Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Anim Cogn ; 26(5): 1505-1519, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37302101

RESUMO

Social comparison is a fundamental human characteristic; however, long-term social comparison may induce psychological stress and can lead to depression and anxiety. Recent studies have shown that nonhuman primates compare themselves with others; however, no studies have investigated whether social comparisons exist among rodents. In the present study, we established a rat model of social comparison. This model was subsequently used to examine the effects of the differential environment of a partner on depression- and anxiety-like behaviors in male rats, as well as to assess the changes in serum, medial prefrontal cortex (mPFC), and dorsal hippocampus brain-derived neurotrophic factor (BDNF) levels induced by long-term social comparison. Compared to rats whose partners were exposed to the same environment, rats whose partners were exposed to two combined enriched environmental stimuli for 14 days showed significantly decreased social novelty preference and sucrose consumption. No anxiety-like behaviors were observed. Rats whose partners were exposed to one enriched environment for 31 days showed significantly increased immobility time in the forced swimming test, and significantly decreased time spent in the center area in the open-field test. Further, rats whose partners were exposed to one enriched environment for 31 days showed lower BDNF levels in the mPFC and dorsal hippocampus, but not following partner exposure for 14 days. These results suggest that social comparisons exist in rats and can induce psychosocial stress and other negative affect. This model will not only provide the possibility to reveal the neurobiological basis of the emotional impact of social comparison, but could also be used to confirm the conservative evolutionary characteristics of social comparison as a behavioral attribute.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Depressão , Animais , Humanos , Masculino , Ratos , Ansiedade/metabolismo , Ansiedade/psicologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Depressão/metabolismo , Depressão/psicologia , Hipocampo/metabolismo , Comparação Social , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia
2.
Hepatol Commun ; 7(7)2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37314747

RESUMO

BACKGROUND AND AIMS: Alcohol-associated liver disease (ALD) and NAFLD often coexist in Western societies that consume energy-rich and cholesterol-containing Western diets. Increased rates of ALD mortality in young people in these societies are likely attributable to binge drinking. It is largely unknown how alcohol binge causes liver damage in the setting of Western diets. APPROACH AND RESULTS: In this study, we showed that a single ethanol binge (5 g/kg body weight) induced severe liver injury as shown by marked increases in serum activities of the 2 aminotransferases AST and ALT in C57BL/6J mice that have been fed a Western diet for 3 weeks. The Western diet plus binge ethanol-fed mice also displayed severe lipid droplet deposition and high contents of triglycerides and cholesterol in the liver, which were associated with increased lipogenic and reduced fatty acid oxidative gene expression. These animals had the highest Cxcl1 mRNA expression and myeloperoxidase (MPO)-positive neutrophils in the liver. Their hepatic ROS and lipid peroxidation were the highest, but their hepatic levels of mitochondrial oxidative phosphorylation proteins remained largely unaltered. Hepatic levels of several ER stress markers, including mRNAs for CHOP, ERO1A, ERO1B, BIM, and BIP, as well as Xbp1 splicing and proteins for BIP/GRP78 and IRE-α were also the highest in these animals. Interestingly, Western diet feeding for 3 weeks or ethanol binge dramatically increased hepatic caspase 3 cleavage, and the combination of the 2 did not further increase it. Thus, we successfully established a murine model of acute liver injury by mimicking human diets and binge drinking. CONCLUSIONS: This simple Western diet plus single ethanol binge model recapitulates major hepatic phenotypes of ALD, including steatosis and steatohepatitis characterized by neutrophil infiltration, oxidative stress, and ER stress.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas , Hepatopatias Alcoólicas , Hepatopatia Gordurosa não Alcoólica , Humanos , Animais , Camundongos , Adolescente , Camundongos Endogâmicos C57BL , Etanol/toxicidade , Dieta Ocidental/efeitos adversos , Consumo Excessivo de Bebidas Alcoólicas/complicações , Hepatopatia Gordurosa não Alcoólica/etiologia
3.
Hepatology ; 77(5): 1688-1701, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35844150

RESUMO

BACKGROUND AND AIMS: Alcohol-associated liver disease (ALD) pathologies include steatosis, inflammation, and injury, which may progress to fibrosis, cirrhosis, and cancer. The liver receives ~60% of fatty acids from adipose tissue triglyceride hydrolysis, but the role of this lipolytic pathway in ALD development has not been directly examined in any genetic animal models with selective inactivation of adipose lipolysis. APPROACH AND RESULTS: Using adipose-specific comparative gene identification-58 (CGI-58) knockout (FAT-KO) mice, a model of impaired adipose lipolysis, we show that mice deficient in adipose lipolysis are almost completely protected against ethanol-induced hepatic steatosis and lipid peroxidation when subjected to the National Institute on Alcohol Abuse and Alcoholism chronic and binge ethanol feeding model. This is unlikely due to reduced lipid synthesis because this regimen of ethanol feeding down-regulated hepatic expression of lipogenic genes similarly in both genotypes. In the pair-fed group, FAT-KO relative to control mice displayed increased hepatocyte injury, neutrophil infiltration, and activation of the transcription factor signal transducer and activator of transcription 3 (STAT3) in the liver; and none of these were exacerbated by ethanol feeding. Activation of STAT3 is associated with a marked increase in hepatic leptin receptor mRNA expression and adipose inflammatory cell infiltration. CONCLUSIONS: Our findings establish a critical role of adipose lipolysis in driving hepatic steatosis and oxidative stress during ALD development.


Assuntos
Fígado Gorduroso , Hepatopatias Alcoólicas , Estados Unidos , Camundongos , Animais , Etanol/farmacologia , Lipólise , Modelos Animais de Doenças , National Institute on Alcohol Abuse and Alcoholism (U.S.) , Fígado Gorduroso/metabolismo , Fígado/patologia , Hepatopatias Alcoólicas/metabolismo , Camundongos Endogâmicos C57BL
4.
Front Microbiol ; 13: 1062282, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36532451

RESUMO

Drug-resistant fungal infections are emerging as an important clinical problem. In general, antifungal resistance results from increased target expression or mutations within the target protein sequence. However, the molecular mechanisms of non-drug target mutations of antifungal resistance in fungal pathogens remain to be explored. Previous studies indicated that the metal chaperone protein Mtm1 is required for mitochondrial Sod2 activation and responses to oxidative stress in yeast and in the fungal pathogen Aspergillus fumigatus, but there is no report of MtmA-related antifungal resistance. In this study, we found that repressed expression of MtmA (only 10% expression) using a conditional promoter resulted in significantly enhanced itraconazole resistance, which was not the result of highly expressed drug targets Erg11A and Erg11B. Furthermore, we demonstrated that repressed expression of MtmA results in upregulation of a series of multidrug resistance-associated transport genes, which may cause multidrug resistance. Further mechanistic studies revealed that inhibition of MtmA expression led to abnormal activation of the calcium signaling system and prompted persistent nucleation of the calcium signaling transcription factor CrzA. Our findings suggest that the metal chaperone protein MtmA is able to negatively regulate fungal resistance via affecting calcium signaling pathway.

5.
Appl Environ Microbiol ; 88(9): e0018222, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35435716

RESUMO

The detoxification system of reactive oxygen species (ROS) plays critical roles in the survival and virulence of fungal pathogens in infected hosts, while superoxide dismutase (SOD) is the primary ROS scavenger. In the model yeast Saccharomyces cerevisiae, the metal chaperone protein Mtm1 is required for mitochondrial Sod2 activation and responses to oxidative stress. However, the function of the S. cerevisiae Mtm1 homolog in the human fungal pathogen Aspergillus fumigatus has not yet been clarified. In this study, we found that mitochondria-localized MtmA in A. fumigatus, a putative homolog of yeast Mtm1, not only has a similar function to Mtm1 in responding to oxidative stress resistance by affecting SodB (MnSOD) activity but is also essential for hyphal growth such that repressed expression of MtmA results in severe growth defects in A. fumigatus. In addition, the chelation of Zn2+ can obviously rescue growth defects caused by repression of MtmA, suggesting that MtmA may be involved in hyphal growth by affecting cellular Zn2+ detoxification. Moreover, MtmA contains four Mito-carr domains, whereas only the first Mito-carr domain is required for the function of MtmA. Therefore, the findings in this study suggest that MtmA in A. fumigatus has an important and unique function that is different from that in yeast. IMPORTANCE Knowledge of the key factors required for the viability of pathogenic fungi can help to explore new antifungal drugs. Here, we demonstrate that MtmA is involved in responding to oxidative stress by activating mitochondrial SodB activity. MtmA, especially for the first Mito-carr domain, is essential for colony growth by regulating cellular Zn2+ equilibrium and responses to oxidative stress in A. fumigatus. This is the first report of the vital and unique role of the MtmA protein in pathogenic fungi, indicating that it might be a potential antifungal drug target.


Assuntos
Aspergillus fumigatus , Saccharomyces cerevisiae , Antifúngicos/metabolismo , Antifúngicos/farmacologia , Aspergillus fumigatus/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
6.
Obesity (Silver Spring) ; 30(1): 153-164, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34825496

RESUMO

OBJECTIVES: Beta-3 adrenergic receptors (ß3-AR) stimulate lipolysis and thermogenesis in white and brown adipose tissue (WAT and BAT). Obesity increases oxidative stress and inflammation that attenuate AT ß3-AR signaling. The objective of this study was to test the hypothesis that the combination of the ß3-AR agonist CL-316,243 (CL) and the antioxidant alpha-lipoic acid (ALA) would lower inflammation in diet-induced obesity (DIO) and improve ß3-AR function. METHODS: A total of 40 DIO mice were separated into four groups: Control (per os and intraperitoneal [IP] vehicle); CL alone (0.01 mg/kg IP daily); ALA alone (250 mg/kg in drinking water); or ALA+CL combination, all for 5 weeks. RESULTS: Food intake was similar in all groups; however, mice receiving ALA+CL showed improved body composition and inflammation as well as lower body weight (+1.7 g Control vs. -2.5 g ALA+CL [-7%]; p < 0.01) and percentage of body fat (-9%, p < 0.001). Systemic and epididymal WAT inflammation was lower with ALA+CL than all other groups, with enhanced recruitment of epididymal WAT anti-inflammatory CD206+ M2 macrophages. ß3-AR signaling in WAT was enhanced in the combination-treatment group, with higher mRNA and protein levels of thermogenic uncoupling protein 1 and AT lipases. CONCLUSIONS: Chronic treatment with ALA and a ß3-AR agonist reduces DIO-induced inflammation. AT immune modulation could be a therapeutic target in patients with obesity.


Assuntos
Ácido Tióctico , Tecido Adiposo Marrom/metabolismo , Agonistas Adrenérgicos/metabolismo , Agonistas Adrenérgicos/farmacologia , Animais , Dieta Hiperlipídica/efeitos adversos , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Obesos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Ácido Tióctico/metabolismo , Ácido Tióctico/farmacologia , Ácido Tióctico/uso terapêutico
7.
Int J Mol Sci ; 22(24)2021 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-34948160

RESUMO

The heart primarily uses fatty acids as energy substrates. Adipose lipolysis is a major source of fatty acids, particularly under stress conditions. In this study, we showed that mice with selective inactivation of the lipolytic coactivator comparative gene identification-58 (CGI-58) in adipose tissue (FAT-KO mice), relative to their littermate controls, had lower circulating FA levels in the fed and fasted states due to impaired adipose lipolysis. They preferentially utilized carbohydrates as energy fuels and were more insulin sensitive and glucose tolerant. Under cold stress, FAT-KO versus control mice had >10-fold increases in glucose uptake in the hearts but no increases in other tissues examined. Plasma concentrations of atrial natriuretic peptide and cardiac mRNAs for atrial and brain-type natriuretic peptides, two sensitive markers of cardiac remodeling, were also elevated. After one week of cold exposure, FAT-KO mice showed reduced cardiac expression of several mitochondrial oxidative phosphorylation proteins. After one month of cold exposure, hearts of these animals showed depressed functions, reduced SERCA2 protein, and increased proteins for MHC-ß, collagen I proteins, Glut1, Glut4 and phospho-AMPK. Thus, CGI-58-dependent adipose lipolysis critically regulates cardiac metabolism and function, especially during cold adaptation. The adipose-heart axis may be targeted for the management of cardiac dysfunction.


Assuntos
Aclimatação , Resposta ao Choque Frio , Glucose/metabolismo , Lipólise , Mitocôndrias Cardíacas/metabolismo , Miocárdio/metabolismo , Animais , Caderinas/deficiência , Caderinas/metabolismo , Glucose/genética , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/genética
8.
Cells ; 10(12)2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34943976

RESUMO

Niemann-Pick C1-like 1 (NPC1L1) mediates intestinal uptake of dietary and biliary cholesterol and is the target of ezetimibe, a cholesterol absorption inhibitor used to treat hypercholesterolemia. Genetic deletion of NPC1L1 or ezetimibe treatment protects mice from high-fat diet (HFD)-induced obesity; however, the molecular mechanisms responsible for this therapeutic benefit remain unknown. A major metabolic fate of cholesterol is its conversion to bile acids. We found that NPC1L1 knockout (L1-KO) mice fed an HFD had increased energy expenditure, bile acid pool size, and fecal bile acid excretion rates. The elevated bile acid pool in the HFD-fed L1-KO mice was enriched with tauro-ß-muricholic acid. These changes in the L1-KO mice were associated with reduced ileal mRNA expression of fibroblast growth factor 15 (FGF15) and increased hepatic mRNA expression of cholesterol 7α-hydroxylase (Cyp7A1) and mitochondrial sterol 27-hydroxylase (Cyp27A1). In addition, mRNA expression of the membrane bile acid receptor Takeda G protein-coupled receptor 5 (TGR5) and type 2 iodothyronine deiodinase (Dio2) were elevated in brown adipose tissue of L1-KO mice, which is known to promote energy expenditure. Thus, altered bile acid homeostasis and signaling may play a role in protecting L1-KO mice against HFD-induced obesity.


Assuntos
Ácidos e Sais Biliares/genética , Fatores de Crescimento de Fibroblastos/genética , Íleo/metabolismo , Proteínas de Membrana Transportadoras/genética , Obesidade/genética , Animais , Ácidos e Sais Biliares/metabolismo , Colestanotriol 26-Mono-Oxigenase/genética , Colesterol/genética , Colesterol/metabolismo , Colesterol 7-alfa-Hidroxilase/genética , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético/genética , Humanos , Íleo/efeitos dos fármacos , Íleo/patologia , Iodeto Peroxidase/genética , Fígado/metabolismo , Fígado/patologia , Proteínas de Membrana Transportadoras/deficiência , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Obesidade/patologia , Receptores Acoplados a Proteínas G/genética , Iodotironina Desiodinase Tipo II
9.
Sci Rep ; 10(1): 4701, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32170127

RESUMO

Humans and rodents with Comparative Gene Identification-58 (CGI-58) mutations manifest nonalcoholic fatty liver disease (NAFLD). Here we show that liver CGI-58 knockout (LivKO) mice fed a Western diet rapidly develop advanced NAFLD, including nonalcoholic steatohepatitis (NASH) and hepatic fibrosis. After 14 weeks of diet challenge, starting at 6 weeks of age, LivKO mice showed increased inflammatory cell infiltration and proinflammatory gene expression in the liver, which was associated with elevated plasma levels of aminotransferases. Hepatic ductular reactions, pericellular fibrosis, and bridging fibrosis were observed only in the LivKO mice. Consistently, the KO mice had a significant increase in hepatic mRNAs for fibrogenic genes. In addition, LivKO mice displayed massive accumulation of lipid droplets (LDs) in hepatocytes. LDs were also observed in the cholangiocytes of the LivKO mice, but not the floxed controls. Four of the five LD coat proteins, including perilipins 2, 3, 4, and 5, were increased in the CGI-58 KO liver. CRISPR/Cas9-mediated knockout of CGI-58 in Huh7 human hepatoma cells induced LD deposition and perilipin expression, suggesting a cell autonomous effect. Our findings establish the Western diet-fed LivKO mice as an animal model of NASH and hepatic fibrosis. These animals may facilitate preclinical screening of therapeutic agents that counter against NAFLD progression.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/deficiência , Dieta Ocidental/efeitos adversos , Suscetibilidade a Doenças , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , Hepatopatia Gordurosa não Alcoólica/complicações , Hepatopatia Gordurosa não Alcoólica/etiologia , Animais , Peso Corporal , Modelos Animais de Doenças , Progressão da Doença , Predisposição Genética para Doença , Glucose/metabolismo , Hepatomegalia , Humanos , Cirrose Hepática/patologia , Camundongos , Camundongos Knockout , Índice de Gravidade de Doença
12.
Cell Metab ; 26(5): 764-777.e5, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-28988822

RESUMO

Lipid droplet (LD) lipolysis in brown adipose tissue (BAT) is generally considered to be required for cold-induced nonshivering thermogenesis. Here, we show that mice lacking BAT Comparative Gene Identification-58 (CGI-58), a lipolytic activator essential for the stimulated LD lipolysis, have normal thermogenic capacity and are not cold sensitive. Relative to littermate controls, these animals had higher body temperatures when they were provided food during cold exposure. The increase in body temperature in the fed, cold-exposed knockout mice was associated with increased energy expenditure and with increased sympathetic innervation and browning of white adipose tissue (WAT). Mice lacking CGI-58 in both BAT and WAT were cold sensitive, but only in the fasted state. Thus, LD lipolysis in BAT is not essential for cold-induced nonshivering thermogenesis in vivo. Rather, CGI-58-dependent LD lipolysis in BAT regulates WAT thermogenesis, and our data uncover an essential role of WAT lipolysis in fueling thermogenesis during fasting.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/metabolismo , Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Lipólise/fisiologia , Termogênese/fisiologia , 1-Acilglicerol-3-Fosfato O-Aciltransferase/genética , Animais , Temperatura Corporal , Temperatura Baixa , Metabolismo Energético , Jejum , Humanos , Hipotermia Induzida , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
13.
Cell Metab ; 26(4): 672-685.e4, 2017 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-28918936

RESUMO

While activation of beige thermogenesis is a promising approach for treatment of obesity-associated diseases, there are currently no known pharmacological means of inducing beiging in humans. Intermittent fasting is an effective and natural strategy for weight control, but the mechanism for its efficacy is poorly understood. Here, we show that an every-other-day fasting (EODF) regimen selectively stimulates beige fat development within white adipose tissue and dramatically ameliorates obesity, insulin resistance, and hepatic steatosis. EODF treatment results in a shift in the gut microbiota composition leading to elevation of the fermentation products acetate and lactate and to the selective upregulation of monocarboxylate transporter 1 expression in beige cells. Microbiota-depleted mice are resistance to EODF-induced beiging, while transplantation of the microbiota from EODF-treated mice to microbiota-depleted mice activates beiging and improves metabolic homeostasis. These findings provide a new gut-microbiota-driven mechanism for activating adipose tissue browning and treating metabolic diseases.


Assuntos
Tecido Adiposo Bege/metabolismo , Tecido Adiposo Branco/metabolismo , Jejum , Microbioma Gastrointestinal , Obesidade/terapia , Animais , Metabolismo Energético , Fígado Gorduroso/complicações , Fígado Gorduroso/metabolismo , Fígado Gorduroso/microbiologia , Fígado Gorduroso/terapia , Fatores de Crescimento de Fibroblastos/metabolismo , Resistência à Insulina , Síndrome Metabólica/complicações , Síndrome Metabólica/metabolismo , Síndrome Metabólica/microbiologia , Síndrome Metabólica/terapia , Camundongos Endogâmicos C57BL , Obesidade/complicações , Obesidade/metabolismo , Obesidade/microbiologia , Transdução de Sinais , Termogênese
14.
J Biol Chem ; 292(9): 3929-3939, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28100772

RESUMO

Sphingolipids are a diverse class of essential cellular lipids that function as structural membrane components and as signaling molecules. Cells acquire sphingolipids by both de novo biosynthesis and recycling of exogenous sphingolipids. The individual importance of these pathways for the generation of essential sphingolipids in differentiated cells is not well understood. To investigate the requirement for de novo sphingolipid biosynthesis in adipocytes, a cell type with highly regulated lipid metabolism, we generated mice with an adipocyte-specific deletion of Sptlc1 Sptlc1 is an obligate subunit of serine palmitoyltransferase, the enzyme responsible for the first and rate-limiting step of de novo sphingolipid biosynthesis. These mice, which initially developed adipose tissue, exhibited a striking age-dependent loss of adipose tissue accompanied by evidence of adipocyte death, increased macrophage infiltration, and tissue fibrosis. Adipocyte differentiation was not affected by the Sptlc1 deletion. The mice also had elevated fasting blood glucose, fatty liver, and insulin resistance. Collectively, these data indicate that de novo sphingolipid biosynthesis is required for adipocyte cell viability and normal metabolic function and that reduced de novo sphingolipid biosynthesis within adipocytes is associated with adipocyte death, adipose tissue remodeling, and metabolic dysfunction.


Assuntos
Adipócitos/citologia , Homeostase , Serina C-Palmitoiltransferase/genética , Esfingolipídeos/biossíntese , Adiposidade , Animais , Diferenciação Celular , Sobrevivência Celular , Deleção de Genes , Inflamação , Resistência à Insulina , Metabolismo dos Lipídeos , Lipogênese , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Serina C-Palmitoiltransferase/metabolismo
15.
Sci Rep ; 5: 10512, 2015 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-26015368

RESUMO

We have previously observed that knockout of Niemann-Pick C1-Like 1 (NPC1L1), a cholesterol transporter essential for intestinal cholesterol absorption, reduces the output of dry stool in mice. As the food intake remains unaltered in NPC1L1-knockout (L1-KO) mice, we hypothesized that NPC1L1 deficiency may alter the gut microbiome to reduce stool output. Consistently, here we demonstrate that the phyla of fecal microbiota differ substantially between L1-KO mice and their wild-type controls. Germ-free (GF) mice have reduced stool output. Inhibition of NPC1L1 by its inhibitor ezetimibe reduces stool output in specific pathogen-free (SPF), but not GF mice. In addition, we show that GF versus SPF mice have reduced intestinal absorption and increased fecal excretion of cholesterol, particularly after treatment with ezetimibe. This negative balance of cholesterol in GF mice is associated with reduced plasma and hepatic cholesterol, and likely caused by reduced expression of NPC1L1 and increased expression of ABCG5 and ABCG8 in small intestine. Expression levels of other genes in intestine and liver largely reflect a state of cholesterol depletion and a decrease in intestinal sensing of bile acids. Altogether, our findings reveal a broad role of microbiota in regulating whole-body cholesterol homeostasis and its response to a cholesterol-lowering drug, ezetimibe.


Assuntos
Bactérias/isolamento & purificação , Colesterol/metabolismo , Regulação da Expressão Gênica , Intestino Delgado/microbiologia , Microbiota , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Membro 8 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Anticolesterolemiantes/farmacologia , Bactérias/genética , Ácidos e Sais Biliares/metabolismo , Peso Corporal/efeitos dos fármacos , Colesterol 7-alfa-Hidroxilase/genética , Colesterol 7-alfa-Hidroxilase/metabolismo , Família 7 do Citocromo P450 , Dieta Hiperlipídica , Ezetimiba/farmacologia , Fezes/microbiologia , Absorção Intestinal/efeitos dos fármacos , Intestino Delgado/metabolismo , Metabolismo dos Lipídeos/genética , Lipídeos/sangue , Lipoproteínas/genética , Lipoproteínas/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Ribossômico 16S/química , RNA Ribossômico 16S/genética , Esteroide Hidroxilases/metabolismo , Regulação para Cima
16.
Endocrinology ; 156(5): 1648-58, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25751639

RESUMO

Intramyocellular accumulation of lipids is often associated with insulin resistance. Deficiency of comparative gene identification-58 (CGI-58) causes cytosolic deposition of triglyceride (TG)-rich lipid droplets in most cell types, including muscle due to defective TG hydrolysis. It was unclear, however, whether CGI-58 deficiency-induced lipid accumulation in muscle influences insulin sensitivity. Here we show that muscle-specific CGI-58 knockout mice relative to their controls have increased glucose tolerance and insulin sensitivity on a Western-type high-fat diet, despite TG accumulation in both heart and oxidative skeletal muscle and cholesterol deposition in heart. Although the intracardiomyocellular lipid deposition results in cardiac ventricular fibrosis and systolic dysfunction, muscle-specific CGI-58 knockout mice show increased glucose uptake in heart and soleus muscle, improved insulin signaling in insulin-sensitive tissues, and reduced plasma concentrations of glucose, insulin, and cholesterol. Hepatic contents of TG and cholesterol are also decreased in these animals. Cardiac steatosis is attributable, at least in part, to decreases in cardiac TG hydrolase activity and peroxisome proliferator-activated receptor-α/peroxisome proliferator-activated receptor-γ coactivator-1-dependent mitochondrial fatty acid oxidation. In conclusion, muscle CGI-58 deficiency causes cardiac dysfunction and fat deposition in oxidative muscles but induces a series of favorable metabolic changes in mice fed a high-fat diet.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/genética , Glucose/metabolismo , Resistência à Insulina , Gotículas Lipídicas/metabolismo , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Disfunção Ventricular Esquerda/metabolismo , Animais , Colesterol/metabolismo , Dieta Hiperlipídica , Ecocardiografia , Ácidos Graxos/metabolismo , Deleção de Genes , Lipase/metabolismo , Gotículas Lipídicas/patologia , Fígado/metabolismo , Masculino , Camundongos , Mitocôndrias/metabolismo , Músculo Esquelético/patologia , Miocárdio/patologia , PPAR alfa/metabolismo , PPAR gama/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/metabolismo , Triglicerídeos/metabolismo , Disfunção Ventricular Esquerda/diagnóstico por imagem , Disfunção Ventricular Esquerda/patologia
17.
Cell Rep ; 9(5): 1798-1811, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25482557

RESUMO

How cancer cells shift metabolism to aerobic glycolysis is largely unknown. Here, we show that deficiency of α/ß-hydrolase domain-containing 5 (Abhd5), an intracellular lipolytic activator that is also known as comparative gene identification 58 (CGI-58), promotes this metabolic shift and enhances malignancies of colorectal carcinomas (CRCs). Silencing of Abhd5 in normal fibroblasts induces malignant transformation. Intestine-specific knockout of Abhd5 in Apc(Min/+) mice robustly increases tumorigenesis and malignant transformation of adenomatous polyps. In colon cancer cells, Abhd5 deficiency induces epithelial-mesenchymal transition by suppressing the AMPKα-p53 pathway, which is attributable to increased aerobic glycolysis. In human CRCs, Abhd5 expression falls substantially and correlates negatively with malignant features. Our findings link Abhd5 to CRC pathogenesis and suggest that cancer cells develop aerobic glycolysis by suppressing Abhd5-mediated intracellular lipolysis.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/genética , Neoplasias Colorretais/genética , Glicólise , Aerobiose , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Progressão da Doença , Transição Epitelial-Mesenquimal , Feminino , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Invasividade Neoplásica , Transplante de Neoplasias
18.
Atherosclerosis ; 237(2): 609-17, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25463095

RESUMO

OBJECTIVE: The correlation between intestinal cholesterol absorption values and plasma low-density lipoprotein-cholesterol (LDL-C) levels remains controversial. Niemann-Pick-C1-Like 1 (NPC1L1) is essential for intestinal cholesterol absorption, and is the target of ezetimibe, a cholesterol absorption inhibitor. However, studies with NPC1L1 knockout mice or ezetimibe cannot definitively clarify this correlation because NPC1L1 expression is not restricted to intestine in humans and mice. In this study we sought to genetically address this issue. METHODS AND RESULTS: We developed a mouse model that lacks endogenous (NPC1L1) and LDL receptor (LDLR) (DKO), but transgenically expresses human NPC1L1 in gastrointestinal tract only (DKO/L1(IntOnly) mice). Our novel model eliminated potential effects of non-intestinal NPC1L1 on cholesterol homeostasis. We found that human NPC1L1 was localized at the intestinal brush border membrane of DKO/L1(IntOnly) mice. Cholesterol feeding induced formation of NPC1L1-positive vesicles beneath this membrane in an ezetimibe-sensitive manner. Compared to DKO mice, DKO/L1(IntOnly) mice showed significant increases in cholesterol absorption and blood/hepatic/biliary cholesterol. Increased blood cholesterol was restricted to very low-density lipoprotein (VLDL) and LDL fractions, which was associated with increased secretion and plasma levels of apolipoproteins B100 and B48. Additionally, DKO/L1(IntOnly) mice displayed decreased fecal cholesterol excretion and hepatic/intestinal expression of cholesterologenic genes. Ezetimibe treatment virtually reversed all of the transgene-related phenotypes in DKO/L1(IntOnly) mice. CONCLUSION: Our findings from DKO/L1(IntOnly) mice clearly demonstrate that NPC1L1-mediated cholesterol absorption is a major determinant of blood levels of apolipoprotein B-containing atherogenic lipoproteins, at least in mice.


Assuntos
LDL-Colesterol/sangue , Regulação da Expressão Gênica , Proteínas de Membrana/metabolismo , Receptores de LDL/metabolismo , Animais , Apolipoproteína B-100 , Apolipoproteína B-48/metabolismo , Apolipoproteínas B/metabolismo , Azetidinas/farmacologia , Colesterol/sangue , Colesterol/metabolismo , Ezetimiba , Homeostase , Humanos , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Fenótipo , Receptores de LDL/genética , Transgenes
19.
Cell Rep ; 7(1): 223-35, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24703845

RESUMO

Overnutrition activates a proinflammatory program in macrophages to induce insulin resistance (IR), but its molecular mechanisms remain incompletely understood. Here, we show that saturated fatty acid and lipopolysaccharide, two factors implicated in high-fat diet (HFD)-induced IR, suppress macrophage CGI-58 expression. Macrophage-specific CGI-58 knockout (MaKO) in mice aggravates HFD-induced glucose intolerance and IR, which is associated with augmented systemic/tissue inflammation and proinflammatory activation of adipose tissue macrophages. CGI-58-deficient macrophages exhibit mitochondrial dysfunction due to defective peroxisome proliferator-activated receptor (PPAR)γ signaling. Consequently, they overproduce reactive oxygen species (ROS) to potentiate secretion of proinflammatory cytokines by activating NLRP3 inflammasome. Anti-ROS treatment or NLRP3 silencing prevents CGI-58-deficient macrophages from oversecreting proinflammatory cytokines and from inducing proinflammatory signaling and IR in the cocultured fat slices. Anti-ROS treatment also prevents exacerbation of inflammation and IR in HFD-fed MaKO mice. Our data thus establish CGI-58 as a suppressor of overnutrition-induced NLRP3 inflammasome activation in macrophages.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/deficiência , Inflamassomos/metabolismo , Resistência à Insulina/fisiologia , Macrófagos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , 1-Acilglicerol-3-Fosfato O-Aciltransferase/biossíntese , Animais , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Regulação para Baixo , Ácidos Graxos/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR
20.
PLoS One ; 9(3): e91652, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24618586

RESUMO

Comparative Gene Identification-58 (CGI-58), a lipid droplet (LD)-associated protein, promotes intracellular triglyceride (TG) hydrolysis in vitro. Mutations in human CGI-58 cause TG accumulation in numerous tissues including intestine. Enterocytes are thought not to store TG-rich LDs, but a fatty meal does induce temporary cytosolic accumulation of LDs. Accumulated LDs are eventually cleared out, implying existence of TG hydrolytic machinery in enterocytes. However, identities of proteins responsible for LD-TG hydrolysis remain unknown. Here we report that intestine-specific inactivation of CGI-58 in mice significantly reduces postprandial plasma TG concentrations and intestinal TG hydrolase activity, which is associated with a 4-fold increase in intestinal TG content and large cytosolic LD accumulation in absorptive enterocytes during the fasting state. Intestine-specific CGI-58 knockout mice also display mild yet significant decreases in intestinal fatty acid absorption and oxidation. Surprisingly, inactivation of CGI-58 in intestine significantly raises plasma and intestinal cholesterol, and reduces hepatic cholesterol, without altering intestinal cholesterol absorption and fecal neutral sterol excretion. In conclusion, intestinal CGI-58 is required for efficient postprandial lipoprotein-TG secretion and for maintaining hepatic and plasma lipid homeostasis. Our animal model will serve as a valuable tool to further define how intestinal fat metabolism influences the pathogenesis of metabolic disorders, such as obesity and type 2 diabetes.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/deficiência , Absorção Intestinal/genética , Mucosa Intestinal/metabolismo , Metabolismo dos Lipídeos/genética , Período Pós-Prandial , 1-Acilglicerol-3-Fosfato O-Aciltransferase/genética , Animais , Colesterol/sangue , Colesterol/metabolismo , Enterócitos/metabolismo , Ácidos Graxos/metabolismo , Feminino , Hidrólise , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Intestinos/patologia , Lipase/metabolismo , Masculino , Camundongos , Camundongos Knockout , Oxirredução , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA