Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
FASEB J ; 33(12): 14296-14306, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31645130

RESUMO

Growing evidence has suggested that autophagy-related protein 7 (ATG7) plays an important role in insulin signaling, but the mechanism of ATG7 in hepatic insulin sensitivity is not fully understood. The purpose of the present study is to clarify the underlying molecular mechanisms of ATG7 in obesity development. Serum and liver samples from mice fed a high fat diet (HFD) were evaluated for metabolic profile data and ATG expressions during obesity development. We found that compared with other ATGs, ATG7 expression increased earlier with lower hepatic insulin sensitivity in the 4-wk HFD-fed mice. For in vitro analyses, silencing ATG7 significantly up-regulated insulin-stimulated phosphorylation of protein kinase B (Akt) and down-regulated phosphatase and tension homolog deleted on chromosome ten (PTEN) in HepG2 cells. Replenishing PTEN to ATG7-silenced hepatocytes restored the phosphorylated Akt level. Furthermore, ATG7 silencing led to higher c-JUN expression, which transcriptionally reduced PTEN expression. These results reveal a novel mechanism by which ATG7 regulates Akt phosphorylation via the c-JUN/PTEN pathway at the early stage of HFD-induced metabolic disorder.-Zhao, D., Zhang, S., Wang, X., Gao, D., Liu, J., Cao, K., Chen, L., Liu, R., Liu, J., Long, J. ATG7 regulates hepatic Akt phosphorylation through the c-JUN/PTEN pathway in high fat diet-induced metabolic disorder.


Assuntos
Proteína 7 Relacionada à Autofagia/metabolismo , Dieta Hiperlipídica/efeitos adversos , Fígado/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proteína 7 Relacionada à Autofagia/genética , Gorduras na Dieta/efeitos adversos , Regulação para Baixo , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Células Hep G2 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Doenças Metabólicas/induzido quimicamente , Doenças Metabólicas/metabolismo , Camundongos , Células PC-3 , PTEN Fosfo-Hidrolase/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética
2.
J Cell Physiol ; 234(3): 2566-2580, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30264458

RESUMO

Obesity is associated with skeletal muscle loss and impaired myogenesis. Increased infiltration of proinflammatory macrophages in skeletal muscle is noted in obesity and is associated with muscle insulin resistance. However, whether the infiltrated macrophages can contribute to obesity-induced muscle loss is unclear. In this study, we investigate macrophage and muscle differentiation markers in the quadriceps (QC), gastrocnemius, tibia anterior, and soleus muscles from obese mice that were fed a high-fat diet for 16 weeks. Then, we examined the effect and mediator of macrophage-secreted factors on myoblast differentiation in vitro. We found markedly increased levels of proinflammatory macrophage markers (F4/80 and CD11c) in the QC muscle compared with the other three muscle groups. Consistent with the increased levels of proinflammatory macrophage infiltration, the QC muscle also showed a significant reduction in the expression of muscle differentiation makers MYOD1 and myosin heavy chain. In in vitro studies, treatment of C2C12 myoblasts with Raw 264.7 macrophage-conditioned medium (CM) significantly promoted cell proliferation and inhibited myoblast differentiation. Neutralization of tumor necrosis factor α (TNF-α) in Raw 264.7 macrophage CM reversed the reduction of myoblast differentiation. Finally, we found that both macrophage CM and TNF-α induced sustained activation of p38 mitogen-activated protein kinase (MAPK) in C2C12 myoblasts. Together, our findings suggest that the increased infiltration of proinflammatory macrophages could contribute toward obesity-induced muscle loss by secreting inflammatory cytokine TNF-α via the p38 MAPK signaling pathway.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Muscular/fisiologia , Obesidade/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Meios de Cultivo Condicionados/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Transdução de Sinais/fisiologia
3.
Front Cell Infect Microbiol ; 12: 1074533, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36776549

RESUMO

Introduction: As a deadly disease induced by Mycobacterium tuberculosis (Mtb), tuberculosis remains one of the top killers among infectious diseases. The low intracellular Mtb killing efficiency of current antibiotics introduced the long duration anti-TB therapy in clinic with strong side effects and increased drug-resistant mutants. Therefore, the exploration of novel anti-TB agents with potent anti-TB efficiency becomes one of the most urgent issues for TB therapies. Methods: Here, we firstly introduced a novel method for the preparation of zinc oxide-selenium nanoparticles (ZnO-Se NPs) by the hybridization of zinc oxide and selenium to combine the anti-TB activities of zinc oxide nanoparticles and selenium nanoparticles. We characterized the ZnO-Se NPs by dynamic laser light scattering and transmission electron microscopy, and then tested the inhibition effects of ZnO-Se NPs on extracellular Mtb by colony-forming units (CFU) counting, bacterial ATP analysis, bacterial membrane potential analysis and scanning electron microscopy imaging. We also analyzed the effects of ZnO-Se NPs on the ROS production, mitochondrial membrane potential, apoptosis, autophagy, polarization and PI3K/Akt/mTOR signaling pathway of Mtb infected THP-1 macrophages. At last, we also tested the effects of ZnO-Se NPs on intracellular Mtb in THP-1 cells by colony-forming units (CFU) counting. Results: The obtained spherical core-shell ZnO-Se NPs with average diameters of 90 nm showed strong killing effects against extracellular Mtb, including BCG and the virulent H37Rv, by disrupting the ATP production, increasing the intracellular ROS level and destroying the membrane structures. More importantly, ZnO-Se NPs could also inhibit intracellular Mtb growth by promoting M1 polarization to increase the production of antiseptic nitric oxide and also promote apoptosis and autophagy of Mtb infected macrophages by increasing the intracellular ROS, disrupting mitochondria membrane potential and inhibiting PI3K/Akt/mTOR signaling pathway. Discussion: These ZnO-Se NPs with synergetic anti-TB efficiency by combining the Mtb killing effects and host cell immunological inhibition effects were expected to serve as novel anti-TB agents for the development of more effective anti-TB strategy.


Assuntos
Antituberculosos , Mycobacterium tuberculosis , Nanopartículas , Selênio , Óxido de Zinco , Trifosfato de Adenosina , Antituberculosos/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Nanopartículas/química , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Espécies Reativas de Oxigênio , Selênio/farmacologia , Serina-Treonina Quinases TOR , Óxido de Zinco/farmacologia , Óxido de Zinco/química
4.
J Nutr Biochem ; 88: 108503, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32956825

RESUMO

The beneficial effects of omega-3 polyunsaturated fatty acids (n-3 PUFAs) on preventing obesity are well known; however, the underlying mechanism by which n-3 PUFAs influence tricarboxylic acid (TCA) cycle under obesity remains unclear. We randomly divided male C57BL/6 mice into 5 groups (n=10) and fed for 12 weeks as follows: mice fed a normal diet (Con, 10% kcal); mice fed a high-fat diet (HFD, lard, 60% kcal); and mice fed a high-fat diet (60% kcal) substituting half the lard with safflower oil (SO), safflower oil and fish oil (SF) and fish oil (FO), respectively. Then we treated HepG2 cells with palmitic acid and DHA for 24 h. We found that body weight in FO group was significantly lower than it in HFD and SO groups. N-3 PUFAs reduced the transcription and translation of TCA cycle enzymes, including IDH1, IDH2, SDHA, FH and MDH2, to enhance mitochondrial function in vivo and vitro. DHA significantly inhibited protein expression of the mTORC1 signaling pathway, increased p-AKT protein expression to alleviate insulin resistance and improved mitochondrial oxygen consumption rate and glycolysis ability in HepG2 cells. In addition, the expressions of IDH2 and SDHB were reduced by rapamycin. N-3 PUFAs could prevent obesity by improving TCA cycle homeostasis and mTORC1 signaling pathway may be upstream.


Assuntos
Ciclo do Ácido Cítrico/efeitos dos fármacos , Ácidos Graxos Ômega-3/farmacologia , Obesidade/prevenção & controle , Animais , Sobrevivência Celular/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Ácidos Docosa-Hexaenoicos/farmacologia , Óleos de Peixe/farmacologia , Glicólise/efeitos dos fármacos , Células Hep G2 , Homeostase/efeitos dos fármacos , Humanos , Resistência à Insulina , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Obesidade/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Ácido Palmítico/farmacologia , Óleo de Cártamo/farmacologia
5.
Mol Nutr Food Res ; 65(3): e2000797, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33296142

RESUMO

SCOPE: Alzheimer's disease (AD) is the most prevalent form of dementia in the aging population; however, no effective therapy has been established. It has been previously demonstrated that daily intake of hydroxytyrosol (HT), a polyphenol in olive oil, at a daily dietary level mildly improves cognition in AD mice. In the present study, HT acetate (HT-ac), which is a natural derivative of HT in olive oil that exhibits better bioactivity than HT improves cognition. METHODS AND RESULTS: HT-ac to APP/PS1 is orally administered to transgenic mice and used Aß-treated neuronal cultures to explore the neuroprotective effects of HT-ac in preventing AD progression. It is found that HT-ac remarkably improved the escape latency, escape distance, and the number of platform crossings of AD mice in the water maze test by ameliorating neuronal apoptosis and decreasing inflammatory cytokine levels. It is further demonstrated that HT-ac stimulated the transcription of ERß and enhanced neuronal viability and electrophysiological activity in primary neurons but that these beneficial effects of HT-ac are abolished upon ERß deficiency. CONCLUSIONS: This study suggests that as the bioactive component of olive oil, HT-ac is a promising neuroprotective nutrient that may be used to alleviate AD-related cognitive dysfunction.


Assuntos
Acetatos/farmacologia , Catecóis/farmacologia , Cognição/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cognição/fisiologia , Modelos Animais de Doenças , Encefalite/tratamento farmacológico , Encefalite/patologia , Receptor beta de Estrogênio/metabolismo , Feminino , Hipocampo/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Presenilina-1/genética , Ratos Wistar
6.
Adv Sci (Weinh) ; 8(11): 2004507, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34141522

RESUMO

Mitochondrial epigenetics is rising as intriguing notion for its potential involvement in aging and diseases, while the details remain largely unexplored. Here it is shown that among the 13 mitochondrial DNA (mtDNA) encoded genes, NADH-dehydrogenase 6 (ND6) transcript is primarily decreased in obese and type 2 diabetes populations, which negatively correlates with its distinctive hypermethylation. Hepatic mtDNA sequencing in mice unveils that ND6 presents the highest methylation level, which dramatically increases under diabetic condition due to enhanced mitochondrial translocation of DNA methyltransferase 1 (DNMT1) promoted by free fatty acid through adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) activation. Hepatic knockdown of ND6 or overexpression of Dnmt1 similarly impairs mitochondrial function and induces systemic insulin resistance both in vivo and in vitro. Genetic or chemical targeting hepatic DNMT1 shows significant benefits against insulin resistance associated metabolic disorders. These findings highlight the pivotal role of ND6 epigenetic network in regulating mitochondrial function and onset of insulin resistance, shedding light on potential preventive and therapeutic strategies of insulin resistance and related metabolic disorders from a perspective of mitochondrial epigenetics.


Assuntos
Metilação de DNA/genética , DNA Mitocondrial/genética , Diabetes Mellitus Tipo 2/genética , Resistência à Insulina/genética , NADH Desidrogenase/genética , Idoso , Animais , Diabetes Mellitus Tipo 2/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
7.
Antioxid Redox Signal ; 32(3): 193-212, 2020 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-31680537

RESUMO

Aims: To study the expression and regulatory role of SOD3 in adipocytes and adipose tissue. Results: SOD3 expression was determined in various tissues of adult C57BL/6J mice, human adipose tissue and epididymal adipose tissue, subcutaneous adipose tissue and brown adipose tissue of high-fat diet (HFD)-induced obese mice. SOD3 expression and release were evaluated in adipocytes differentiated from primary human preadipocytes and murine bone marrow-derived mesenchymal stem cells (BM-MSCs). The regulatory role for SOD3 was determined by SOD3 lentivirus knockdown in human adipocytes and global sod3 knockout (KO) mice. SOD3 was expressed at high levels in white adipose tissue, and adipocytes were the main cells expressing SOD3 in adipose tissue. SOD3 expression was significantly elevated in adipose tissue of HFD-fed mice. Moreover, SOD3 expression and release were markedly increased in differentiated human adipocytes and adipocytes differentiated from mouse BM-MSCs compared with undifferentiated cells. In addition, SOD3 silencing in human adipocytes increased expression of genes involved in lipid metabolic pathways such as PPARγ and SREBP1c and promoted the accumulation of triglycerides. Finally, global sod3 KO mice were more obese and insulin resistant with enlarged adipose tissue and increased triglyceride accumulation. Innovation: Our data showed that SOD3 is secreted from adipocytes and regulates lipid metabolism in adipose tissue. This important discovery may open up new avenues of research for the cytoprotective role of SOD3 in obesity and its associated metabolic disorders. Conclusion: SOD3 is a protective factor secreted by adipocytes in response to HFD-induced obesity and regulates adipose tissue lipid metabolism.


Assuntos
Adipócitos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Inflamação/metabolismo , Resistência à Insulina/fisiologia , Insulina/metabolismo , Obesidade/metabolismo , Superóxido Dismutase/metabolismo , Tecido Adiposo/metabolismo , Adulto , Animais , Diferenciação Celular/fisiologia , Feminino , Humanos , Metabolismo dos Lipídeos/fisiologia , Redes e Vias Metabólicas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , PPAR gama/metabolismo , Triglicerídeos/metabolismo
8.
Neurobiol Aging ; 56: 115-126, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28528772

RESUMO

Alzheimer's disease (AD) is considered a multifactorial disease that affects the central nervous system and periphery. A decline in brain glucose metabolism is an early feature of AD and is accompanied by a phenotypic shift from aerobic glycolysis to ketogenesis. The liver is responsible for the generation of the ketone body. However, the mechanism that underlies hepatic ketogenesis in AD remains unclear. Here, we investigated hepatic ketogenesis during the early stage of AD pathogenesis in amyloid precursor protein (APPSWE) and presenilin (PSEN1dE9) (APP/PS1) mice. We observed that ß-hydroxybutyric acid was increased in the brain of the postmortem mild cognitive impairment and AD subjects and in 3-month-old APP/PS1 AD mice. A rise in 3-hydroxy-3-methylglutary-CoA synthase 2 (HMGCS2), a key enzyme for catalyzing ß-hydroxybutyric acid production, was observed in early AD mice. We further showed that proinflammatory cytokines were activated in the liver prior to their activation in the brain of 3-month-old APP/PS1 mice. Among the cytokines, interleukin-6 significantly activated HMGCS2 through the binding of nuclear factor κB (NF-κB) p65 to the HMGCS2 promoter. Additionally, interleukin-6 stimulated phosphorylation of p38 mitogen activated protein kinases, an upstream molecule for NF-κB p65 signaling. We have demonstrated that a hepatic inflammatory factor enhances ketogenesis through HMGCS2 signaling activation by p38/NF-κB p65. These results provide a novel peripheral metabolic mechanism for enhanced ketone production and suggest a plausible early AD phenotype to diagnose AD.


Assuntos
Hidroximetilglutaril-CoA Sintase/metabolismo , Interleucina-6/fisiologia , Corpos Cetônicos/metabolismo , Fígado/metabolismo , NF-kappa B/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/metabolismo , Animais , Biomarcadores/metabolismo , Encéfalo/metabolismo , Hidroxibutiratos/metabolismo , Camundongos Transgênicos , Fosforilação/genética
9.
Mol Nutr Food Res ; 61(3)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27615442

RESUMO

SCOPE: Apple pomace polysaccharides (APP), a free radical scavenger, is one of the major compounds derived from apple pomace. However, whether APP has beneficial effects on metabolic disorders is still unknown. METHODS AND RESULTS: In the present study, water-soluble APP was isolated from the pomace of the locally abundant "Qinguan" apple and chemically characterized. Then, APP was orally administrated to high-fat diet (HFD)-induced obese mice. We found that APP significantly reduced HFD-induced body weight gain and ameliorated HFD-induced hepatic metabolic disorders and oxidative stress. In a palmitate-loaded HepG2 cell model, APP protected the cells from palmitate-induced insulin resistance and loss of viability by suppressing mitochondrial reactive oxygen species and rescuing mitochondrial respiratory function. CONCLUSION: Our work suggests that APP, a promising bioactive food component, successfully improved obesity-associated hepatic metabolic disorder, most likely though the activation of hepatic mitochondrial function and the suppression of mitochondria oxidative stress.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Fígado/efeitos dos fármacos , Malus/química , Obesidade/dietoterapia , Polissacarídeos/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Feminino , Células Hep G2 , Humanos , Resistência à Insulina , Fígado/metabolismo , Fígado/fisiopatologia , Masculino , Camundongos Obesos , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Obesidade/etiologia , Estresse Oxidativo , Palmitatos/efeitos adversos , Polissacarídeos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA