Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Hepatol ; 69(2): 385-395, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29572095

RESUMO

BACKGROUND AND AIMS: Although the majority of patients with non-alcoholic fatty liver disease (NAFLD) have only steatosis without progression, a sizeable fraction develop non-alcoholic steatohepatitis (NASH), which can lead to cirrhosis and hepatocellular carcinoma (HCC). Many established diet-induced mouse models for NASH require 24-52 weeks, which makes testing for drug response costly and time consuming. METHODS: We have sought to establish a murine NASH model with rapid progression of extensive fibrosis and HCC by using a western diet (WD), which is high-fat, high-fructose and high-cholesterol, combined with low weekly dose of intraperitoneal carbon tetrachloride (CCl4), which serves as an accelerator. RESULTS: C57BL/6J mice were fed a normal chow diet ±â€¯CCl4 or WD ±â€¯CCl4 for 12 and 24 weeks. Addition of CCl4 exacerbated histological features of NASH, fibrosis, and tumor development induced by WD, which resulted in stage 3 fibrosis at 12 weeks and HCC development at 24 weeks. Furthermore, whole liver transcriptomic analysis indicated that dysregulated molecular pathways in WD/CCl4 mice and immunologic features were similar to those of human NASH. CONCLUSIONS: Our mouse NASH model exhibits rapid progression of advanced fibrosis and HCC, and mimics histological, immunological and transcriptomic features of human NASH, suggesting that it will be a useful experimental tool for preclinical drug testing. LAY SUMMARY: A carefully characterized model has been developed in mice that recapitulates the progressive stages of human fatty liver disease, from simple steatosis, to inflammation, fibrosis and cancer. The functional pathways of gene expression and immune abnormalities in this model closely resemble human disease. The ease and reproducibility of this model make it ideal to study disease pathogenesis and test new treatments.


Assuntos
Dieta Ocidental , Fígado Gorduroso , Cirrose Hepática , Neoplasias Hepáticas , Camundongos Endogâmicos C57BL , Animais , Tetracloreto de Carbono/farmacologia , Modelos Animais de Doenças , Progressão da Doença , Fígado Gorduroso/etiologia , Fígado Gorduroso/imunologia , Fígado Gorduroso/patologia , Perfilação da Expressão Gênica/métodos , Inflamação/patologia , Cirrose Hepática/etiologia , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Neoplasias Hepáticas/etiologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Camundongos , Reprodutibilidade dos Testes
3.
Sci Signal ; 16(787): eadf6696, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37253090

RESUMO

Organ size is maintained by the controlled proliferation of distinct cell populations. In the mouse liver, hepatocytes in the midlobular zone that are positive for cyclin D1 (CCND1) repopulate the parenchyma at a constant rate to preserve liver mass. Here, we investigated how hepatocyte proliferation is supported by hepatic stellate cells (HSCs), pericytes that are in close proximity to hepatocytes. We used T cells to ablate nearly all HSCs in the murine liver, enabling the unbiased characterization of HSC functions. In the normal liver, complete loss of HSCs persisted for up to 10 weeks and caused a gradual reduction in liver mass and in the number of CCND1+ hepatocytes. We identified neurotrophin-3 (Ntf-3) as an HSC-produced factor that induced the proliferation of midlobular hepatocytes through the activation of tropomyosin receptor kinase B (TrkB). Treating HSC-depleted mice with Ntf-3 restored CCND1+ hepatocytes in the midlobular region and increased liver mass. These findings establish that HSCs form the mitogenic niche for midlobular hepatocytes and identify Ntf-3 as a hepatocyte growth factor.


Assuntos
Células Estreladas do Fígado , Fígado , Neurotrofina 3 , Animais , Camundongos , Proliferação de Células , Células Estreladas do Fígado/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , Neurotrofina 3/metabolismo
4.
Lipids Health Dis ; 11: 75, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22698140

RESUMO

BACKGROUND: Resynthesis of triglycerides in enterocytes of the small intestine plays a critical role in the absorption of dietary fat. Acyl-CoA:monoacylglycerol acyltransferase-2 (MGAT2) is highly expressed in the small intestine and catalyzes the synthesis of diacylglycerol from monoacylglycerol and acyl-CoA. To determine the physiological importance of MGAT2 in metabolic disorders and lipid metabolism in the small intestine, we constructed and analyzed Mgat2-deficient mice. RESULTS: In oral fat tolerance test (OFTT), Mgat2-deficient mice absorbed less fat into the circulation. When maintained on a high-fat diet (HFD), Mgat2-deficient mice were protected from HFD-induced obesity and insulin resistance. Heterozygote (Mgat2+/-) mice had an intermediate phenotype between Mgat2+/+ and Mgat2-/- and were partially protected from metabolic disorders. Despite of a decrease in fat absorption in the Mgat2-deficient mice, lipid levels in the feces and small intestine were comparable among the genotypes. Oxygen consumption was increased in the Mgat2-deficient mice when maintained on an HFD. A prominent upregulation of the genes involved in fatty acid oxidation was observed in the duodenum but not in the liver of the Mgat2-deficient mice. CONCLUSION: These results suggest that MGAT2 has a pivotal role in lipid metabolism in the small intestine, and the inhibition of MGAT2 activity may be a promising strategy for the treatment of obesity-related metabolic disorders.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Gorduras/metabolismo , Resistência à Insulina/fisiologia , Absorção Intestinal/fisiologia , Obesidade/metabolismo , Animais , Composição Corporal/genética , Composição Corporal/fisiologia , Teste de Tolerância a Glucose , Resistência à Insulina/genética , Absorção Intestinal/genética , Camundongos , Camundongos Knockout , N-Acetilglucosaminiltransferases , Obesidade/genética , Reação em Cadeia da Polimerase
5.
Nihon Yakurigaku Zasshi ; 154(4): 203-209, 2019.
Artigo em Japonês | MEDLINE | ID: mdl-31597900

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is a rising cause of chronic liver disease worldwide. Although majority of patients with NAFLD are benign and non-progressive, having only steatosis, some fraction of patients develop non-alcoholic steatohepatitis (NASH), which can lead to cirrhosis, hepatocellular carcinoma, and eventually increased liver-related mortality. Among histological features of NAFLD, it has been reported that liver fibrosis is the most important predictor of long-term outcomes. Liver fibrosis is a dynamic process characterized by the over-accumulation of extracellular matrix due to chronic liver injury resulting from any etiology including not only NASH, but also viral infection and alcoholic liver disease. Activation of hepatic stellate cells (HSCs) has been well established as a central driver of fibrosis in experimental animal models and human liver injury. It is a transdifferentiation of quiescent, vitamin-A­storing cells into proliferative and fibrogenic myofibroblasts. However, the discovery of novel pathways and mediators reveals the complexity of HSC activation. These emerging pathways include hedgehog, autophagy, free cholesterol, YAP1, hepcidin, and nuclear/G-protein coupled receptor-mediated signals. In addition, pathways of HSC clearance have been uncovered such as apoptosis, senescence, and reversion to an inactivated state. Thus, clarifying the underlying mechanisms of HSC activation could lead to the identification of novel therapeutic targets for NASH, and several drug candidates are currently being developed in clinical trials.


Assuntos
Células Estreladas do Fígado/citologia , Cirrose Hepática/terapia , Hepatopatia Gordurosa não Alcoólica/terapia , Animais , Transdiferenciação Celular , Humanos
6.
Biol Open ; 8(5)2019 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-31097447

RESUMO

Iron overload in the liver causes oxidative stress and inflammation, which result in organ dysfunction, making it a risk factor for non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma. We aimed to evaluate the effect of dietary iron restriction on disease progression in rats fed a choline-deficient L-amino acid-defined (CDAA) diet. Male F344 rats were fed a choline-sufficient amino acid-defined (control) diet, a CDAA diet or an iron-restricted CDAA diet for 4, 8 and 12 weeks. At each time point, hepatic iron levels, oxidative stress, inflammation and fibrosis were evaluated by immunohistochemistry. The iron-restricted CDAA diet significantly decreased serum iron levels for 12 weeks compared with the CDAA diet. Histological analysis confirmed that feeding with the CDAA diet induced hepatic iron overload and that this was associated with oxidative stress (number of 8-hydroxydeoxyguanosine-positive cells), inflammation (CD68 positive area) and fibrosis (Sirius Red positive area). Iron restriction with the CDAA diet significantly led to a reduction in the hepatic iron levels, oxidative stress, inflammation and fibrosis. Therefore, dietary iron restriction could be a useful therapeutic approach for NASH patients with hepatic iron overload.

7.
Biol Open ; 8(5)2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-31023717

RESUMO

Non-alcoholic steatohepatitis (NASH) is a fast-growing liver disease in the Western world. Currently, only a few animal models show both the metabolic and histological features of human NASH. We aimed to explore murine NASH models in a time dependent manner that exhibit metabolic, histological and transcriptomic hallmarks of human NASH. For this, the murine strains C57BL/6J, ob/ob, and KK-Ay were used and three types of nutritional regimes were administered: normal chow diet (NCD); high-fat, high-fructose, and high-cholesterol diet (fast food diet; FFD); or choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD), for 2, 4, 8, 12, 18, 24, and 30 weeks. All strains under the FFD and CDAHFD regimes developed steatohepatitis. Among the strains treated with FFD, the non-alcoholic fatty liver disease (NAFLD) activity score, fibrosis progression and metabolic abnormalities such as hyperinsulinemia and obesity were more pronounced in ob/ob mice than in C57BL/6J and KK-Ay mice. In ob/ob mice fed FFD, the development of hepatic crown-like structures was confirmed. Furthermore, molecular pathways involved in steatohepatitis and fibrosis showed significant changes from as early as 2 weeks of starting the FFD regime. Ob/ob mice fed FFD showed metabolic, histological, and transcriptomic dysfunctions similar to human NASH, suggesting their potential as an experimental model to discover novel drugs for NASH.

8.
Nat Rev Gastroenterol Hepatol ; 14(7): 397-411, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28487545

RESUMO

Hepatic fibrosis is a dynamic process characterized by the net accumulation of extracellular matrix resulting from chronic liver injury of any aetiology, including viral infection, alcoholic liver disease and NASH. Activation of hepatic stellate cells (HSCs) - transdifferentiation of quiescent, vitamin-A-storing cells into proliferative, fibrogenic myofibroblasts - is now well established as a central driver of fibrosis in experimental and human liver injury. Yet, the continued discovery of novel pathways and mediators, including autophagy, endoplasmic reticulum stress, oxidative stress, retinol and cholesterol metabolism, epigenetics and receptor-mediated signals, reveals the complexity of HSC activation. Extracellular signals from resident and inflammatory cells including macrophages, hepatocytes, liver sinusoidal endothelial cells, natural killer cells, natural killer T cells, platelets and B cells further modulate HSC activation. Finally, pathways of HSC clearance have been greatly clarified, and include apoptosis, senescence and reversion to an inactivated state. Collectively, these findings reinforce the remarkable complexity and plasticity of HSC activation, and underscore the value of clarifying its regulation in hopes of advancing the development of novel diagnostics and therapies for liver disease.


Assuntos
Transdiferenciação Celular/fisiologia , Células Estreladas do Fígado/patologia , Cirrose Hepática/patologia , Transdiferenciação Celular/genética , Transdiferenciação Celular/imunologia , Citocinas/imunologia , Epigênese Genética/fisiologia , Células Estreladas do Fígado/imunologia , Humanos , Imunidade Inata , Cirrose Hepática/genética , Cirrose Hepática/imunologia , Transdução de Sinais/fisiologia
9.
Sci Rep ; 7: 42563, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28256512

RESUMO

We have used a computational approach to identify anti-fibrotic therapies by querying a transcriptome. A transcriptome signature of activated hepatic stellate cells (HSCs), the primary collagen-secreting cell in liver, and queried against a transcriptomic database that quantifies changes in gene expression in response to 1,309 FDA-approved drugs and bioactives (CMap). The flavonoid apigenin was among 9 top-ranked compounds predicted to have anti-fibrotic activity; indeed, apigenin dose-dependently reduced collagen I in the human HSC line, TWNT-4. To identify proteins mediating apigenin's effect, we next overlapped a 122-gene signature unique to HSCs with a list of 160 genes encoding proteins that are known to interact with apigenin, which identified C1QTNF2, encoding for Complement C1q tumor necrosis factor-related protein 2, a secreted adipocytokine with metabolic effects in liver. To validate its disease relevance, C1QTNF2 expression is reduced during hepatic stellate cell activation in culture and in a mouse model of alcoholic liver injury in vivo, and its expression correlates with better clinical outcomes in patients with hepatitis C cirrhosis (n = 216), suggesting it may have a protective role in cirrhosis progression.These findings reinforce the value of computational approaches to drug discovery for hepatic fibrosis, and identify C1QTNF2 as a potential mediator of apigenin's anti-fibrotic activity.


Assuntos
Antifibrinolíticos/farmacologia , Apigenina/farmacologia , Descoberta de Drogas , Reposicionamento de Medicamentos , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Transcriptoma , Animais , Biomarcadores , Linhagem Celular , Humanos , Camundongos
10.
Sci Rep ; 6: 39342, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27996033

RESUMO

Autophagy and the unfolded protein response (UPR) both promote activation of hepatic stellate cells (HSC), however the link between the two stimuli remains unclear. Here we have explored the role of X-box binding protein 1 (XBP1), one of three UPR effector pathways and sought to establish the interdependence between autophagy and the UPR during HSC activation. XBP1 induction accompanied both culture-based HSC activation and ER stress induced by tunicamycin. Ectopic overexpression of XBP1 induced collagen 1-alpha expression in HSCs, which was inhibited by knockdown of ATG7, a critical autophagy mediator. Genome-wide transcriptomic profiling indicated an upregulation of collagen synthesis pathways, but not of the transforming growth factor (TGF)-b pathway, a canonical fibrogenic driver, suggesting that XBP1 activates a specific subset of fibrogenesis pathways independent of TGF-ß1. XBP1 target gene signatures were significantly induced in rodent liver fibrosis models (n = 3-5) and in human samples of non-alcoholic fatty liver disease (NAFLD) (n = 72-135). Thus, XBP1-mediated UPR contributes to fibrogenic HSC activation and is functionally linked to cellular autophagy.


Assuntos
Autofagia/fisiologia , Células Estreladas do Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Resposta a Proteínas não Dobradas/fisiologia , Proteína 1 de Ligação a X-Box/metabolismo , Animais , Proteína 7 Relacionada à Autofagia/genética , Linhagem Celular , Colágeno Tipo I/biossíntese , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células HEK293 , Humanos , Cirrose Hepática/patologia , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , RNA Interferente Pequeno , Tunicamicina/efeitos adversos , Proteína 1 de Ligação a X-Box/genética
11.
PLoS One ; 10(12): e0145849, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26720709

RESUMO

Type 2 diabetes (T2D) occurs when there is insufficient insulin release to control blood glucose, due to insulin resistance and impaired ß-cell function. The GPR39 receptor is expressed in metabolic tissues including pancreatic ß-cells and has been proposed as a T2D target. Specifically, GPR39 agonists might improve ß-cell function leading to more adequate and sustained insulin release and glucose control. The present study aimed to test the hypothesis that GPR39 agonism would improve glucose stimulated insulin secretion in vivo. A high throughput screen, followed by a medicinal chemistry program, identified three novel potent Zn2+ modulated GPR39 agonists. These agonists were evaluated in acute rodent glucose tolerance tests. The results showed a lack of glucose lowering and insulinotropic effects not only in lean mice, but also in diet-induced obese (DIO) mice and Zucker fatty rats. It is concluded that Zn2+ modulated GPR39 agonists do not acutely stimulate insulin release in rodents.


Assuntos
Descoberta de Drogas , Insulina/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Animais , Glicemia/efeitos dos fármacos , Relação Dose-Resposta a Droga , Teste de Tolerância a Glucose , Ensaios de Triagem em Larga Escala , Humanos , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Ratos , Ratos Zucker , Receptores Acoplados a Proteínas G/genética , Bibliotecas de Moléculas Pequenas , Zinco/metabolismo , Zinco/farmacologia
12.
Metabolism ; 61(7): 944-53, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22154323

RESUMO

Type 2 diabetes mellitus is frequently accompanied by fatty liver/nonalcoholic fatty liver disease. Hence, accumulation of lipids in the liver is considered to be one of the risk factors for insulin resistance and metabolic syndrome. Ursodeoxycholic acid (UDCA) is widely used for the treatment of liver dysfunction. We investigated the therapeutic effects of UDCA on type 2 diabetes mellitus exacerbating hepatic steatosis and the underlying mechanisms of its action using KK-A(y) mice fed a high-fat diet. KK-A(y) mice were prefed a high-fat diet; and 50, 150, and 450 mg/kg of UDCA was orally administered for 2 or 3 weeks. Administration of UDCA decreased fasting hyperglycemia and hyperinsulinemia. Hyperinsulinemic-euglycemic clamp analyses showed that UDCA improved hepatic (but not peripheral) insulin resistance. Hepatic triglyceride and cholesterol contents were significantly reduced by treatment with UDCA, although the genes involved in the synthesis of fatty acids and cholesterol, including fatty acid synthase and 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase, were upregulated. Fecal levels of bile acids, neutral sterols, fatty acids, and phospholipids were significantly increased by UDCA treatment. The gene expression levels and protein phosphorylation levels of endoplasmic reticulum stress markers were not changed by UDCA treatment. These results indicate that UDCA ameliorates hyperglycemia and hyperinsulinemia by improving hepatic insulin resistance and steatosis in high-fat diet-fed KK-A(y) mice. Reduction of hepatic lipids might be due to their excretion in feces, followed by enhanced utilization of glucose for the synthesis of fatty acids and cholesterol. Ursodeoxycholic acid should be effective for the treatment of type 2 diabetes mellitus accompanying hepatic steatosis.


Assuntos
Colagogos e Coleréticos/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/tratamento farmacológico , Resistência à Insulina , Ácido Ursodesoxicólico/uso terapêutico , Animais , Colesterol/análise , Colesterol/biossíntese , Colesterol/genética , Diabetes Mellitus Tipo 2/tratamento farmacológico , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ácidos Graxos/análise , Ácidos Graxos/biossíntese , Ácidos Graxos/genética , Fígado Gorduroso/metabolismo , Perfilação da Expressão Gênica , Hiperglicemia/tratamento farmacológico , Hiperinsulinismo/tratamento farmacológico , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa