Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Obes (Lond) ; 48(7): 941-953, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38424257

RESUMO

OBJECTIVE: In our previous study, we identified a notable increase in miR-548ag content after obesity, which contributes to the progression of Type 2 diabetes Mellitus(T2DM) through the up-regulation of Dipeptidyl Peptidase-4(DPP4) expression within the liver. However, the precise molecular mechanisms underlying the upregulation of DPP4 by miR-548ag remain elusive. Mature miRNAs rich in GU sequences can activate the TLR(7/8)/NF-κB signalling pathway, which transcriptionally activates DPP4 expression. Notably, the proportion of GU sequences in hsa-miR-548ag was found to be 47.6%. The study proposes a hypothesis suggesting that miR-548ag could potentially increase DPP4 expression in hepatocytes by activating the TLR(7/8)/NF-κB signalling pathway. METHODS: Male C57BL/6J mice were fed normal chow diet (NCD, n = 16) or high-fat diet (HFD, n = 16) for 12 weeks. For a duration of 6 weeks, NCD mice received intraperitoneal injections of a miR-548ag mimic, while HFD mice and db/db mice (n = 16) were administered intraperitoneal injections of a miR-548ag inhibitor. qRT-PCR and Western Blot were used to detect the expression level of miR-548ag, DPP4 and the activation of TLR(7/8)/NF-κB signalling pathway. HepG2 and L02 cells were transfected with miR-548ag mimic, miR-548ag inhibitor, TLR7/8 interfering fragment, and overexpression of miR-548ag while inhibiting TLR7/8, respectively. RESULTS: (1) We observed elevated levels of miR-548ag in the serum, adipose tissue, and liver of obese mice, accompanied by an upregulation of TLR7/8, pivotal protein in the NF-κB pathway, and DPP4 expression in the liver. (2) miR-548ag promotes DPP4 expression in hepatocytes via the TLR(7/8)/NF-κB signalling pathway, resulting in a reduction in the glucose consumption capacity of hepatocytes. (3) The administration of a miR-548ag inhibitor enhanced glucose tolerance and insulin sensitivity in db/db mice. CONCLUSIONS: MiR-548ag promotes the expression of DPP4 in hepatocytes by activating the TLR(7/8)/NF-κB signalling pathway. MiR-548ag may be a potential target for the treatment of T2DM.


Assuntos
Dipeptidil Peptidase 4 , Hepatócitos , Camundongos Endogâmicos C57BL , MicroRNAs , NF-kappa B , Transdução de Sinais , Animais , Camundongos , Masculino , MicroRNAs/metabolismo , MicroRNAs/genética , Hepatócitos/metabolismo , Dipeptidil Peptidase 4/metabolismo , Dipeptidil Peptidase 4/genética , NF-kappa B/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Obesidade/metabolismo , Obesidade/genética , Dieta Hiperlipídica , Modelos Animais de Doenças , Regulação para Cima , Receptor 7 Toll-Like/metabolismo , Receptor 7 Toll-Like/genética
2.
FASEB J ; 37(7): e23033, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37342904

RESUMO

In the obesity context, inflammatory cytokines secreted by adipocytes lead to insulin resistance and are key to metabolic syndrome development. In our previous study, we found that the transcription factor KLF7 promoted the expression of p-p65 and IL-6 in adipocytes. However, the specific molecular mechanism remained unclear. In the present study, we found that the expression of KLF7, PKCζ, p-IκB, p-p65, and IL-6 in epididymal white adipose tissue (Epi WAT) in mice fed a high-fat diet (HFD) was significantly increased. In contrast, the expression of PKCζ, p-IκB, p-p65, and IL-6 was significantly decreased in Epi WAT of KLF7 fat conditional knockout mice. In 3T3-L1 adipocytes, KLF7 promoted the expression of IL-6 via the PKCζ/NF-κB pathway. In addition, we performed luciferase reporter and chromatin immunoprecipitation assays, which confirmed that KLF7 upregulated the expression of PKCζ transcripts in HEK-293T cells. Collectively, our results show that KLF7 promotes the expression of IL-6 by upregulating PKCζ expression and activating the NF-κB signaling pathway in adipocytes.


Assuntos
Transtornos do Metabolismo de Glucose , NF-kappa B , Animais , Camundongos , Células 3T3-L1 , Adipócitos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Transtornos do Metabolismo de Glucose/metabolismo , Proteínas I-kappa B/metabolismo , Inflamação/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , NF-kappa B/metabolismo
3.
Int J Mol Sci ; 24(3)2023 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-36769291

RESUMO

The present study aimed to explore the molecular mechanism underlying the regulation of glucose metabolism by miR-548ag. For the first time, we found that miR-548ag expression was elevated in the abdominal adipose tissue and serum of subjects with obesity and type 2 diabetes mellitus (T2DM). The conditional knockout of adipose tissue Dicer notably reduced the expression and content of miR-548ag in mouse adipose tissue, serum, and liver tissue. The combined use of RNAseq, an miRNA target gene prediction software, and the dual luciferase reporter assay confirmed that miR-548ag exerts a targeted regulatory effect on DNMT3B and DPP4. miR-548ag and DPP4 expression was increased in the adipose tissue, serum, and liver tissue of diet-induced obese mice, while DNMT3B expression was decreased. It was subsequently confirmed both in vitro and in vivo that adipose tissue-derived miR-548ag impaired glucose tolerance and insulin sensitivity by inhibiting DNMT3B and upregulating DPP4. Moreover, miR-548ag inhibitors significantly improved the adverse metabolic phenotype in both obese mice and db/db mice. These results revealed that the expression of the adipose tissue-derived miR-548ag increased in obese subjects, and that this could upregulate the expression of DPP4 by targeting DNMT3B, ultimately leading to glucose metabolism disorder. Therefore, miR-548ag could be utilized as a potential target in the treatment of T2DM.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , MicroRNAs , Camundongos , Animais , Dipeptidil Peptidase 4/genética , Dipeptidil Peptidase 4/metabolismo , Regulação para Cima , Camundongos Obesos , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Tecido Adiposo/metabolismo , Fígado/metabolismo , Obesidade/genética , Obesidade/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Resistência à Insulina/genética , Camundongos Endogâmicos C57BL
4.
Development ; 146(2)2019 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-30635284

RESUMO

Protein modification by ubiquitin and ubiquitin-like proteins (UBLs) regulates numerous biological functions. The UFM1 system, a novel UBL conjugation system, is implicated in mouse development and hematopoiesis. However, its broad biological functions and working mechanisms remain largely elusive. CDK5RAP3, a possible ufmylation substrate, is essential for epiboly and gastrulation in zebrafish. Herein, we report a crucial role of CDK5RAP3 in liver development and hepatic functions. Cdk5rap3 knockout mice displayed prenatal lethality with severe liver hypoplasia, as characterized by delayed proliferation and compromised differentiation. Hepatocyte-specific Cdk5rap3 knockout mice suffered post-weaning lethality, owing to serious hypoglycemia and impaired lipid metabolism. Depletion of CDK5RAP3 triggered endoplasmic reticulum stress and activated unfolded protein responses in hepatocytes. We detected the in vivo interaction of CDK5RAP3 with UFL1, the defined E3 ligase in ufmylation. Notably, loss of CDK5RAP3 altered the ufmylation profile in liver cells, suggesting that CDK5RAP3 serves as a novel substrate adaptor for this UBL modification. Collectively, our study identifies CDK5RAP3 as an important regulator of ufmylation and suggests the involvement of ufmylation in mammalian development.


Assuntos
Fígado/embriologia , Fígado/metabolismo , Proteínas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Proliferação de Células , Perda do Embrião/patologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Retículo Endoplasmático/metabolismo , Deleção de Genes , Células Hep G2 , Hepatócitos/citologia , Hepatócitos/metabolismo , Homeostase , Humanos , Fígado/patologia , Camundongos Knockout , Ligação Proteica , Especificidade por Substrato , Proteínas Supressoras de Tumor
5.
Acta Pharmacol Sin ; 43(9): 2362-2372, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35105957

RESUMO

Bile acid (BA) homeostasis is regulated by the extensive cross-talk between liver and intestine. Many bile-acid-activated signaling pathways have become attractive therapeutic targets for the treatment of metabolic disorders. In this study we investigated the regulatory mechanisms of BA in the intestine. We showed that the BA levels in the gallbladder and faeces were significantly increased, whereas serum BA levels decreased in systemic Krüppel-like factor 9 (Klf9) deficiency (Klf9-/-) mice. These phenotypes were also observed in the intestine-specific Klf9-deleted (Klf9vil-/-) mice. In contrast, BA levels in the gallbladder and faeces were reduced, whereas BA levels in the serum were increased in intestinal Klf9 transgenic (Klf9Rosa26+/+) mice. By using a combination of biochemical, molecular and functional assays, we revealed that Klf9 promoted the expression of apical sodium-dependent bile acid transporter (Asbt) in the terminal ileum to enhance BA absorption in the intestine. Reabsorbed BA affected liver BA synthetic enzymes by regulating Fgf15 expression. This study has identified a previously neglected transcriptional pathway that regulates BA homeostasis.


Assuntos
Ácidos e Sais Biliares , Fatores de Transcrição Kruppel-Like/metabolismo , Simportadores , Animais , Ácidos e Sais Biliares/metabolismo , Circulação Êntero-Hepática , Intestinos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Simportadores/metabolismo , Fatores de Transcrição/metabolismo
6.
Gut ; 70(11): 2183-2195, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33257471

RESUMO

OBJECTIVE: Impaired hepatic fatty acids oxidation results in lipid accumulation and redox imbalance, promoting the development of fatty liver diseases and insulin resistance. However, the underlying pathogenic mechanism is poorly understood. Krüppel-like factor 16 (KLF16) is a transcription factor that abounds in liver. We explored whether and by what mechanisms KLF16 affects hepatic lipid catabolism to improve hepatosteatosis and insulin resistance. DESIGN: KLF16 expression was determined in patients with non-alcoholic fatty liver disease (NAFLD) and mice models. The role of KLF16 in the regulation of lipid metabolism was investigated using hepatocyte-specific KLF16-deficient mice fed a high-fat diet (HFD) or using an adenovirus/adeno-associated virus to alter KLF16 expression in mouse primary hepatocytes (MPHs) and in vivo livers. RNA-seq, luciferase reporter gene assay and ChIP analysis served to explore the molecular mechanisms involved. RESULTS: KLF16 expression was decreased in patients with NAFLD, mice models and oleic acid and palmitic acid (OA and PA) cochallenged hepatocytes. Hepatic KLF16 knockout impaired fatty acid oxidation, aggravated mitochondrial stress, ROS burden, advancing hepatic steatosis and insulin resistance. Conversely, KLF16 overexpression reduced lipid deposition and improved insulin resistance via directly binding the promoter of peroxisome proliferator-activated receptor α (PPARα) to accelerate fatty acids oxidation and attenuate mitochondrial stress, oxidative stress in db/db and HFD mice. PPARα deficiency diminished the KLF16-evoked protective effects against lipid deposition in MPHs. Hepatic-specific PPARα overexpression effectively rescued KLF16 deficiency-induced hepatic steatosis, altered redox balance and insulin resistance. CONCLUSIONS: These findings prove that a direct KLF16-PPARα pathway closely links hepatic lipid homeostasis and redox balance, whose dysfunction promotes insulin resistance and hepatic steatosis.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , PPAR alfa/metabolismo , Animais , Biomarcadores/sangue , Hepatócitos/metabolismo , Humanos , Resistência à Insulina , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Sheng Li Xue Bao ; 73(5): 772-780, 2021 Oct 25.
Artigo em Zh | MEDLINE | ID: mdl-34708234

RESUMO

The development of nonalcoholic fatty liver disease (NAFLD) is closely related to the fatty acid (FA) uptake. This study aimed to investigate the effect of Krüppel-like factor 9 (KLF9) on CD36 (typical fatty acid translocase), hepatocellular lipid metabolism as well as the development and progression of nonalcoholic fatty liver. High-fat diet-induced obese C57BL/6J mice and db/db mice were used to test the expression levels of Klf9 and Cd36 in the livers. The primary hepatocytes were isolated from C57BL/6J mice, treated with Ad-GFP, Ad-Klf9, Ad-shCtrl or Ad-shKlf9, and then incubated with oleic acid and palmitic acid for 24 h. Liver-specific knockout of Klf9 mice were established. The protein levels and relative mRNA levels were examined by Western blot and real-time PCR, respectively. Triglyceride content was determined by using an assay kit. Lipid content was determined by Oil Red O staining. The results showed that: (1) Klf9 expression levels were increased in the livers of high-fat diet-induced obese mice and db/db mice, compared to their respective control mice. (2) Adenovirus-mediated overexpression of Klf9 in primary hepatocytes increased Cd36 expression and cellular triglyceride contents. (3) In contrast, adenovirus-mediated knockdown of Klf9 expression in primary hepatocytes by Ad-shKlf9 decreased Cd36 expression and cellular triglyceride contents. (4) Finally, Klf9 deficiency decreased liver Cd36 expression and alleviated fatty liver phenotype of high-fat diet-induced obese mice. These results suggest that KLF9 can regulate hepatic lipid metabolism and development of NAFLD by promoting the expression of CD36.


Assuntos
Metabolismo dos Lipídeos , Hepatopatia Gordurosa não Alcoólica , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Dieta Hiperlipídica , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ácido Oleico/metabolismo
8.
Sheng Li Xue Bao ; 73(5): 821-827, 2021 Oct 25.
Artigo em Zh | MEDLINE | ID: mdl-34708239

RESUMO

ß3-adrenergic agonists induce adaptive thermogenesis and promote beiging of white fat. However, it remains unclear which metabolites mediate the stimulatory effects of ß3-adrenergic agonists on thermogenesis of brown and beige fat. In this study, adipose tissue was isolated from 8-week-old C57/BL6J male mice by intraperitoneal administration of ß3-adrenergic agonist CL316,243 for RNA-Seq, which revealed that histidine decarboxylase, a key enzyme in histamine synthesis, was strongly induced in adipose by CL316,243. Therefore, we speculated that histamine might be involved in the process of thermogenesis in adipose tissue. We determined the physiological role and mechanism by which histamine promotes fat thermogenesis by intravenous administering histamine to C57BL/6J mice fed a normal or a high-fat diet. The results showed that intravenous injection of histamine into C57BL/6J mice fed a normal diet stimulated the expression of thermogenic genes, including peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and uncoupling protein 1 (UCP1), in brown adipose tissue (BAT) and inguinal white adipose tissue (iWAT). H&E staining also suggested that histamine treatment decreased the size of lipid droplets in adipocytes. Moreover, histamine treatment also enhanced thermogenesis of fat in high-fat diet induced obese mice, and improved glucose intolerance and fatty liver phenotype. Finally, we demonstrated that the effects of histamine on the thermogenic program were cell autonomous. Our data suggest that histamine may mediate the effects of ß3-adrenergic agonists on thermogenesis of fat.


Assuntos
Tecido Adiposo Bege , Histamina , Tecido Adiposo Marrom , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Termogênese , Proteína Desacopladora 1/genética
9.
New Phytol ; 223(2): 705-721, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30891753

RESUMO

Wild and cultivated rice show a significant difference in anthocyanin biosynthesis in the leaf. The regulation system of anthocyanin biosynthesis in rice leaf and the causal mechanism of the difference in this biosynthesis between wild and cultivated rice remain largely unknown. In this study, a genome-wide association study and transcriptome analysis were performed to identify the determinant factors and dissect the regulatory system for anthocyanin biosynthesis in rice leaves. OsC1, OsRb and OsDFR were identified as the determinants of anthocyanin biosynthesis in rice leaves. Artificial selection of certain null mutations of OsC1 and OsRb was the main causal mechanism underlying the loss of anthocyanin pigmentation in most cultivated rice. OsP1 and the MYB-bHLH-WD40 complexes regulate anthocyanin biosynthetic genes in rice leaves with partial functional overlap. OsP1 specifically activates upstream biosynthetic genes (OsCHS, OsCHI and OsF3'H) for anthocyanin biosynthesis, whereas the ternary MYB-bHLH-WD40 complex activates all anthocyanin biosynthetic genes including OsCHS, OsCHI, OsF3'H, OsF3H, OsDFR and OsANS. OsC1 and OsRb are tissue-specific regulators that do not influence anthocyanin biosynthesis in the pericarp. Our results reveal the determinant factors, regulatory system and domestication of anthocyanin biosynthesis in rice leaves, and show the potential of engineering anthocyanin biosynthesis in rice.


Assuntos
Antocianinas/biossíntese , Vias Biossintéticas , Domesticação , Oryza/metabolismo , Folhas de Planta/metabolismo , Sequência de Bases , Variação Genética , Estudo de Associação Genômica Ampla , Haplótipos/genética , Oryza/genética , Fenótipo , Proteínas de Plantas/metabolismo , Regiões Promotoras Genéticas/genética , Transcrição Gênica
10.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 101-112, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27816442

RESUMO

Metformin is widely used to treat hyperglycemia. However, metformin treatment may induce intrahepatic cholestasis and liver injury in a few patients with type II diabetes through an unknown mechanism. Here we show that metformin decreases SIRT1 protein levels in primary hepatocytes and liver. Both metformin-treated wild-type C57 mice and hepatic SIRT1-mutant mice had increased hepatic and serum bile acid levels. However, metformin failed to change systemic bile acid levels in hepatic SIRT1-mutant mice. Molecular mechanism study indicates that SIRT1 directly interacts with and deacetylates Foxa2 to inhibit its transcriptional activity on expression of genes involved in bile acids synthesis and transport. Hepatic SIRT1 mutation elevates Foxa2 acetylation levels, which promotes Foxa2 binding to and activating genes involved in bile acids metabolism, impairing hepatic and systemic bile acid homeostasis. Our data clearly suggest that hepatic SIRT1 mediates metformin effects on systemic bile acid metabolism and modulation of SIRT1 activity in liver may be an attractive approach for treatment of bile acid-related diseases such as cholestasis.


Assuntos
Ácidos e Sais Biliares/metabolismo , Colestase Intra-Hepática/genética , Fator 3-beta Nuclear de Hepatócito/genética , Hipoglicemiantes/efeitos adversos , Metformina/efeitos adversos , Sirtuína 1/genética , Acetilação , Animais , Colestase Intra-Hepática/induzido quimicamente , Colestase Intra-Hepática/metabolismo , Colestase Intra-Hepática/patologia , Regulação da Expressão Gênica , Células Hep G2 , Fator 3-beta Nuclear de Hepatócito/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Homeostase/efeitos dos fármacos , Homeostase/genética , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Cultura Primária de Células , Transdução de Sinais , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/metabolismo
11.
J Biol Chem ; 292(52): 21631-21642, 2017 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-29123026

RESUMO

Krüppel-like factor 14 (KLF14) is a member of the Cys2/His2 zinc-finger DNA-binding proteins. Despite strong evidence showing that a polymorphism in the Klf14 gene is closely linked to the development of type 2 diabetes, the physiological and metabolic functions of KLF14 still remain unclear. In the present study, we investigated the role of KLF14 in the regulation of hepatic gluconeogenesis. Adenoviruses expressing KLF14 (Ad-Klf14) or KLF14-specific shRNAs (Ad-shKlf14) were injected into normal C57BL/6J, db/db diabetic, or high-fat diet-induced obese (DIO) mice. Gene expression, hepatic glucose production, glucose tolerance, and insulin resistance were tested in C57BL/6J, db/db, and DIO mice and primary hepatocytes. Our results demonstrate that KLF14 expression is induced in the livers of normal C57BL/6J mice upon fasting and significantly up-regulated in the livers of db/db mice, suggesting a physiological link between KLF14 and gluconeogenesis. Adenovirus-mediated overexpression of KLF14 in primary hepatocytes increased both the mRNA and protein levels of peroxisome proliferator-activated receptor-γ coactivator 1α (Pgc-1α, also known as Ppargc1a), thereby stimulating cellular glucose production. Conversely, knockdown of KLF14 expression led to a reduction in PGC-1α, subsequently inhibiting glucose output in primary hepatocytes. Finally, forced expression of KLF14 in the livers of normal mice increased the plasma glucose levels and impaired glucose tolerance; in contrast, KLF14 knockdown in diabetic mouse livers improved glucose tolerance. Taken together, our data strongly indicate that KLF14 modulates hepatic gluconeogenesis.


Assuntos
Gluconeogênese/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica , Gluconeogênese/genética , Glucose/metabolismo , Intolerância à Glucose/metabolismo , Hepatócitos/metabolismo , Resistência à Insulina/fisiologia , Fatores de Transcrição Kruppel-Like/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo
12.
Diabetologia ; 60(12): 2443-2452, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28836014

RESUMO

AIM/HYPOTHESIS: Abnormal activation of hepatic gluconeogenesis leads to hyperglycaemia. However, the molecular mechanisms underlying dysregulated hepatic gluconeogenesis remain to be fully defined. Here, we explored the physiological role of Krüppel-like factor 10 (KLF10) in regulating hepatic glucose metabolism in mice. METHODS: Hepatic KLF10 expression in wild-type C57BL/6J mice, the db/db mouse model of diabetes, the ob/ob mouse model of obesity and high-fat-diet-induced obese (DIO) mice was measured. Adenoviruses expressing Klf10 or Klf10-specific short-hairpin RNA were injected into wild-type C57BL/6J mice, db/db or DIO mice. Expression of gluconeogenic genes in the liver and blood glucose levels were measured. GTTs and pyruvate tolerance tests were performed. The molecular mechanism by which KLF10 regulates hepatic glucose metabolism was explored. RESULTS: Hepatic KLF10 expression was regulated by nutritional status in wild-type mice and upregulated in diabetic, obese and DIO mice. Overexpression of KLF10 in primary hepatocytes increased the expression of gluconeogenic genes and cellular glucose output. C57BL/6J mice with KLF10 overexpression in the liver displayed increased blood glucose levels and impaired glucose tolerance. Conversely, hepatic KLF10 knockdown in db/db and DIO mice decreased blood glucose levels and improved glucose tolerance. Furthermore, luciferase reporter gene assay and chromatin immunoprecipitation analysis indicated that KLF10 activates Pgc-1α (also known as Ppargc1a) gene transcription via directly binding to its promoter region. CONCLUSIONS/INTERPRETATION: KLF10 is an important regulator of hepatic glucose metabolism and modulation of KLF10 expression in the liver may be an attractive approach for the treatment of type 2 diabetes.


Assuntos
Glicemia/metabolismo , Fatores de Transcrição de Resposta de Crescimento Precoce/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Fígado/metabolismo , Adenoviridae/genética , Animais , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Fatores de Transcrição de Resposta de Crescimento Precoce/genética , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo
13.
Am J Physiol Endocrinol Metab ; 313(4): E493-E505, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28765271

RESUMO

Because of the mass and functions in metabolism, skeletal muscle is one of the major organs regulating whole body metabolic homeostasis. SIRT6, a histone deacetylase, has been shown to regulate metabolism in liver and brain; however, its specific role in skeletal muscle is undetermined. In the present study we explored physiological function of SIRT6 in muscle. We generated a muscle-specific SIRT6 knockout mouse model. The mice with SIRT6 deficiency in muscle displayed impaired glucose homeostasis and insulin sensitivity, attenuated whole body energy expenditure, and weakened exercise performance. Mechanistically, deletion of SIRT6 in muscle decreased expression of genes involved in glucose and lipid uptake, fatty acid oxidation, and mitochondrial oxidative phosphorylation in muscle cells because of the reduced AMP-activated protein kinase (AMPK) activity. In contrast, overexpression of SIRT6 in C2C12 myotubes activates AMPK. Our results from both gain- and loss-of-function experiments identify SIRT6 as a physiological regulator of muscle mitochondrial function. These findings indicate that SIRT6 is a potential therapeutic target for treatment of type 2 diabetes mellitus.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Metabolismo Energético/genética , Glucose/metabolismo , Resistência à Insulina/genética , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Sirtuínas/genética , Animais , Linhagem Celular , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/genética , Homeostase , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Knockout , Fibras Musculares Esqueléticas , Oxirredução , Fosforilação Oxidativa , Condicionamento Físico Animal , Sirtuínas/metabolismo
14.
Diabetologia ; 59(10): 2229-39, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27421728

RESUMO

AIM/HYPOTHESIS: Hepatic forkhead box q1 (FOXQ1) expression levels are regulated by nutritional and pathophysiological status. In this study we investigated the role of FOXQ1 in the regulation of hepatic gluconeogenesis. METHODS: We used multiple mouse and cell models to study the role of FOXQ1 in regulating expression of gluconeogenic genes, and cellular and hepatic glucose production. RESULTS: Expression of hepatic FOXQ1 was regulated by fasting in normal mice and was dysregulated in diabetic mice. Overexpression of FOXQ1 in primary hepatocytes inhibited expression of gluconeogenic genes and decreased cellular glucose output. Hepatic FOXQ1 rescue in db/db and high-fat diet-induced obese mice markedly decreased blood glucose level and improved glucose intolerance. In contrast, wild-type C57 mice with hepatic FOXQ1 deficiency displayed increased blood glucose levels and impaired glucose tolerance. Interestingly, studies into molecular mechanisms indicated that FOXQ1 interacts with FOXO1, thereby blocking FOXO1 activity on hepatic gluconeogenesis, preventing it from directly binding to insulin response elements mapped in the promoter region of gluconeogenic genes. CONCLUSIONS/INTERPRETATION: FOXQ1 is a novel factor involved in regulating hepatic gluconeogenesis, and the decreased FOXQ1 expression in liver may contribute to the development of type 2 diabetes.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Dieta Hiperlipídica/efeitos adversos , Jejum/sangue , Fatores de Transcrição Forkhead/genética , Gluconeogênese/genética , Gluconeogênese/fisiologia , Intolerância à Glucose , Hepatócitos/metabolismo , Insulina/metabolismo , Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
15.
J Biol Chem ; 290(51): 30607-15, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26504089

RESUMO

Dysregulation of hepatic gluconeogenesis contributes to the pathogenesis of diabetes, yet the detailed molecular mechanisms remain to be fully elucidated. Here we show that FOXP1, a transcriptional repressor, plays a key role in the regulation of systemic glucose homeostasis. Hepatic expression levels of FOXP1 are decreased in diabetic mice. Modest hepatic overexpression of FOXP1 in mice inhibited the expression of gluconeogenic genes, such as peroxisome proliferators-activated receptor γ coactivator-1α (PGC-1α), phosphoenolpyruvate carboxykinase (PEPCK), and glucose-6-phosphatase (G6PC), leading to a decrease in hepatic glucose production and fasting blood glucose levels in normal mice and different mouse models of diabetes, including db/db diabetic and high-fat diet-induced obese mice. FOXP1 physically interacted with FOXO1 in vivo and competed with FOXO1 for binding to the insulin response element in the promoter region of gluconeogenic genes, thereby interfering expression of these genes. These results identify a previously unrecognized role for FOXP1 in the transcriptional control of hepatic glucose homeostasis.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Gluconeogênese , Glucose/metabolismo , Homeostase , Fígado/metabolismo , Proteínas Repressoras/metabolismo , Animais , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Fatores de Transcrição Forkhead/genética , Glucose/genética , Masculino , Camundongos , Camundongos Obesos , Obesidade/induzido quimicamente , Obesidade/genética , Obesidade/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fosfoenolpiruvato Carboxiquinase (GTP) , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Biochem Biophys Res Commun ; 471(4): 444-9, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26903296

RESUMO

Previous study showed mammalian Ste20-like kinase (Mst1) may serve as target for the development of new therapies for diabetes. However, the function of Mst1 involved in liver lipid metabolism has remained elusive. In this study, we report that the liver of Mst1 knockout (Mst1(-/-)) mice showed more severe liver metabolic damage under fasting and high-fat diet than that of control mice. And fasting induced hepatic Mst1 expression. Mst1 overexpression inhibited Srebp-1c expression and increased the expression of antioxidant genes in primary hepatocytes. We also found that fasting-induced expression of hepatic Sirt1 was attenuated in Mst1(-/-) mice. Mst1 overexpression promoted Sirt1 expression, probably due to inhibiting Sirt1 ubiquitination. In summary, our study suggests that Mst1 regulates hepatic lipid metabolism by inhibiting Sirt1 ubiquitination in mice.


Assuntos
Metabolismo dos Lipídeos/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Sirtuína 1/metabolismo , Animais , Antioxidantes/metabolismo , Dieta Hiperlipídica/efeitos adversos , Jejum , Regulação da Expressão Gênica , Hepatócitos/fisiologia , Fígado/metabolismo , Fígado/patologia , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Sirtuína 1/genética , Ubiquitinação
17.
J Biol Chem ; 289(34): 23332-42, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-24993831

RESUMO

Hepatic steatosis, characterized by ectopic hepatic triglyceride accumulation, is considered as the early manifestation of non-alcoholic fatty liver diseases (NAFLD). Increased SREBP-1c level and activity contribute to excessive hepatic triglyceride accumulation in NAFLD patients; however, negative regulators of Srebp-1c are not well defined. In this study, we show that Dec1, a critical regulator of circadian rhythm, negatively regulates hepatic Srebp-1c expression. Hepatic Dec1 expression levels are markedly decreased in NAFLD mouse models. Restored Dec1 gene expression levels in NAFLD mouse livers decreased the expression of Srebp-1c and lipogenic genes, subsequently ameliorating the fatty liver phenotype. Conversely, knockdown of Dec1 expression by an adenovirus expressing Dec1-specific shRNA led to an increase in hepatic TG content in normal mouse livers. Correspondingly, expression levels of lipogenic genes, including Srebp-1c, Fas, and Acc, were increased in livers of mice with Dec1 knockdown. Moreover, a functional lipogenesis assay suggested that Dec1 overexpression repressed lipid synthesis in primary hepatocytes. Finally, a luciferase reporter gene assay indicates that DEC1 inhibits Srebp-1c gene transcription via the E-box mapped to the promoter region. Chromatin immunoprecipitation confirmed that DEC1 proteins bound to the identified E-box element. Our studies indicate that DEC1 is an important regulator of Srebp-1c expression and links circadian rhythm to hepatic lipogenesis. Activation of Dec1 can alleviate the nonalcoholic fatty liver phenotype.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Proteínas de Homeodomínio/fisiologia , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Proteína de Ligação a Elemento Regulador de Esterol 1/antagonistas & inibidores , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Imunoprecipitação da Cromatina , Primers do DNA , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/metabolismo
18.
J Hepatol ; 63(3): 713-21, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26026874

RESUMO

BACKGROUND & AIMS: Heme oxygenase 1 (HO-1)-mediated increases in adiponectin, ameliorate the deleterious effects of obesity and metabolic syndrome; however, the effect of HO-1 on hepatic lipid metabolism remains elusive. The aim of this study is to evaluate the role of HO-1 in hepatic lipid metabolism. METHODS: Functional studies were performed using C57BL/6J (WT) mice and Sirt1 liver specific mutant (Sirt1-deficient) mice. The molecular mechanism was explored in primary hepatocytes and mouse liver. RESULTS: Chronic exposure to high-fat diet (HFD) induced hepatic steatosis in WT mice. Treatment of WT mice on HFD with cobalt protoporphyrin (CoPP), an inducer of HO-1 activity, decreased body weight and visceral fat content, reduced intracellular hepatic triglyceride and serum total cholesterol concentrations, and decreased liver lipid droplet formation. Compared with WT mice, the administration of CoPP to Sirt1-deficient mice on HFD increased visceral fat content, and slightly promoted liver lipid droplet formation. CoPP improved glucose tolerance and insulin sensitivity in WT mice on HFD, but compromised insulin sensitivity in Sirt1-deficient mice on HFD. Furthermore, CoPP-induced Sirt1 expression and decreased sterol regulatory element binding protein 1c (SREBP-1c) expression in WT mice on HFD. However, CoPP promoted SREBP-1c expression in Sirt1-deficient hepatocytes, which was reversed by a protein tyrosine phosphatase 1b inhibitor. Additionally, while the administration of CoPP to WT mice on HFD improved antioxidant and anti-inflammatory states, these CoPP-mediated effects were abolished in Sirt1-deficient mice. CONCLUSIONS: Sirt1 mediates the effect of CoPP on ameliorating liver metabolic damage caused by HFD.


Assuntos
Fígado Gorduroso/prevenção & controle , Heme Oxigenase-1/fisiologia , Fígado/efeitos dos fármacos , Protoporfirinas/farmacologia , Sirtuína 1/fisiologia , Animais , Células Cultivadas , Dieta Hiperlipídica , Resistência à Insulina , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Proteína Tirosina Fosfatase não Receptora Tipo 1/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 1/fisiologia
19.
Chemosphere ; 359: 142386, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38777196

RESUMO

The resuspension of phosphorus (P) in sediments has the most significant contribution to the overlying water. The PP release characterization during resuspension was investigated. The results indicated that the P in suspensions had more release risk compared to the sediments. The particulate P (PP) concentration (0.54 mg L-1) under high-intensity rotational speed (250 rad min-1) was about five times higher than others (0.11 mg L-1). The sorption parameters of zero equilibrium P concentration (EPC0F) and soluble reactive P (SRP) were significantly correlated with each other (p < 0.01, r = 0.73). Suspended solids expressed stronger P source than sediments. The values of EPC0F was highly significantly correlated with the sorption coefficient (KF) and native adsorbed P (NAP) (p < 0.01). The mean values of NAP were 0.0612 mg g-1 and 0.0604 mg g-1 in the Prophase and Metaphase, respectively, and 0.0586 mg g-1 at Anaphase. The values of P sorption index (PSI) ranged from 0.4359 to 0.6862 L g-1, with mean values of 0.5350 L g-1 (Prophase), 0.6061 L g-1 (Metaphase), and 0.4967 L g-1 (Anaphase). The degree of P saturation (DPS) decreased in the order of Anaphase (2.73%) > Prophase (2.53%) > Metaphase (2.12%). The release risk index of P (ERI) decreased in the order of Anaphase (5.47%) > Prophase (4.72%) > Metaphase (3.59%), with a range of 2.12%-8.56%. To fast and slow scale, the results of NaOH-P (V1<0, V2>0) contribution indicated that the persistent disturbance promoted the release of adsorbed dissolved PP from NaOH-P in suspended sediment to the overlying water. The contribution of HCl-P (V2 > 0) was positive in the Anaphase of the slow scale, and HCl-P was a PP source in the frequently disturbing conditions.


Assuntos
Monitoramento Ambiental , Sedimentos Geológicos , Fósforo , Poluentes Químicos da Água , Fósforo/análise , Fósforo/química , Sedimentos Geológicos/química , Poluentes Químicos da Água/análise , Adsorção
20.
Chemosphere ; 352: 141276, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38280652

RESUMO

Microbes may induce endogenous phosphorus (P) migration from lacustrine sediment. This study focused on the role of phosphate-solubilizing bacteria (PSB) disturbance in affecting the sediment P release and further contributing to cyanobacterial recruitment in Meiliang Bay, Lake Taihu. Gluconic acid was the main mechanism of phosphate solubilizing by PSB. The dominant PSB (Burkholderia) isolated from eutrophic lake sediments was used as a representative to investigate the effects of disturbance on endogenous P release using diffusive gradients in thin films (DGT) and high-resolution dialysis (HR-Peeper). The results show that soluble reactive phosphorus (SRP) and iron (Fe (II)) concentrations could reach 0.51 mg L-1 and 33.56 mg L-1 in pore water, respectively. And the sediment DGT-P and DGT-Fe were relatively reduced by PSB. Subsequent the chlorophyll a (Chl a) concentrations reached peaks of 344.8 µg L-1 in overlying water. The abundance of the dominant PSB (Burkholderia-Caballeronia-Paraburkholderia) were significantly associated with Chl a (P < 0.05) and algal effective state phosphorus (AAP) (P < 0.05), respectively. PSB mainly regulates AAP leaching to pore water and then diffusing across the sediment-water interface to the overlying water, producing the effect of cyanobacteria recruitment. The results provide new insights into early management of cyanobacterial resuscitation in a large eutrophic lake.


Assuntos
Cianobactérias , Poluentes Químicos da Água , Fosfatos , Lagos , Clorofila A , Sedimentos Geológicos , Poluentes Químicos da Água/análise , Monitoramento Ambiental/métodos , Diálise Renal , Fósforo/análise , Água , China
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA