these pathways, and provide rationale and data to highlight carbohydrate transport as a broad, proximal intervention to block the deleterious sequelae of hepatic glucose and fructose metabolism.


Assuntos
Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Moduladores de Transporte de Membrana/uso terapêutico , Síndrome Metabólica/prevenção & controle , Obesidade/prevenção & controle , Hipernutrição/prevenção & controle , Animais , Transporte Biológico/efeitos dos fármacos , Restrição Calórica/métodos , Metabolismo dos Carboidratos/efeitos dos fármacos , Metabolismo dos Carboidratos/genética , Modelos Animais de Doenças , Jejum/metabolismo , Expressão Gênica , Glucose/antagonistas & inibidores , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Terapia de Alvo Molecular/métodos , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Hipernutrição/genética , Hipernutrição/metabolismo , Hipernutrição/patologia
10.
J Biol Chem ; 295(34): 12279-12289, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32651233

RESUMO

Nonalcoholic fatty liver diseases (NAFLDs), especially nonalcoholic steatohepatitis (NASH), have become a major cause of liver transplant and liver-associated death. However, the pathogenesis of NASH is still unclear. Currently, there is no FDA-approved medication to treat this devastating disease. AMP-activated protein kinase (AMPK) senses energy status and regulates metabolic processes to maintain homeostasis. The activity of AMPK is regulated by the availability of nutrients, such as carbohydrates, lipids, and amino acids. AMPK activity is increased by nutrient deprivation and inhibited by overnutrition, inflammation, and hypersecretion of certain anabolic hormones, such as insulin, during obesity. The repression of hepatic AMPK activity permits the transition from simple steatosis to hepatocellular death; thus, activation might ameliorate multiple aspects of NASH. Here we review the pathogenesis of NAFLD and the impact of AMPK activity state on hepatic steatosis, inflammation, liver injury, and fibrosis during the transition of NAFL to NASH and liver failure.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Falência Hepática/enzimologia , Fígado/enzimologia , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hipernutrição/enzimologia , Humanos , Fígado/patologia , Falência Hepática/patologia , Hepatopatia Gordurosa não Alcoólica/patologia , Hipernutrição/patologia
11.
Mamm Genome ; 31(5-6): 119-133, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32350605

RESUMO

Nutritional constraints including not only caloric restriction or protein deficiency, but also energy-dense diets affect metabolic health and frequently lead to obesity and insulin resistance, as well as glucose intolerance and type 2 diabetes. The effects of these environmental factors are often mediated via epigenetic modifiers that target the expression of metabolic genes. More recently, it was discovered that such parentally acquired metabolic changes can alter the metabolic health of the filial and grand-filial generations. In mammals, this epigenetic inheritance can either follow an intergenerational or transgenerational mode of inheritance. In the case of intergenerational inheritance, epimutations established in gametes persist through the first round of epigenetic reprogramming occurring during preimplantation development. For transgenerational inheritance, epimutations persist additionally throughout the reprogramming that occurs during germ cell development later in embryogenesis. Differentially expressed transcripts, genomic cytosine methylations, and several chemical modifications of histones are prime candidates for tangible marks which may serve as epimutations in inter- and transgenerational inheritance and which are currently being investigated experimentally. We review, here, the current literature in support of epigenetic inheritance of metabolic traits caused by nutritional constraints and potential mechanisms in man and in rodent model systems.


Assuntos
Diabetes Mellitus Tipo 2/genética , Intolerância à Glucose/genética , Padrões de Herança , Desnutrição/genética , Obesidade/genética , Hipernutrição/genética , Animais , Metilação de DNA , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Desenvolvimento Embrionário/genética , Disruptores Endócrinos/administração & dosagem , Feminino , Interação Gene-Ambiente , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Histonas/genética , Histonas/metabolismo , Humanos , Resistência à Insulina/genética , Masculino , Desnutrição/complicações , Desnutrição/metabolismo , Desnutrição/patologia , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Hipernutrição/complicações , Hipernutrição/metabolismo , Hipernutrição/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
12.
Eur J Pharmacol ; 881: 173200, 2020 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-32445706

RESUMO

Nutritional imbalance in early life may disrupt the hypothalamic control of energy homeostasis and increase the risk of metabolic disease. The hypothalamic serotonin (5-hydroxytryptamine; 5-HT) system based in the hypothalamus plays an important role in the homeostatic control of energy balance, however the mechanisms underlying the regulation of energy metabolism by 5-HT remain poorly described. Several crucial mitochondrial functions are altered by mitochondrial stress. Adaptations to this stress include changes in mitochondrial multiplication (i.e, mitochondrial biogenesis). Due to the scarcity of evidence regarding the effects of serotonin reuptake inhibitors (SSRI) such as fluoxetine (FLX) on mitochondrial function, we sought to investigate the potential contribution of FLX on changes in mitochondrial function and biogenesis occurring in overfed rats. Using a neonatal overfeeding model, male Wistar rats were divided into 4 groups between 39 and 59 days of age based on nutrition and FLX administration: normofed + vehicle (NV), normofed + FLX (NF), overfed + vehicle (OV) and overfed + FLX (OF). We found that neonatal overfeeding impaired mitochondrial respiration and increased oxidative stress biomarkers in the hypothalamus. FLX administration in overfed rats reestablished mitochondrial oxygen consumption, increased mitochondrial uncoupling protein 2 (Ucp2) expression, reduced total reactive species (RS) production and oxidative stress biomarkers, and up-regulated mitochondrial biogenesis-related genes. Taken together our results suggest that FLX administration in overfed rats improves mitochondrial respiratory chain activity and oxidative balance and increases the transcription of genes employed in mitochondrial biogenesis favoring mitochondrial energy efficiency in response to early nutritional imbalance.


Assuntos
Fármacos Antiobesidade/farmacologia , Metabolismo Energético/efeitos dos fármacos , Fluoxetina/farmacologia , Hipotálamo/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Biogênese de Organelas , Hipernutrição/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Fatores Etários , Fenômenos Fisiológicos da Nutrição Animal , Animais , Animais Recém-Nascidos , Animais Lactentes , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Estado Nutricional , Hipernutrição/metabolismo , Hipernutrição/patologia , Hipernutrição/fisiopatologia , Oxirredução , Consumo de Oxigênio , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo
13.
Cell Metab ; 31(3): 580-591.e5, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32032542

RESUMO

It is well documented that the rate of aging can be slowed, but it remains unclear to which extent aging-associated conditions can be reversed. How the interface of immunity and metabolism impinges upon the diabetes pandemic is largely unknown. Here, we show that NLRP3, a pattern recognition receptor, is modified by acetylation in macrophages and is deacetylated by SIRT2, an NAD+-dependent deacetylase and a metabolic sensor. We have developed a cell-based system that models aging-associated inflammation, a defined co-culture system that simulates the effects of inflammatory milieu on insulin resistance in metabolic tissues during aging, and aging mouse models; and demonstrate that SIRT2 and NLRP3 deacetylation prevent, and can be targeted to reverse, aging-associated inflammation and insulin resistance. These results establish the dysregulation of the acetylation switch of the NLRP3 inflammasome as an origin of aging-associated chronic inflammation and highlight the reversibility of aging-associated chronic inflammation and insulin resistance.


Assuntos
Envelhecimento/patologia , Inflamassomos/metabolismo , Inflamação/patologia , Resistência à Insulina , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Doença Crônica , Modelos Animais de Doenças , Glucose/metabolismo , Homeostase , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Proteína 3 que Contém Domínio de Pirina da Família NLR/química , Hipernutrição/patologia , Peptídeos/química , Sirtuína 2/metabolismo
14.
Indian J Med Res ; 149(6): 695-705, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31496522

RESUMO

Large population-based surveys by the Government of India and several other regional studies have reconfirmed the coexisting burden of over- and undernutrition. While time trends from the 2nd, 3rd and 4th rounds of the National Family Health Survey show declining trends in the prevalence of the underweight, it also highlights increasing rates in the overweight/obesity. Dose-response relationships with different micro- and macronutrient consumption with overweight/obesity prevalence have been established. In this context, it was attempted to identify the specific diet pattern and socio-behavioural determinants of overnutrition along with its combat strategies. This review highlights that while the proportion of chronic energy deficiency is decreasing in India, the intake of micronutrients and food groups continues to be below the recommended dietary allowance set by the Indian Council of Medical Research. Distal factors that determine the nutritional imbalance among Indians are presented under (i) household contextual factors, (ii) peer and socio-cultural influencers, and (iii) business and neighbourhood environment. Accumulation of such factors increases the density of obesogenic environment around individuals. Further, the review offers action points at individual, society and policy levels, presented in a 'logframe matrix' for bringing convergence actions across sectors in consultation with programme managers from different ministries/departments.


Assuntos
Obesidade/epidemiologia , Hipernutrição/epidemiologia , Sobrepeso/epidemiologia , Magreza/epidemiologia , Dieta/efeitos adversos , Ingestão de Energia , Feminino , Humanos , Índia/epidemiologia , Masculino , Estado Nutricional , Obesidade/patologia , Obesidade/prevenção & controle , Hipernutrição/patologia , Hipernutrição/prevenção & controle , Sobrepeso/fisiopatologia , Magreza/patologia
15.
J Neuroendocrinol ; 31(8): e12757, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31233242

RESUMO

The early-life environment is important in programming brain development, and metabolic disruptions at this time can have long-lasting effects. Previously, we have shown that rats overfed for the first 3 weeks of their neonatal life maintain obesity into adulthood. Neonatal overfeeding also leads to primed hypothalamic and hippocampal microglia that are hyper-responsive to an immune challenge in adulthood. However, whether this microglial priming contributes to the obese phenotype and whether it is possible to reverse either the obesity or the microglial priming are not clear. In the present study, we hypothesised that an intervention with minocycline during the juvenile period (postnatal day 21-42) would normalise both the microglial priming and obesity. To induce obesity in neonatal Wistar rats, we manipulated the litter sizes in which they were suckled, yielding litters of 12 (control-fed) or four (neonatally overfed). After weaning, we administered minocycline i.p. every second day for a 3-week period and examined body composition and microglial profiles 24 hours following an immune challenge with lipopolysaccharide. As demonstrated previously, neonatal overfeeding resulted in prolonged weight gain. However, minocycline failed to reverse this effect. Minocycline did reverse microglial priming in feeding-related regions of the hypothalamus, with minimal effects on pro-inflammatory cytokines and on microglial number and morphology in the hippocampus. Thus, the programming effect of neonatal overfeeding on microglial priming can be ameliorated by minocycline later in life. However, the persistent obesity seen after neonatal overfeeding is likely not driven by changes in hypothalamic inflammation and microglial activity.


Assuntos
Encefalite/fisiopatologia , Hipotálamo/patologia , Microglia/fisiologia , Obesidade/etiologia , Hipernutrição/complicações , Animais , Animais Recém-Nascidos , Reprogramação Celular/efeitos dos fármacos , Encefalite/complicações , Encefalite/patologia , Feminino , Hipotálamo/efeitos dos fármacos , Masculino , Microglia/efeitos dos fármacos , Microglia/patologia , Minociclina/farmacologia , Obesidade/patologia , Obesidade/fisiopatologia , Hipernutrição/patologia , Hipernutrição/fisiopatologia , Gravidez , Ratos , Ratos Wistar , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/fisiologia
16.
Appl Physiol Nutr Metab ; 44(2): 164-171, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30058348

RESUMO

There is a strong correlation between inadequate gestational and postpartum nutrition and the occurrence of cardiovascular diseases. The present study investigated the effects of a maternal low-protein diet and neonatal overfeeding on the oxidative balance and morphology of the renal cortex of male Wistar rats. Two independent protocols were used. First, pregnant Wistar rats received diets containing either 17% (normal protein) or 8% (low protein) casein throughout pregnancy and lactation. Second, the litter size was reduced by one-third on the third postnatal day to induce overnourishment in offspring. At 30 days, the oxidative balance and morphology of the renal cortex were analyzed. There was a small but significant increase in renal corpuscle area in the low protein (LP, 5%) and overnutrition (ON, 8%) groups. Glomerular tuft area also increased in LP (6%) and ON (9%), as did glomerular cellularity (LP, +11%; ON, +12%). In the oxidative stress analyses, both nutritional insults significantly elevated lipid peroxidation (LP, +18%; ON, +135%) and protein oxidation (LP, +40%; ON, +65%) while significantly reducing nonenzymatic antioxidant defenses, measured as reduced glutathione (LP, -32%; ON, -45%) and total thiol content (LP, -28%; ON, -24%). We also observed a decrease in superoxide dismutase (LP, -78%; ON, -51%), catalase (LP, -18%; ON, -61%), and glutathione S-transferase (only in ON, -44%) activities. Our results demonstrate that nutritional insults, even those of a very different nature, during perinatal development can result in similar changes in oxidative parameters and glomerular morphology in the renal cortex.


Assuntos
Dieta com Restrição de Proteínas/efeitos adversos , Córtex Renal/metabolismo , Glomérulos Renais/patologia , Hipernutrição/metabolismo , Hipernutrição/patologia , Estresse Oxidativo , Animais , Animais Recém-Nascidos , Antioxidantes/metabolismo , Peso Corporal , Feminino , Córtex Renal/patologia , Glomérulos Renais/metabolismo , Peroxidação de Lipídeos , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Wistar
17.
J Clin Invest ; 128(12): 5335-5350, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30226474

RESUMO

Obesity is a major risk factor for developing nonalcoholic fatty liver disease (NAFLD). NAFLD is the most common form of chronic liver disease and is closely associated with insulin resistance, ultimately leading to cirrhosis and hepatocellular carcinoma. However, knowledge of the intracellular regulators of obesity-linked fatty liver disease remains incomplete. Here we showed that hepatic Rho-kinase 1 (ROCK1) drives obesity-induced steatosis in mice through stimulation of de novo lipogenesis. Mice lacking ROCK1 in the liver were resistant to diet-induced obesity owing to increased energy expenditure and thermogenic gene expression. Constitutive expression of hepatic ROCK1 was sufficient to promote adiposity, insulin resistance, and hepatic lipid accumulation in mice fed a high-fat diet. Correspondingly, liver-specific ROCK1 deletion prevented the development of severe hepatic steatosis and reduced hyperglycemia in obese diabetic (ob/ob) mice. Of pathophysiological significance, hepatic ROCK1 was markedly upregulated in humans with fatty liver disease and correlated with risk factors clustering around NAFLD and insulin resistance. Mechanistically, we found that hepatic ROCK1 suppresses AMPK activity and a ROCK1/AMPK pathway is necessary to mediate cannabinoid-induced lipogenesis in the liver. Furthermore, treatment with metformin, the most widely used antidiabetes drug, reduced hepatic lipid accumulation by inactivating ROCK1, resulting in activation of AMPK downstream signaling. Taken together, our findings establish a ROCK1/AMPK signaling axis that regulates de novo lipogenesis, providing a unique target for treating obesity-related metabolic disorders such as NAFLD.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Lipogênese , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hipernutrição/enzimologia , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Humanos , Resistência à Insulina/genética , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Obesos , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/complicações , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Hipernutrição/complicações , Hipernutrição/genética , Hipernutrição/patologia , Quinases Associadas a rho/genética
18.
Adv Exp Med Biol ; 1061: 19-44, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29956204

RESUMO

Overnutrition, usually with obesity and genetic predisposition, lead to insulin resistance, which is an invariable accompaniment of nonalcoholic fatty liver disease (NAFLD). The associated metabolic abnormalities, pre- or established diabetes, hypertension and atherogenic dyslipidemia (clustered as metabolic syndrome) tend to be worse for nonalcoholic steatohepatitis (NASH), revealing it as part of a continuum of metabolic pathogenesis. The origins of hepatocellular injury and lobular inflammation which distinguish NASH from simple steatosis have intrigued investigators, but it is now widely accepted that NASH results from liver lipotoxicity. The key issue is not the quantity of liver fat but the type(s) of lipid molecules that accumulate, and how they are "packaged" to avoid subcellular injury. Possible lipotoxic mediators include free (unesterified) cholesterol, saturated free fatty acids, diacylglycerols, lysophosphatidyl-choline, sphingolipids and ceramide. Lipid droplets are intracellular storage organelles for non-structural lipid whose regulation is influenced by genetic polymorphisms, such as PNPLA3. Cells unable to sequester chemically reactive lipid molecules undergo mitochondrial injury, endoplasmic reticulum (ER) stress and autophagy, all processes of interest for NASH pathogenesis. Lipotoxicity kills hepatocytes by apoptosis, a highly regulated, non-inflammatory form of cell death, but also by necrosis, necroptosis and pyroptosis; the latter involve mitochondrial injury, oxidative stress, activation of c-Jun N-terminal kinase (JNK) and release of danger-associated molecular patterns (DAMPs). DAMPs stimulate innate immunity by binding pattern recognition receptors, such as Toll-like receptor 4 (TLR4) and the NOD-like receptor protein 3 (NLRP3) inflammasome, which release a cascade of pro-inflammatory chemokines and cytokines. Thus, lipotoxic hepatocellular injury attracts inflammatory cells, particularly activated macrophages which surround ballooned hepatocytes as crown-like structures. In both experimental and human NASH, livers contain cholesterol crystals which are a second signal for NLRP3 activation; this causes interleukin (IL)-1ß and IL18 secretion to attract and activate macrophages and neutrophils. Injured hepatocytes also liberate plasma membrane-derived extracellular vesicles; these have been shown to circulate in NASH and to be pro-inflammatory. The way metabolic dysfunction leads to lipotoxicity, innate immune responses and the resultant pattern of cellular inflammation in the liver are likely also relevant to hepatic fibrogenesis and hepatocarcinogenesis. Pinpointing the key molecules involved pharmacologically should eventually lead to effective pharmacotherapy against NASH.


Assuntos
Metabolismo dos Lipídeos , Fígado , Hepatopatia Gordurosa não Alcoólica , Hipernutrição , Animais , Humanos , Inflamação/metabolismo , Inflamação/patologia , Fígado/metabolismo , Fígado/patologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Hipernutrição/metabolismo , Hipernutrição/patologia
19.
J Clin Invest ; 128(1): 141-156, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29202465

RESUMO

The incorporation of excess saturated free fatty acids (SFAs) into membrane phospholipids within the ER promotes ER stress, insulin resistance, and hepatic gluconeogenesis. Thioesterase superfamily member 2 (Them2) is a mitochondria-associated long-chain fatty acyl-CoA thioesterase that is activated upon binding phosphatidylcholine transfer protein (PC-TP). Under fasting conditions, the Them2/PC-TP complex directs saturated fatty acyl-CoA toward ß-oxidation. Here, we showed that during either chronic overnutrition or acute induction of ER stress, Them2 and PC-TP play critical roles in trafficking SFAs into the glycerolipid biosynthetic pathway to form saturated phospholipids, which ultimately reduce ER membrane fluidity. The Them2/PC-TP complex activated ER stress pathways by enhancing translocon-mediated efflux of ER calcium. The increased cytosolic calcium, in turn, led to the phosphorylation of calcium/calmodulin-dependent protein kinase II, which promoted both hepatic insulin resistance and gluconeogenesis. These findings delineate a mechanistic link between obesity and insulin resistance and establish the Them2/PC-TP complex as an attractive target for the management of hepatic steatosis and insulin resistance.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Estresse do Retículo Endoplasmático , Homeostase , Fígado/enzimologia , Proteínas Mitocondriais/metabolismo , Tioléster Hidrolases/metabolismo , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Resistência à Insulina , Fígado/patologia , Fluidez de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/genética , Obesidade/enzimologia , Obesidade/genética , Obesidade/patologia , Hipernutrição/enzimologia , Hipernutrição/genética , Hipernutrição/patologia , Tioléster Hidrolases/genética
20.
Sci Rep ; 7(1): 12911, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-29018245

RESUMO

Unbalanced nutrition early in life is increasingly recognized as an important factor in the development of chronic, non-communicable diseases at adulthood, including metabolic diseases. We aimed to determine whether transient postnatal overfeeding (OF) leads to liver stress-induced premature senescence (SIPS) of hepatocytes in association with liver structure and hepatic function alterations. Litters sizes of male C57BL/6 mice were adjusted to 9 pups (normal feeding, NF) or reduced to 3 pups during the lactation period to induce transient postnatal OF. Compared to the NF group, seven-month-old adult mice transiently overfed during the postnatal period were overweight and developed glucose intolerance and insulin resistance. Their livers showed microsteatosis and fibrosis, while hepatic insulin signaling and glucose transporter protein expressions were altered. Increased hepatic oxidative stress (OS) was observed, with increased superoxide anion production, glucose-6-phosphate dehydrogenase protein expression, oxidative DNA damage and decreased levels of antioxidant defense markers, such as superoxide dismutase and catalase proteins. Hepatocyte senescence was characterized by increased p21WAF, p53, Acp53, p16INK4a and decreased pRb/Rb and Sirtuin-1 (SIRT-1) protein expression levels. Transient postnatal OF induces liver OS at adulthood, associated with hepatocyte SIPS and alterations in liver structure and hepatic functions, which could be mediated by a SIRT-1 deficiency.


Assuntos
Envelhecimento/patologia , Fígado/patologia , Hipernutrição/patologia , Estresse Fisiológico , Animais , Animais Recém-Nascidos , Composição Corporal , Peso Corporal , Dano ao DNA , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Insulina/metabolismo , Fígado/metabolismo , Cirrose Hepática/patologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Transdução de Sinais , Coloração e Rotulagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...