Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
J Physiol ; 595(21): 6635-6652, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28759122

RESUMO

KEY POINTS: Offspring of overweight and obese women are at greater risk for respiratory complications at birth. We determined the effect of late gestation maternal overnutrition (LGON) in sheep on surfactant maturation, glucose transport and fatty acid metabolism in the lung in fetal and postnatal life. There were significant decreases in surfactant components and numerical density of surfactant producing cells in the alveolar epithelium due to LGON in the fetal lung. However, there were no differences in the levels of these surfactant components between control and LGON lambs at 30 days of age. The reduced capacity for surfactant production in fetuses as a result of LGON may affect the transition to air breathing at birth. There was altered glucose transport and fatty acid metabolism in the lung as a result of LGON in postnatal life. However, there is a normalisation of surfactant components that suggests accelerated maturation in the lungs after birth. ABSTRACT: With the increasing incidence of obesity worldwide, the proportion of women entering pregnancy overweight or obese has increased dramatically. The fetus of an overnourished mother experiences numerous metabolic changes that may modulate lung development and hence successful transition to air breathing at birth. We used a sheep model of maternal late gestation overnutrition (LGON; from 115 days' gestation, term 147 ± 3 days) to determine the effect of exposure to an increased plane of nutrition in late gestation on lung development in the fetus (at 141 days' gestation) and the lamb (30 days after birth). We found a decrease in the numerical density of surfactant protein positive cells, as well as a reduction in mRNA expression of surfactant proteins (SFTP-A, -B and -C), a rate limiting enzyme in surfactant phospholipid synthesis (phosphate cytidylyltransferase 1, choline, α; PCYT1A), and glucose transporters (SLC2A1 and SLC2A4) in the fetal lung. In lambs at 30 days after birth, there were no differences between Control and LGON groups in the surfactant components that were downregulated in the LGON fetuses. However, mRNA expression of SFTP-A, PCYT1A, peroxisome proliferator activated receptor-γ, fatty acid synthase and fatty acid transport protein were increased in LGON lambs compared to controls. These results indicate a reduced capacity for surfactant production in late gestation. While these deficits are normalised by 30 days after birth, the lungs of LGON lambs exhibited altered glucose transport and fatty acid metabolism, which is consistent with an enhanced capacity for surfactant synthesis and restoration of surfactant maturity in these animals.


Assuntos
Pulmão/embriologia , Hipernutrição/metabolismo , Complicações na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Proteínas Associadas a Surfactantes Pulmonares/metabolismo , Animais , Ácidos Graxos/metabolismo , Feminino , Glucose/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Hipernutrição/patologia , Gravidez , Complicações na Gravidez/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Proteínas Associadas a Surfactantes Pulmonares/genética , Mucosa Respiratória/embriologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Ovinos
8.
Biochem Biophys Res Commun ; 478(3): 1310-6, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27562717

RESUMO

Dieting often leads to body weight cycling involving repeated weight loss and regain. However, little information is available regarding rapid-response serum markers of overnutrition that predict body weight alterations during weight cycling. Here, we report the rapid response of serum leukocyte cell-derived chemotaxin 2 (LECT2), a hepatokine that induces insulin resistance in skeletal muscle, during diet-induced weight cycling in mice. A switch from a high-fat diet (HFD) to a regular diet (RD) in obese mice gradually decreased body weight but rapidly decreased serum LECT2 levels within 10 days. In contrast, a switch from a RD to a HFD rapidly elevated serum LECT2 levels. Serum LECT2 levels showed a positive correlation with liver triglyceride contents but not with adipose tissue weight. This study demonstrates the rapid response of LECT2 preceding body weight alterations during weight cycling in mice and suggests that measurement of serum LECT2 may be clinically useful in the management of obesity.


Assuntos
Peso Corporal , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Tecido Adiposo/patologia , Adiposidade , Animais , Biomarcadores/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Insulina/sangue , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Hipernutrição/sangue , Hipernutrição/patologia
9.
J Nutr ; 144(12): 1943-51, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25411031

RESUMO

BACKGROUND: Intrauterine growth restriction (IUGR) is intimately linked with postnatal catch-up growth, leading to impaired lung structure and function. However, the impact of catch-up growth induced by early postnatal hyperalimentation (HA) on the lung has not been addressed to date. OBJECTIVE: The aim of this study was to investigate whether prevention of HA subsequent to IUGR protects the lung from 1) deregulation of the transforming growth factor-ß(TGF-ß)/bone morphogenetic protein (BMP) pathway, 2) activation of interleukin (IL)-6 signaling, and 3) profibrotic processes. METHODS: IUGR was induced in Wistar rats by isocaloric protein restriction during gestation by feeding a control (Co) or a low-protein diet with 17% or 8% casein, respectively. On postnatal day 1 (P1), litters from both groups were randomly reduced to 6 pups per dam to induce HA or adjusted to 10 pups and fed with standard diet: Co, Co with HA (Co-HA), IUGR, and IUGR with HA (IUGR-HA). RESULTS: Birth weights in rats after IUGR were lower than in Co rats (P < 0.05). HA during lactation led to accelerated body weight gain from P1 to P23 (Co vs. Co-HA, IUGR vs. IUGR-HA; P < 0.05). At P70, prevention of HA after IUGR protected against the following: 1) activation of both TGF-ß [phosphorylated SMAD (pSMAD) 2; plasminogen activator inhibitor 1 (Pai1)] and BMP signaling [pSMAD1; inhibitor of differentiation (Id1)] compared with Co (P < 0.05) and Co or IUGR (P < 0.05) rats, respectively; 2) greater mRNA expression of interleukin (Il) 6 and Il13 (P < 0.05) as well as activation of signal transducer and activator of transcription 3 (STAT3) signaling (P < 0.05) after IUGR-HA; and 3) greater gene expression of collagen Iα1 and osteopontin (P < 0.05) and increased deposition of bronchial subepithelial connective tissue in IUGR-HA compared with Co and IUGR rats. Moreover, HA had a significant additive effect (P < 0.05) on the increased enhanced pause (indicator of airway resistance) in the IUGR group (P < 0.05) at P70. CONCLUSIONS: This study demonstrates a dual mechanism in IUGR-associated lung disease that is 1) IUGR-dependent and 2) HA-mediated and thereby offers new avenues to develop innovative preventive strategies for perinatal programming of adult lung diseases.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Interleucina-6/metabolismo , Pulmão/crescimento & desenvolvimento , Hipernutrição/prevenção & controle , Fator de Crescimento Transformador beta/metabolismo , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Proteínas Morfogenéticas Ósseas/genética , Dieta com Restrição de Proteínas , Feminino , Retardo do Crescimento Fetal/terapia , Regulação da Expressão Gênica , Interleucina-6/genética , Lactação , Pulmão/patologia , Pneumopatias/prevenção & controle , Masculino , Hipernutrição/patologia , Ratos , Ratos Wistar , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Aumento de Peso/efeitos dos fármacos
10.
Pediatr Res ; 75(2): 259-65, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24232634

RESUMO

BACKGROUND: We evaluated the influence of postnatal early overnutrition on renal pathophysiological changes in aging rats. METHODS: Three or 10 male pups per mother were assigned to either the small litter (SL) or normal litter (control) groups, respectively, during the first 21 d of life. The effects of early postnatal overnutrition were determined at 12 mo. RESULTS: SL rats weighed more than controls between 4 d and 6 mo of age (P < 0.05). However, between 6 and 12 mo, body weights in both groups were not different. In the SL group, at 12 mo, systolic blood pressure was higher and creatinine clearance was lower than the same in controls (P < 0.05). Numbers of CD68 (ED1)-positive macrophages and apoptotic cells in renal cortex were higher in SL rats (P < 0.05). Furthermore, index scores for glomerulosclerosis and tubulointerstitial fibrosis were higher in the SL group (P < 0.05). Significantly less glomeruli per section area were found in aging SL rats (P < 0.05). Immunoblotting and immunohistochemistry showed decreased intrarenal renin expression in SL rats (P < 0.05). CONCLUSION: Early postnatal overnutrition can potentiate structural and functional abnormalities in the aging kidney and can lead to systolic hypertension with reduced intrarenal renin activity.


Assuntos
Envelhecimento , Nefropatias/patologia , Rim/patologia , Hipernutrição/patologia , Ciências da Nutrição Animal , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Apoptose , Peso Corporal , Creatinina/sangue , Regulação da Expressão Gênica , Glomérulos Renais/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Ratos , Renina/metabolismo , Sístole , Fatores de Tempo
11.
Horm Metab Res ; 46(4): 259-68, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24627101

RESUMO

Childhood obesity is growing in prevalence. Obesity and bone dysfunctions may be related disorders, and therefore our aim was to study the impact of the early overfeeding (EO) in offspring bone health since weaning up to adulthood. To induce EO during lactation, litter size was adjusted to 3 male rats per litter (SL). Litter containing 10 pups per mother was the control (NL). Bone tissue was evaluated by dual-energy X-ray absorptiometry, computed tomography, microcomputed tomography, biomechanical tests, and serum analyses. SL offspring presented higher body weight, fat mass, lean mass from 21 up to 180 days, hyperphagia, and higher visceral fat mass. Bone analysis showed that SL offspring presented higher total bone mineral density (BMD) only at 180 days, and higher total bone mineral content and higher bone area from 21 until 180 days. At 180 days, SL offspring presented higher femur BMD and fourth lumbar vertebra (LV4) BMD, higher femoral head radiodensity and LV4 vertebral body radiodensity, lower trabecular pattern factor and trabecular separation, however with higher trabecular number, higher maximal load, resilience, stiffness and break load, and lower break deformation. SL group had, at 180 days, higher osteocalcin and lower C-terminal cross-linked telopeptide of type I collagen (CTX I). We have shown that the excess of fat mass contributed to an increased bone mass, and hypothesized that this increase could be mediated by the hypothyroidism and previous higher thyroid hormone action and hyperleptinemia at weaning. Furthermore, the increased biomechanical loading due to increased body weight probably help us to understand the protective effects obesity exerts upon bone health.


Assuntos
Comportamento Alimentar , Fêmur/patologia , Fêmur/fisiopatologia , Hipernutrição/patologia , Hipernutrição/fisiopatologia , Absorciometria de Fóton , Animais , Animais Recém-Nascidos , Fenômenos Biomecânicos , Peso Corporal , Densidade Óssea , Colágeno Tipo I/metabolismo , Feminino , Fêmur/diagnóstico por imagem , Gordura Intra-Abdominal/fisiopatologia , Masculino , Tamanho do Órgão , Osteocalcina/metabolismo , Hipernutrição/diagnóstico por imagem , Peptídeos/metabolismo , Ratos Wistar , Desmame , Suporte de Carga , Microtomografia por Raio-X
12.
J Neurosci ; 32(33): 11486-94, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22895731

RESUMO

It is increasingly accepted that alterations of the early life environment may have lasting impacts on physiological functions. In particular, epidemiological and animal studies have indicated that changes in growth and nutrition during childhood and adolescence can impair reproductive function. However, the precise biological mechanisms that underlie these programming effects of neonatal nutrition on reproduction are still poorly understood. Here, we used a mouse model of divergent litter size to investigate the effects of early postnatal overnutrition and undernutrition on the maturation of hypothalamic circuits involved in reproductive function. Neonatally undernourished females display attenuated postnatal growth associated with delayed puberty and defective development of axonal projections from the arcuate nucleus to the preoptic region. These alterations persist into adulthood and specifically affect the organization of neural projections containing kisspeptin, a key neuropeptide involved in pubertal activation and fertility. Neonatal overfeeding also perturbs the development of neural projections from the arcuate nucleus to the preoptic region, but it does not result in alterations in kisspeptin projections. These studies indicate that alterations in the early nutritional environment cause lasting and deleterious effects on the organization of neural circuits involved in the control of reproduction, and that these changes are associated with lifelong functional perturbations.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipotálamo/citologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Estado Nutricional/fisiologia , Reprodução/fisiologia , Fatores Etários , Animais , Animais Recém-Nascidos , Peso Corporal , Mapeamento Encefálico , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/crescimento & desenvolvimento , Kisspeptinas/metabolismo , Tamanho da Ninhada de Vivíparos , Hormônio Luteinizante/metabolismo , Masculino , Desnutrição/metabolismo , Desnutrição/patologia , Camundongos , Fibras Nervosas/fisiologia , Neurocinina B/metabolismo , Neurônios/metabolismo , Ovariectomia , Hipernutrição/metabolismo , Hipernutrição/patologia , Fatores Sexuais
13.
Clin Exp Pharmacol Physiol ; 40(11): 803-16, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24033542

RESUMO

With the worldwide obesity epidemic, the proportion of women entering pregnancy overweight or obese has increased significantly in recent years. Babies born to obese women are at an increased risk of respiratory complications at birth and in childhood. In addition to maternal diabetes, there are a number of metabolic changes that the fetus of an overnourished mother experiences in utero that may modulate lung development and represent the mechanisms underlying the increased risk of respiratory complications. Herein we highlight a series of factors associated with the intrauterine environment of an overnourished mother that may impact on fetal lung development and lead to an increased risk of complications at birth or in postnatal life.


Assuntos
Modelos Animais de Doenças , Desenvolvimento Fetal , Pulmão/embriologia , Fenômenos Fisiológicos da Nutrição Materna , Hipernutrição/fisiopatologia , Animais , Secreções Corporais/metabolismo , Criança , Desenvolvimento Infantil , Pré-Escolar , Diabetes Gestacional/epidemiologia , Diabetes Gestacional/etiologia , Feminino , Humanos , Lactente , Recém-Nascido , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Pulmão/patologia , Masculino , Obesidade/patologia , Obesidade/fisiopatologia , Hipernutrição/patologia , Gravidez , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Transtornos Respiratórios/epidemiologia , Transtornos Respiratórios/etiologia , Risco
14.
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
15.
Am J Physiol Endocrinol Metab ; 302(12): E1586-98, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22510708

RESUMO

Both overnutrition and an incorrect nutrient balance have contributed to the rise in obesity. Moreover, it is now clear that poor nutrition during early life augments the possibility of excess weight gain in later years. Our aim was to determine how neonatal overnutrition affects later responses to a sucrose-enriched diet and whether this varies depending upon when the diet is introduced in postnatal life. Male Wistar rats raised in litters of four or 12 pups were given a 33% sucrose solution instead of water from weaning (day 21) or postnatal day (PND) 65. All rats received normal chow ad libitum until they were euthanized on PND 80. Body weight (BW) and food and liquid intake were monitored throughout the study. Fat mass, adipocyte morphology, serum biochemical and hormonal parameters, and hypothalamic neuropeptide mRNA levels were measured at study termination. Neonatal overnutrition increased food intake, BW, and leptin levels, induced adipocyte hypertrophy, and decreased total ghrelin levels. The sucrose-enriched diet increased total energy intake, adipose accrual, and leptin, adiponectin, and acylated ghrelin levels but decreased BW. Most of these responses were accentuated in neonatally overnourished rats, which also had increased insulin and triglyceride levels. However, long-term sucrose intake induced adipocyte hypertrophy in rats from normal-sized litters but not in neonatally overfed rats. The results reported here indicate that neonatal overnutrition increases the detrimental response to a diet rich in sucrose later in life. Moreover, the timing and duration of the exposure to a sucrose-enriched diet alter the adverse metabolic outcomes.


Assuntos
Tecido Adiposo/patologia , Dieta , Sacarose Alimentar/farmacologia , Hipernutrição/patologia , Adiponectina/metabolismo , Animais , Animais Recém-Nascidos , Proteínas Sanguíneas/metabolismo , Composição Corporal/fisiologia , Colesterol/sangue , Corticosterona/metabolismo , Ingestão de Alimentos/fisiologia , Ensaio de Imunoadsorção Enzimática , Ácidos Graxos não Esterificados/sangue , Feminino , Grelina/metabolismo , Teste de Tolerância a Glucose , Glicerol/sangue , Insulina/metabolismo , Leptina/metabolismo , Masculino , Hipernutrição/psicologia , Gravidez , Radioimunoensaio , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Aumento de Peso/fisiologia
16.
Microvasc Res ; 84(1): 94-8, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22484032

RESUMO

The aims of our study were to investigate effects of postnatal overnutrition, obtained by restricting the number of pups per litter, on microcirculatory reactivity, fat depots, its total percentage and lipid profile. Microvascular reactivity was evaluated in the cremaster muscle of 24 hamsters divided into four groups, with 6 animals in each one: normal (NL) and restricted (RL) litter groups, both at 6th and 21st weeks of age. The NL group had 8-9 pups and the RL 3 pups per litter and to avoid the litter effect, only one animal was used per litter. The results have shown that the RL group had higher velocity of weight, body mass and fat gain compared to the NL one at weeks 6 and 21. Significant differences were also observed on urogenital fat depot, total cholesterol and low density lipoprotein between groups. At the lowest concentration of Ach, the RL group showed smaller arteriolar dilatation at the 21st than at the 6th week [5(3-13) vs 19(8-40)%, p<0.01] while the NL one did not show any difference within the group. The highest concentration of Ach at the 21th week pointed to endothelial-dependent microvascular dysfunction in RL compared to NL [3(8-26) vs. 13(8-26)%, p<0.05]. Endothelial-independent microvascular reactivity was similar between groups. Our data suggest that postnatal overnutrition is associated to muscle endothelial-dependent microvascular dysfunction, greater body mass and total percentage of fat and impaired the lipid profile. In conclusion, the imprinting promoted by this experimental model of obesity was able to influence microvascular reactivity later in life.


Assuntos
Arteríolas/patologia , Endotélio Vascular/patologia , Músculo Esquelético/irrigação sanguínea , Obesidade/patologia , Hipernutrição/patologia , Tecido Adiposo/patologia , Animais , Arteríolas/fisiopatologia , Tamanho Corporal , Cricetinae , Endotélio Vascular/fisiopatologia , Feminino , Humanos , Gordura Intra-Abdominal , Tamanho da Ninhada de Vivíparos , Masculino , Mesocricetus , Obesidade/fisiopatologia , Hipernutrição/fisiopatologia , Aumento de Peso
17.
Br J Nutr ; 107(4): 510-22, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21733295

RESUMO

Both high and low maternal dietary intakes adversely affect fetal nutrient supply in adolescent sheep pregnancies. Aims were: (a) to assess the impact of prenatal nutrition on pregnancy outcome, offspring growth and offspring glucose metabolism and (b) to determine whether the offspring metabolic phenotype could then be altered by modifying postnatal nutrition. Dams carrying a single fetus were offered either an optimal control (C) intake to maintain adiposity throughout pregnancy, undernourished to maintain weight at conception but deplete maternal reserves (UN), or overnourished to promote rapid maternal growth and adiposity (ON). Placental weight and gestation length were reduced in ON dams and lamb birth weights were C>UN>ON (P < 0·001). All offspring were fed ad libitum from weaning to 6 months of age. ON offspring exhibited rapid catch-up growth and had increased fasting glucose and relative glucose intolerance compared with C offspring (P < 0·05). Irrespective of prenatal diet and sex, birth weight correlated negatively with these indices of glucose metabolism. From 7 to 12 months offspring either had continued ad libitum diet (ADLIB; to induce an obesogenic state) or a decreased ration appropriate for normal growth (NORM). At 12 months, the negative relationship between birth weight and indices of glucose metabolism persisted in ADLIB females (for example, fasting glucose, r - 0·632; P < 0·03) but was absent in NORM females and in both male groups. Therefore, low-birth-weight offspring from differentially achieved prenatal malnutrition exhibit an early adverse metabolic phenotype, and this can apparently be ameliorated by postnatal nutrition in females but not in males.


Assuntos
Retardo do Crescimento Fetal/dietoterapia , Desnutrição/fisiopatologia , Fenômenos Fisiológicos da Nutrição Materna , Hipernutrição/fisiopatologia , Animais , Animais Recém-Nascidos , Peso ao Nascer , Cruzamentos Genéticos , Transferência Embrionária , Feminino , Desenvolvimento Fetal , Retardo do Crescimento Fetal/sangue , Retardo do Crescimento Fetal/etiologia , Intolerância à Glucose/etiologia , Hiperglicemia/etiologia , Masculino , Desnutrição/patologia , Tamanho do Órgão , Hipernutrição/patologia , Placenta/patologia , Gravidez , Nascimento Prematuro/etiologia , Caracteres Sexuais , Carneiro Doméstico
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA