Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nat Immunol ; 21(10): 1219-1231, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32778760

RESUMO

Chronic inflammation is a common feature of obesity, with elevated cytokines such as interleukin-1 (IL-1) in the circulation and tissues. Here, we report an unconventional IL-1R-MyD88-IRAK2-PHB/OPA1 signaling axis that reprograms mitochondrial metabolism in adipocytes to exacerbate obesity. IL-1 induced recruitment of IRAK2 Myddosome to mitochondria outer membranes via recognition by TOM20, followed by TIMM50-guided translocation of IRAK2 into mitochondria inner membranes, to suppress oxidative phosphorylation and fatty acid oxidation, thereby attenuating energy expenditure. Adipocyte-specific MyD88 or IRAK2 deficiency reduced high-fat-diet-induced weight gain, increased energy expenditure and ameliorated insulin resistance, associated with a smaller adipocyte size and increased cristae formation. IRAK2 kinase inactivation also reduced high-fat diet-induced metabolic diseases. Mechanistically, IRAK2 suppressed respiratory super-complex formation via interaction with PHB1 and OPA1 upon stimulation of IL-1. Taken together, our results suggest that the IRAK2 Myddosome functions as a critical link between inflammation and metabolism, representing a novel therapeutic target for patients with obesity.


Assuntos
Adipócitos/imunologia , Inflamação/imunologia , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-1/metabolismo , Membranas Mitocondriais/metabolismo , Obesidade/imunologia , Adipócitos/patologia , Animais , Células Cultivadas , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Masculino , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fosforilação Oxidativa , Proibitinas , Transporte Proteico , Receptores de Interleucina-1/metabolismo , Transdução de Sinais
2.
Circulation ; 147(14): 1079-1096, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-37011073

RESUMO

BACKGROUND: Large-scale human and mechanistic mouse studies indicate a strong relationship between the microbiome-dependent metabolite trimethylamine N-oxide (TMAO) and several cardiometabolic diseases. This study aims to investigate the role of TMAO in the pathogenesis of abdominal aortic aneurysm (AAA) and target its parent microbes as a potential pharmacological intervention. METHODS: TMAO and choline metabolites were examined in plasma samples, with associated clinical data, from 2 independent patient cohorts (N=2129 total). Mice were fed a high-choline diet and underwent 2 murine AAA models, angiotensin II infusion in low-density lipoprotein receptor-deficient (Ldlr-/-) mice or topical porcine pancreatic elastase in C57BL/6J mice. Gut microbial production of TMAO was inhibited through broad-spectrum antibiotics, targeted inhibition of the gut microbial choline TMA lyase (CutC/D) with fluoromethylcholine, or the use of mice genetically deficient in flavin monooxygenase 3 (Fmo3-/-). Finally, RNA sequencing of in vitro human vascular smooth muscle cells and in vivo mouse aortas was used to investigate how TMAO affects AAA. RESULTS: Elevated TMAO was associated with increased AAA incidence and growth in both patient cohorts studied. Dietary choline supplementation augmented plasma TMAO and aortic diameter in both mouse models of AAA, which was suppressed with poorly absorbed oral broad-spectrum antibiotics. Treatment with fluoromethylcholine ablated TMAO production, attenuated choline-augmented aneurysm initiation, and halted progression of an established aneurysm model. In addition, Fmo3-/- mice had reduced plasma TMAO and aortic diameters and were protected from AAA rupture compared with wild-type mice. RNA sequencing and functional analyses revealed choline supplementation in mice or TMAO treatment of human vascular smooth muscle cells-augmented gene pathways associated with the endoplasmic reticulum stress response, specifically the endoplasmic reticulum stress kinase PERK. CONCLUSIONS: These results define a role for gut microbiota-generated TMAO in AAA formation through upregulation of endoplasmic reticulum stress-related pathways in the aortic wall. In addition, inhibition of microbiome-derived TMAO may serve as a novel therapeutic approach for AAA treatment where none currently exist.


Assuntos
Aneurisma da Aorta Abdominal , Microbioma Gastrointestinal , Humanos , Camundongos , Animais , Suínos , Camundongos Endogâmicos C57BL , Colina , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/prevenção & controle
3.
FASEB J ; 33(1): 1098-1109, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30102568

RESUMO

Alterations in gut microbiota are known to affect intestinal inflammation and obesity. Antibiotic treatment can affect weight gain by elimination of histone deacetylase (HDAC) inhibitor-producing microbes, which are anti-inflammatory by augmenting regulatory T (Treg) cells. We asked whether mice that lack HDAC6 and have potent suppressive Treg cells are protected from microbiota-induced accelerated weight gain. We crossed wild-type and HDAC6-deficient mice and subjected the offspring to perinatal penicillin, inducing weight gain via microbiota disturbance. We observed that male HDAC6-deficient mice were not protected and developed profoundly accelerated weight gain. The antibiotic-exposed HDAC6-deficient mice showed a mixed immune phenotype with increased CD4+ and CD8+ T-cell activation yet maintained the enhanced Treg cell-suppressive function phenotype characteristic of HDAC6-deficient mice. 16S rRNA sequencing of mouse fecal samples reveals that their microbiota diverged with time, with HDAC6 deletion altering microbiome composition. On a high-fat diet, HDAC6-deficient mice were depleted in representatives of the S24-7 family and Lactobacillus but enriched with Bacteroides and Parabacteroides; these changes are associated with obesity. Our findings further our understanding of the influence of HDACs on microbiome composition and are important for the development of HDAC6 inhibitors in the treatment of human diseases.-Lieber, A. D., Beier, U. H., Xiao, H., Wilkins, B. J., Jiao, J., Li, X. S., Schugar, R. C., Strauch, C. M., Wang, Z., Brown, J. M., Hazen, S. L., Bokulich, N. A., Ruggles, K. V., Akimova, T., Hancock, W. W., Blaser, M. J. Loss of HDAC6 alters gut microbiota and worsens obesity.


Assuntos
Microbioma Gastrointestinal , Desacetilase 6 de Histona/fisiologia , Obesidade/genética , Obesidade/microbiologia , Animais , Bacteroides/isolamento & purificação , Dieta Hiperlipídica , Fígado Gorduroso/genética , Fezes , Vida Livre de Germes , Desacetilase 6 de Histona/genética , Hiperlipidemias/genética , Lactobacillus/isolamento & purificação , Masculino , Mesentério/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/imunologia , Baço/patologia , Linfócitos T Reguladores/imunologia , Regulação para Cima , Aumento de Peso
4.
Arterioscler Thromb Vasc Biol ; 39(6): 1045-1054, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31070450

RESUMO

Objective- FMO (flavin-containing monooxygenase) 3 converts bacterial-derived trimethylamine to trimethylamine N-oxide (TMAO), an independent risk factor for cardiovascular disease. We generated FMO3 knockout (FMO3KO) mouse to study its effects on plasma TMAO, lipids, glucose/insulin metabolism, thrombosis, and atherosclerosis. Approach and Results- Previous studies with an antisense oligonucleotide (ASO) knockdown strategy targeting FMO3 in LDLRKO (low-density lipoprotein receptor knockout) mice resulted in major reductions in TMAO levels and atherosclerosis, but also showed effects on plasma lipids, insulin, and glucose. Although FMO3KO mice generated via CRISPR/Cas9 technology bred onto the LDLRKO background did exhibit similar effects on TMAO levels, the effects on lipid metabolism were not as pronounced as with the ASO knockdown model. These differences could result from either off-target effects of the ASO or from a developmental adaptation to the FMO3 deficiency. To distinguish these possibilities, we treated wild-type and FMO3KO mice with control or FMO3 ASOs. FMO3-ASO treatment led to the same extent of lipid-lowering effects in the FMO3KO mice as the wild-type mice, indicating off-target effects. The levels of TMAO in LDLRKO mice fed an atherogenic diet are very low in both wild-type and FMO3KO mice, and no significant effect was observed on atherosclerosis. When FMO3KO and wild-type mice were maintained on a 0.5% choline diet, FMO3KO showed a marked reduction in both TMAO and in vivo thrombosis potential. Conclusions- FMO3KO markedly reduces systemic TMAO levels and thrombosis potential. However, the previously observed large effects of an FMO3 ASO on plasma lipid levels appear to be due partly to off-target effects.


Assuntos
Aterosclerose/metabolismo , Colina/metabolismo , Metilaminas/metabolismo , Oxigenases/genética , Trombose/metabolismo , Animais , Aterosclerose/genética , Colina/farmacologia , Modelos Animais de Doenças , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigenases/metabolismo , Reação em Cadeia da Polimerase/métodos , Distribuição Aleatória , Valores de Referência , Trombose/fisiopatologia
5.
Arterioscler Thromb Vasc Biol ; 38(1): 218-231, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29074585

RESUMO

OBJECTIVE: Human genetic variants near the FADS (fatty acid desaturase) gene cluster (FADS1-2-3) are strongly associated with cardiometabolic traits including dyslipidemia, fatty liver, type 2 diabetes mellitus, and coronary artery disease. However, mechanisms underlying these genetic associations are unclear. APPROACH AND RESULTS: Here, we specifically investigated the physiological role of the Δ-5 desaturase FADS1 in regulating diet-induced cardiometabolic phenotypes by treating hyperlipidemic LDLR (low-density lipoprotein receptor)-null mice with antisense oligonucleotides targeting the selective knockdown of Fads1. Fads1 knockdown resulted in striking reorganization of both ω-6 and ω-3 polyunsaturated fatty acid levels and their associated proinflammatory and proresolving lipid mediators in a highly diet-specific manner. Loss of Fads1 activity promoted hepatic inflammation and atherosclerosis, yet was associated with suppression of hepatic lipogenesis. Fads1 knockdown in isolated macrophages promoted classic M1 activation, whereas suppressing alternative M2 activation programs, and also altered systemic and tissue inflammatory responses in vivo. Finally, the ability of Fads1 to reciprocally regulate lipogenesis and inflammation may rely in part on its role as an effector of liver X receptor signaling. CONCLUSIONS: These results position Fads1 as an underappreciated regulator of inflammation initiation and resolution, and suggest that endogenously synthesized arachidonic acid and eicosapentaenoic acid are key determinates of inflammatory disease progression and liver X receptor signaling.


Assuntos
Aorta/enzimologia , Doenças da Aorta/enzimologia , Aterosclerose/enzimologia , Dislipidemias/enzimologia , Ácidos Graxos Dessaturases/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/enzimologia , Lipogênese , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Ácido Araquidônico/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Dessaturase de Ácido Graxo Delta-5 , Modelos Animais de Doenças , Dislipidemias/genética , Dislipidemias/patologia , Ácido Eicosapentaenoico/metabolismo , Ácidos Graxos Dessaturases/genética , Inflamação/genética , Inflamação/patologia , Fígado/metabolismo , Receptores X do Fígado/metabolismo , Ativação de Macrófagos , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Placa Aterosclerótica , Receptores de LDL/deficiência , Receptores de LDL/genética
6.
Curr Opin Lipidol ; 26(5): 426-31, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26218418

RESUMO

PURPOSE OF REVIEW: Atherosclerosis and associated cardiovascular disease still remain the largest cause of mortality worldwide. Several recent studies have discovered that metabolism of common nutrients by gut microbes can produce a proatherogenic metabolite called trimethylamine-N-oxide (TMAO). The goal of this review is to discuss emerging evidence that the hepatic enzyme that generates TMAO, flavin monooxygenase 3 (FMO3), plays a regulatory role in maintaining whole body cholesterol balance and atherosclerosis development. RECENT FINDINGS: Several independent studies have recently uncovered a link between either FMO3 itself or its enzymatic product TMAO with atherosclerosis and hepatic insulin resistance. These recent studies show that inhibition of FMO3 stimulates macrophage reverse cholesterol transport and protects against atherosclerosis in mice. SUMMARY: A growing body of work demonstrates that nutrients present in high-fat foods (phosphatidylcholine, choline and L-carnitine) can be metabolized by the gut microbial enzymes to generate trimethylamine, which is then further metabolized by the host enzyme FMO3 to produce proatherogenic TMAO. Here, we discuss emerging evidence that the TMAO-producing enzyme FMO3 is centrally involved in the pathogenesis of atherosclerosis by regulating cholesterol metabolism and insulin resistance, and how these new insights provide exciting new avenues for cardiovascular disease therapies.


Assuntos
Aterosclerose/enzimologia , Colesterol/metabolismo , Oxigenases/fisiologia , Animais , Humanos , Resistência à Insulina , Metabolismo dos Lipídeos
7.
Am J Physiol Endocrinol Metab ; 304(4): E363-74, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23233542

RESUMO

During states of low carbohydrate intake, mammalian ketone body metabolism transfers energy substrates originally derived from fatty acyl chains within the liver to extrahepatic organs. We previously demonstrated that the mitochondrial enzyme coenzyme A (CoA) transferase [succinyl-CoA:3-oxoacid CoA transferase (SCOT), encoded by nuclear Oxct1] is required for oxidation of ketone bodies and that germline SCOT-knockout (KO) mice die within 48 h of birth because of hyperketonemic hypoglycemia. Here, we use novel transgenic and tissue-specific SCOT-KO mice to demonstrate that ketone bodies do not serve an obligate energetic role within highly ketolytic tissues during the ketogenic neonatal period or during starvation in the adult. Although transgene-mediated restoration of myocardial CoA transferase in germline SCOT-KO mice is insufficient to prevent lethal hyperketonemic hypoglycemia in the neonatal period, mice lacking CoA transferase selectively within neurons, cardiomyocytes, or skeletal myocytes are all viable as neonates. Like germline SCOT-KO neonatal mice, neonatal mice with neuronal CoA transferase deficiency exhibit increased cerebral glycolysis and glucose oxidation, and, while these neonatal mice exhibit modest hyperketonemia, they do not develop hypoglycemia. As adults, tissue-specific SCOT-KO mice tolerate starvation, exhibiting only modestly increased hyperketonemia. Finally, metabolic analysis of adult germline Oxct1(+/-) mice demonstrates that global diminution of ketone body oxidation yields hyperketonemia, but hypoglycemia emerges only during a protracted state of low carbohydrate intake. Together, these data suggest that, at the tissue level, ketone bodies are not a required energy substrate in the newborn period or during starvation, but rather that integrated ketone body metabolism mediates adaptation to ketogenic nutrient states.


Assuntos
Envelhecimento , Coenzima A-Transferases/metabolismo , Corpos Cetônicos/metabolismo , Cetose/fisiopatologia , Músculo Esquelético/enzimologia , Miócitos Cardíacos/enzimologia , Neurônios/enzimologia , Adaptação Fisiológica , Animais , Animais Recém-Nascidos , Restrição Calórica/efeitos adversos , Coenzima A-Transferases/biossíntese , Coenzima A-Transferases/genética , Heterozigoto , Hipoglicemia/etiologia , Corpos Cetônicos/sangue , Cetose/sangue , Cetose/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Especificidade de Órgãos , Oxirredução
8.
Am J Physiol Heart Circ Physiol ; 304(8): H1060-76, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23396451

RESUMO

Ketone bodies are metabolized through evolutionarily conserved pathways that support bioenergetic homeostasis, particularly in brain, heart, and skeletal muscle when carbohydrates are in short supply. The metabolism of ketone bodies interfaces with the tricarboxylic acid cycle, ß-oxidation of fatty acids, de novo lipogenesis, sterol biosynthesis, glucose metabolism, the mitochondrial electron transport chain, hormonal signaling, intracellular signal transduction pathways, and the microbiome. Here we review the mechanisms through which ketone bodies are metabolized and how their signals are transmitted. We focus on the roles this metabolic pathway may play in cardiovascular disease states, the bioenergetic benefits of myocardial ketone body oxidation, and prospective interactions among ketone body metabolism, obesity, metabolic syndrome, and atherosclerosis. Ketone body metabolism is noninvasively quantifiable in humans and is responsive to nutritional interventions. Therefore, further investigation of this pathway in disease models and in humans may ultimately yield tailored diagnostic strategies and therapies for specific pathological states.


Assuntos
Doenças Cardiovasculares/metabolismo , Corpos Cetônicos/metabolismo , Animais , Cardiomiopatias/metabolismo , Coenzima A-Transferases/metabolismo , Ácidos Graxos/metabolismo , Humanos , Hidroximetilglutaril-CoA Sintase/metabolismo , Lipogênese/fisiologia , Fígado/metabolismo , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Hepáticas/metabolismo , Miocárdio/metabolismo , Obesidade/metabolismo , Oxirredução , Transdução de Sinais
9.
Curr Opin Clin Nutr Metab Care ; 15(4): 374-80, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22617564

RESUMO

PURPOSE OF REVIEW: Obesity-associated nonalcoholic fatty liver disease (NAFLD) is highly prevalent, for which weight loss is the generally recommended clinical management. Low-carbohydrate ketogenic diets have been successful in promoting weight loss, but variations in the range of metabolic responses to these diets indicate that the effects of altering macronutrient content are not completely understood. This review focuses on the most recent findings that reveal the relationship between low-carbohydrate diets and NAFLD in rodent models and humans. RECENT FINDINGS: Low-carbohydrate diets have been shown to promote weight loss, decrease intrahepatic triglyceride content, and improve metabolic parameters of patients with obesity. These ketogenic diets also provoke weight loss in rodents. However, long-term maintenance on a ketogenic diet stimulates the development of NAFLD and systemic glucose intolerance in mice. The relationship between ketogenic diets and systemic insulin resistance in both humans and rodents remains to be elucidated. SUMMARY: Because low-carbohydrate ketogenic diets are increasingly employed for treatment of obesity, NAFLD, and neurological diseases such as epilepsy, understanding the long-term systemic effects of low-carbohydrate diets is crucial to the development of efficacious and safe dietary interventions.


Assuntos
Dieta com Restrição de Carboidratos , Dieta Cetogênica , Dieta Redutora , Fígado Gorduroso/etiologia , Intolerância à Glucose/etiologia , Obesidade/dietoterapia , Redução de Peso/fisiologia , Animais , Glicemia/metabolismo , Carboidratos da Dieta/metabolismo , Fígado Gorduroso/metabolismo , Intolerância à Glucose/metabolismo , Humanos , Resistência à Insulina/fisiologia , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica , Obesidade/complicações , Obesidade/metabolismo , Triglicerídeos/metabolismo
10.
J Cell Biol ; 177(1): 73-86, 2007 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-17420291

RESUMO

We have shown that muscle-derived stem cells (MDSCs) transplanted into dystrophic (mdx) mice efficiently regenerate skeletal muscle. However, MDSC populations exhibit heterogeneity in marker profiles and variability in regeneration abilities. We show here that cell sex is a variable that considerably influences MDSCs' regeneration abilities. We found that the female MDSCs (F-MDSCs) regenerated skeletal muscle more efficiently. Despite using additional isolation techniques and cell cloning, we could not obtain a male subfraction with a regeneration capacity similar to that of their female counterparts. Rather than being directly hormonal or caused by host immune response, this difference in MDSCs' regeneration potential may arise from innate sex-related differences in the cells' stress responses. In comparison with F-MDSCs, male MDSCs have increased differentiation after exposure to oxidative stress induced by hydrogen peroxide, which may lead to in vivo donor cell depletion, and a proliferative advantage for F-MDSCs that eventually increases muscle regeneration. These findings should persuade researchers to report cell sex, which is a largely unexplored variable, and consider the implications of relying on cells of one sex.


Assuntos
Músculo Esquelético/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Regeneração/genética , Fatores Sexuais , Transplante de Células-Tronco , Células-Tronco/classificação
11.
Elife ; 112022 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-35072627

RESUMO

Obesity has repeatedly been linked to reorganization of the gut microbiome, yet to this point obesity therapeutics have been targeted exclusively toward the human host. Here, we show that gut microbe-targeted inhibition of the trimethylamine N-oxide (TMAO) pathway protects mice against the metabolic disturbances associated with diet-induced obesity (DIO) or leptin deficiency (Lepob/ob). Small molecule inhibition of the gut microbial enzyme choline TMA-lyase (CutC) does not reduce food intake but is instead associated with alterations in the gut microbiome, improvement in glucose tolerance, and enhanced energy expenditure. We also show that gut microbial CutC inhibition is associated with reorganization of host circadian control of both phosphatidylcholine and energy metabolism. This study underscores the relationship between microbe and host metabolism and provides evidence that gut microbe-derived trimethylamine (TMA) is a key regulator of the host circadian clock. This work also demonstrates that gut microbe-targeted enzyme inhibitors have potential as anti-obesity therapeutics.


Assuntos
Colina/análogos & derivados , Ritmo Circadiano/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Obesidade/metabolismo , Animais , Colina/administração & dosagem , Colina/metabolismo , Dieta Hiperlipídica , Inibidores Enzimáticos/farmacologia , Leptina/deficiência , Liases/efeitos dos fármacos , Masculino , Metilaminas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/microbiologia
12.
Elife ; 112022 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-35084335

RESUMO

There is mounting evidence that microbes residing in the human intestine contribute to diverse alcohol-associated liver diseases (ALD) including the most deadly form known as alcohol-associated hepatitis (AH). However, mechanisms by which gut microbes synergize with excessive alcohol intake to promote liver injury are poorly understood. Furthermore, whether drugs that selectively target gut microbial metabolism can improve ALD has never been tested. We used liquid chromatography tandem mass spectrometry to quantify the levels of microbe and host choline co-metabolites in healthy controls and AH patients, finding elevated levels of the microbial metabolite trimethylamine (TMA) in AH. In subsequent studies, we treated mice with non-lethal bacterial choline TMA lyase (CutC/D) inhibitors to blunt gut microbe-dependent production of TMA in the context of chronic ethanol administration. Indices of liver injury were quantified by complementary RNA sequencing, biochemical, and histological approaches. In addition, we examined the impact of ethanol consumption and TMA lyase inhibition on gut microbiome structure via 16S rRNA sequencing. We show the gut microbial choline metabolite TMA is elevated in AH patients and correlates with reduced hepatic expression of the TMA oxygenase flavin-containing monooxygenase 3 (FMO3). Provocatively, we find that small molecule inhibition of gut microbial CutC/D activity protects mice from ethanol-induced liver injury. CutC/D inhibitor-driven improvement in ethanol-induced liver injury is associated with distinct reorganization of the gut microbiome and host liver transcriptome. The microbial metabolite TMA is elevated in patients with AH, and inhibition of TMA production from gut microbes can protect mice from ethanol-induced liver injury.


Assuntos
Bactérias/metabolismo , Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Microbioma Gastrointestinal , Hepatite/metabolismo , Metilaminas/metabolismo , Animais , Etanol/efeitos adversos , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória
13.
Am J Physiol Gastrointest Liver Physiol ; 300(6): G956-67, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21454445

RESUMO

Low-carbohydrate diets are used to manage obesity, seizure disorders, and malignancies of the central nervous system. These diets create a distinctive, but incompletely defined, cellular, molecular, and integrated metabolic state. Here, we determine the systemic and hepatic effects of long-term administration of a very low-carbohydrate, low-protein, and high-fat ketogenic diet, serially comparing these effects to a high-simple-carbohydrate, high-fat Western diet and a low-fat, polysaccharide-rich control chow diet in C57BL/6J mice. Longitudinal measurement of body composition, serum metabolites, and intrahepatic fat content, using in vivo magnetic resonance spectroscopy, reveals that mice fed the ketogenic diet over 12 wk remain lean, euglycemic, and hypoinsulinemic but accumulate hepatic lipid in a temporal pattern very distinct from animals fed the Western diet. Ketogenic diet-fed mice ultimately develop systemic glucose intolerance, hepatic endoplasmic reticulum stress, steatosis, cellular injury, and macrophage accumulation, but surprisingly insulin-induced hepatic Akt phosphorylation and whole-body insulin responsiveness are not impaired. Moreover, whereas hepatic Pparg mRNA abundance is augmented by both high-fat diets, each diet confers splice variant specificity. The distinctive nutrient milieu created by long-term administration of this low-carbohydrate, low-protein ketogenic diet in mice evokes unique signatures of nonalcoholic fatty liver disease and whole-body glucose homeostasis.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Dieta com Restrição de Carboidratos/efeitos adversos , Dieta Cetogênica/efeitos adversos , Retículo Endoplasmático/metabolismo , Fígado Gorduroso/etiologia , Inflamação/etiologia , Fígado/metabolismo , Estresse Fisiológico , Análise de Variância , Animais , Biomarcadores/sangue , Glicemia/metabolismo , Composição Corporal , Dieta com Restrição de Proteínas , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/metabolismo , Retículo Endoplasmático/patologia , Ingestão de Energia , Ácidos Graxos não Esterificados/sangue , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/fisiopatologia , Regulação da Expressão Gênica , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/metabolismo , Insulina/sangue , Resistência à Insulina , Fígado/patologia , Fígado/fisiopatologia , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , PPAR gama/genética , PPAR gama/metabolismo , Fatores de Tempo , Triglicerídeos/sangue , Resposta a Proteínas não Dobradas
14.
J Biomed Biotechnol ; 2009: 789526, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20037738

RESUMO

Human umbilical cord blood is an excellent primitive source of noncontroversial stem cells for treatment of hematologic disorders; meanwhile, new stem cell candidates in the umbilical cord (UC) tissue could provide therapeutic cells for nonhematologic disorders. We show novel in situ characterization to identify and localize a panel of some markers expressed by mesenchymal stromal cells (MSCs; CD44, CD105, CD73, CD90) and CD146 in the UC. We describe enzymatic isolation and purification methods of different UC cell populations that do not require manual separation of the vessels and stroma of the coiled, helical-like UC tissue. Unique quantitation of in situ cell frequency and stromal cell counts upon harvest illustrate the potential to obtain high numerical yields with these methods. UC stromal cells can differentiate to the osteogenic and chondrogenic lineages and, under specific culturing conditions, they exhibit high expandability with unique long-term stability of their phenotype. The remarkable stability of the phenotype represents a novel finding for human MSCs, from any source, and supports the use of these cells as highly accessible stromal cells for both basic studies and potentially therapeutic applications such as allogeneic clinical use for musculoskeletal disorders.


Assuntos
Técnicas de Cultura de Células/métodos , Sangue Fetal/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Engenharia Tecidual/métodos , Antígeno CD146/metabolismo , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Humanos , Fenótipo
15.
Elife ; 82019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31621579

RESUMO

Recent studies have identified a genetic variant rs641738 near two genes encoding membrane bound O-acyltransferase domain-containing 7 (MBOAT7) and transmembrane channel-like 4 (TMC4) that associate with increased risk of non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcohol-related cirrhosis, and liver fibrosis in those infected with viral hepatitis (Buch et al., 2015; Mancina et al., 2016; Luukkonen et al., 2016; Thabet et al., 2016; Viitasalo et al., 2016; Krawczyk et al., 2017; Thabet et al., 2017). Based on hepatic expression quantitative trait loci analysis, it has been suggested that MBOAT7 loss of function promotes liver disease progression (Buch et al., 2015; Mancina et al., 2016; Luukkonen et al., 2016; Thabet et al., 2016; Viitasalo et al., 2016; Krawczyk et al., 2017; Thabet et al., 2017), but this has never been formally tested. Here we show that Mboat7 loss, but not Tmc4, in mice is sufficient to promote the progression of NAFLD in the setting of high fat diet. Mboat7 loss of function is associated with accumulation of its substrate lysophosphatidylinositol (LPI) lipids, and direct administration of LPI promotes hepatic inflammatory and fibrotic transcriptional changes in an Mboat7-dependent manner. These studies reveal a novel role for MBOAT7-driven acylation of LPI lipids in suppressing the progression of NAFLD.


Assuntos
Aciltransferases/genética , Proteínas de Membrana/genética , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade/genética , Acilação , Animais , Progressão da Doença , Humanos , Camundongos
16.
Crit Rev Eukaryot Gene Expr ; 18(2): 173-88, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18304031

RESUMO

Sex is well known to influence life expectancy and disposition to disease. Stem and progenitor cells are believed to persist throughout life, and they contribute to the repair and healthy maintenance of tissue; consequently, sex-related differences demonstrated by stem cells may provide insight to sex-related differences in aging, disease, and healing. However, cell sex is an often overlooked variable in stem cell biology.


Assuntos
Células-Tronco Adultas/fisiologia , Músculo Esquelético/fisiologia , Caracteres Sexuais , Envelhecimento/fisiologia , Animais , Técnicas de Cultura de Células , Doença/etiologia , Estrogênios/fisiologia , Feminino , Expressão Gênica/fisiologia , Humanos , Masculino , Modelos Biológicos , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/anatomia & histologia , Músculo Esquelético/metabolismo , Tamanho do Órgão , Estresse Oxidativo/fisiologia , Pericitos/fisiologia , Células Satélites de Músculo Esquelético/fisiologia
17.
Cell Biochem Biophys ; 52(2): 85-102, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18841496

RESUMO

The field of regenerative medicine offers hope for the development of a cell-based therapy for the repair of articular cartilage (AC). Yet, the greatest challenge in the use of stem cells for tissue repair, is understanding how the cells respond to stimuli and using that knowledge to direct cell fate. Novel methods that utilize stem cells in cartilage regeneration will require specific spatio-temporal controls of the biochemical and biophysical signaling environments. Current chondrogenic differentiation research focuses on the roles of biochemical stimuli like growth factors, hormones, and small molecules, and the role of the physical environment and mechanical stimuli, such as compression and shear stress, which likely act through mechanical receptors. Numerous signals are associated with chondrogenic-like activity of cells in different systems, however many variables for a controlled method still need to be optimized; e.g., spatial and temporal application of the stimuli, and time of transplantation of an engineered construct. Understanding the necessary microenvironmental signals for cell differentiation will advance cell therapy for cartilage repair.


Assuntos
Cartilagem Articular/citologia , Diferenciação Celular/fisiologia , Condrogênese/fisiologia , Células-Tronco/citologia , Engenharia Tecidual , Animais , Cartilagem Articular/fisiologia , Ensaio de Unidades Formadoras de Colônias/métodos , Hormônios/fisiologia , Humanos , Mecanorreceptores/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia
18.
Adipocyte ; 7(1): 49-56, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29172946

RESUMO

The human body is an integrated circuit between microbial symbionts and our Homo sapien genome, which communicate bi-directionally to maintain homeostasis within the human meta-organism. There is now strong evidence that microbes resident in the human intestine can directly contribute to the pathogenesis of obesity and associated cardiometabolic disorders. In fact, gut microbes represent a filter of our greatest environmental exposure - the foods we consume. It is now clear that we each experience a given meal differently, based on our unique gut microbial communities. Biologically active gut microbe-derived metabolites, such as short chain fatty acids, secondary bile acids, and trimethylamine-N-oxide (TMAO), are now uniquely recognized as contributors to obesity and related cardiometabolic disorders. However, mechanistic insights into how microbe-derived metabolites promote obesity are largely unknown. Recent work has demonstrated that the meta-organismal production of the bacterial co-metabolite TMAO is linked to suppression of beiging of white adipose tissue in mice and humans. Furthermore, the TMAO pathway is becoming an increasingly attractive therapeutic target in obesity-associated diseases such as type 2 diabetes, kidney failure, and cardiovascular disease. In this commentary we discuss recent findings linking the TMAO pathway to obesity-associated disorders, and provide additional insights into potential mechanisms driving this microbe-host interaction.


Assuntos
Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Colina/metabolismo , Microbioma Gastrointestinal/fisiologia , Animais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/microbiologia , Humanos , Período Pós-Prandial
19.
JCI Insight ; 2(22)2017 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-29202451

RESUMO

Three Akt isoforms, encoded by 3 separate genes, are expressed in mammals. While the roles of Akt1 and Akt2 in metabolism are well established, it is not yet known whether Akt3 plays a role in metabolic diseases. We now report that Akt3 protects mice from high-fat diet-induced obesity by suppressing an alternative pathway of adipogenesis via with no lysine protein kinase-1 (WNK1) and serum/glucocorticoid-inducible kinase 1 (SGK1). We demonstrate that Akt3 specifically phosphorylates WNK1 at T58 and promotes its degradation via the ubiquitin-proteasome pathway. A lack of Akt3 in adipocytes increases the WNK1 protein level, leading to activation of SGK1. SGK1, in turn, promotes adipogenesis by phosphorylating and inhibiting transcription factor FOXO1 and, subsequently, activating the transcription of PPARγ in adipocytes. Akt3-deficient mice have an increased number of adipocytes and, when fed a high-fat diet, display increased weight gain, white adipose tissue expansion, and impaired glucose homeostasis. Pharmacological blockade of SGK1 in high-fat diet-fed Akt3-deficient mice suppressed adipogenesis, prevented excessive weight gain and adiposity, and ameliorated metabolic parameters. Thus, Akt3/WNK1/SGK1 represents a potentially novel signaling pathway controlling the development of obesity.


Assuntos
Adipogenia/efeitos dos fármacos , Proteínas Imediatamente Precoces/metabolismo , Obesidade/prevenção & controle , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Proteína Quinase 1 Deficiente de Lisina WNK/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Adipócitos/patologia , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/patologia , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Proteína Forkhead Box O1/metabolismo , Glucose/metabolismo , Homeostase , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/sangue , Masculino , Camundongos , Camundongos Knockout , PPAR gama/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/sangue , Proteínas Proto-Oncogênicas c-akt/genética , Fatores de Transcrição/efeitos dos fármacos , Ubiquitina/metabolismo , Proteína Quinase 1 Deficiente de Lisina WNK/sangue , Aumento de Peso
20.
Cell Rep ; 19(12): 2451-2461, 2017 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28636934

RESUMO

Emerging evidence suggests that microbes resident in the human intestine represent a key environmental factor contributing to obesity-associated disorders. Here, we demonstrate that the gut microbiota-initiated trimethylamine N-oxide (TMAO)-generating pathway is linked to obesity and energy metabolism. In multiple clinical cohorts, systemic levels of TMAO were observed to strongly associate with type 2 diabetes. In addition, circulating TMAO levels were associated with obesity traits in the different inbred strains represented in the Hybrid Mouse Diversity Panel. Further, antisense oligonucleotide-mediated knockdown or genetic deletion of the TMAO-producing enzyme flavin-containing monooxygenase 3 (FMO3) conferred protection against obesity in mice. Complimentary mouse and human studies indicate a negative regulatory role for FMO3 in the beiging of white adipose tissue. Collectively, our studies reveal a link between the TMAO-producing enzyme FMO3 and obesity and the beiging of white adipose tissue.


Assuntos
Metilaminas/sangue , Obesidade/enzimologia , Oxigenases/fisiologia , Gordura Subcutânea/enzimologia , Adipócitos Bege/enzimologia , Animais , Diabetes Mellitus Tipo 2/sangue , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/sangue , Obesidade/patologia , Gordura Subcutânea/patologia , Gordura Subcutânea/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA