Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 238(6): 1226-1236, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37120832

RESUMEN

The perisinusoidal hepatic stellate cells (HSCs) have been investigated extensively for their role as the major fibrogenic cells during chronic liver injury. HSCs also produce numerous cytokines, chemokines, and growth mediators, and express cell adhesion molecules constitutively and in response to stimulants such as endotoxin (lipopolysaccharide). With this property and by interacting with resident and recruited immune and inflammatory cells, HSCs regulate hepatic immune homeostasis, inflammation, and acute injury. Indeed, experiments with HSC-depleted animal models and cocultures have provided evidence for the prominent role of HSCs in the initiation and progression of inflammation and acute liver damage due to various toxic agents. Thus HSCs and/or mediators derived thereof during acute liver damage may be considered as potential therapeutic targets.


Asunto(s)
Hepatopatías , Hígado , Animales , Hígado/metabolismo , Citocinas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Hepatopatías/metabolismo , Inflamación/patología , Cirrosis Hepática/patología
2.
J Cell Physiol ; 238(7): 1530-1541, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37098757

RESUMEN

Binding of lipopolysaccharide (LPS) to CD14 is required for its cellular effects via TLR4. A role of LPS/TLR4-mediated signaling in activated hepatic stellate cells (aHSCs), the major fibrogenic cells, in liver fibrosis has been reported. We investigated effects of LPS on carbon tetrachloride (CCl4)-induced fibrosis in CD14-knockout (KO) mice in vivo, and culture-activated HSCs in vitro. CCl4 (biweekly; 4 weeks)-treated wild type (WT) and CD14-KO mice were challenged with single LPS administration for 24 h. Liver injury, inflammation and fibrosis were determined. Culture-activated HSCs from WT or CD14-KO mice were stimulated with LPS. Parameters of fibrogenic activity (expression of collagen1a1 [Col1a1], α-smooth muscle actin [αSMA] and TGFß1) and inflammatory cytokines/chemokines were measured. CCl4 treatment caused similar liver injury and fibrosis in WT and CD14-KO mice. LPS increased liver injury and inflammation similarly in CCl4-treated WT and CD14-KO mice, but downregulated Timp1 and upregulated Mmp13. LPS elicited similar NFκB activation and inflammatory response in WT and CD14-KO aHSCs. LPS similarly downregulated Acta2 (encodes αSMA), Pdgfrb, Col1a1 and Mmp13 expression but did not affect Timp1 expression in WT and CD14-KO aHSCs. LPS did not alter Tgfb1 but increased expression of decorin (Dcn) (inhibitor of TGFß1) expression in WT and CD14-KO aHSCs. The results indicate that the effects of LPS on HSCs are CD14-independent, and CD14 is not required for hepatic fibrosis. LPS-induced down-modulation of fibrogenic markers in aHSCs is also CD14-independent.


Asunto(s)
Tetracloruro de Carbono , Receptores de Lipopolisacáridos , Lipopolisacáridos , Animales , Ratones , Tetracloruro de Carbono/efectos adversos , Células Estrelladas Hepáticas/metabolismo , Inflamación/patología , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Hígado/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Toll-Like 4/metabolismo
3.
Am J Pathol ; 192(3): 518-535, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34954210

RESUMEN

Acetaminophen (APAP)-induced liver injury is influenced by inflammatory Gram-negative bacterial endotoxin [lipopolysaccharide (LPS)], mechanisms of which are not completely understood. Because LPS-stimulated perisinusoidal hepatic stellate cells (HSCs) produce cytokines that affect survival of hepatocytes, this study investigated their role in APAP-induced liver injury. Fed (nonstarved) rats were administered 5 mg/kg LPS or phosphate-buffered saline (PBS) vehicle, followed by 200 mg/kg APAP or PBS an hour later, and euthanized at 6 hours. Control rats received PBS at both time points. Both LPS and APAP caused mild hepatocyte injury (apoptosis), as assessed by histopathology, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining, and caspase-3 activation. The liver injury was augmented in rats administered LPS + APAP, in association with increased nuclear translocation of interferon-regulatory factor-1 (IRF1). In vitro, APAP augmented LPS/HSC-conditioned medium-induced inhibition of DNA and protein synthesis, apoptosis, and nuclear IRF1 in hepatocytes. LPS-stimulated HSCs produced interferon-ß (IFN-ß), and LPS/HSC + APAP-induced hepatocyte apoptosis was inhibited by anti-IFN-ß antibody. Finally, HSC-depleted mice produced significantly lower IFN-ß and tumor necrosis factor-α, exhibited less oxidative stress, and were protected from excessive injury due to high APAP dose (600 mg/kg), as well as LPS (5 mg/kg overnight) followed by APAP. In co-culture with or without LPS, HSCs increased expression of proinflammatory cytokines by Kupffer cells. These results suggest that HSCs play a critical role in APAP-induced liver injury without or with LPS preconditioning, and it involves INF-ß-IRF1 signaling.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Enfermedad Hepática Inducida por Sustancias y Drogas , Acetaminofén/metabolismo , Acetaminofén/toxicidad , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Citocinas/metabolismo , Endotoxinas/metabolismo , Endotoxinas/toxicidad , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Lipopolisacáridos/toxicidad , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas
4.
J Cell Physiol ; 237(11): 4215-4225, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36098042

RESUMEN

Liver depleted of hepatic stellate cells (HSCs) is resistant to ischemia/reperfusion-, concanavalin A-, and acetaminophen-induced acute injury. Whether HSCs regulate carbon tetrachloride (CCl4 )-induced acute liver injury is not known. CCl4 treatment damages pericentral hepatocytes that express CCl4 -metabolizing Cyp2E1 and activates HSCs. We investigated whether HSC-depletion in mice transgenic for thymidine kinase under the glial fibrillary acidic protein promoter (GFAP-TK-Tg) confers resistance to injury and inflammation due to CCl4 rechallenge. GFAP-TK-Tg or wild type (WT) mice were administered 0.16 ml/kg CCl4 (3× at 3 days intervals), then 40 µg/g/day ganciclovir for 10 days. The treatment depletes ~70%-75% HSCs from GFAP-TK-Tg but not WT mice while the liver recovers from earlier CCl4 -induced injury. Mice were then administered CCl4 , and liver injury and inflammation were determined at 24 h. HSC-depleted and HSC-sufficient mice showed similar CCl4 -induced hepatocyte necrosis and oxidative stress. However, increase in F4/80+ macrophages, but not CD68+ cells, was greater in CCl4 rechallenged HSC-depleted compared to HSC-sufficient mice. Expression of tumor necrosis factor-α (TNF-α), CCL2, and CXCL1 increased similarly, whereas increase in interleukin-6 (IL6), IL1ß, and IL10 expression was higher in CCl4 rechallenged HSC-depleted compared to HSC-sufficient mice. CCl4 rechallenge of HSC-sufficient mice rapidly activated HSCs causing significant fibrosis with increased expression of Col1a1, transforming growth factor ß1 (TGFß1), tissue inhibitors of metalloproteinases 1 (TIMP1); increase in TIPM1 was much lower and metalloproteinases 13 (MMP13) greater in CCl4 rechallenged HSC-depleted mice. Interestingly, hepatic recruitment of both profibrogenic (Ly6Chi ) and antifibrogenic restorative (Ly6Clo ) macrophages, and neutrophils was significantly greater in CCl4 rechallenged HSC-depleted mice. These data suggest that CCl4 directly damages hepatocytes but HSCs regulate inflammation. Rapid fibrogenesis in CCl4 rechallenged HSC-sufficient mice recovered from earlier injury indicates that even transiently activated HSCs that had reverted to the quiescent phenotype remain primed to become reactivated.


Asunto(s)
Tetracloruro de Carbono , Enfermedad Hepática Inducida por Sustancias y Drogas , Ratones , Animales , Tetracloruro de Carbono/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Células Estrelladas Hepáticas/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Inflamación/patología , Cirrosis Hepática/genética
5.
J Hepatol ; 77(5): 1410-1421, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35777586

RESUMEN

Augmenter of liver regeneration (ALR), a ubiquitous fundamental life protein, is expressed more abundantly in the liver than other organs. Expression of ALR is highest in hepatocytes, which also constitutively secrete it. ALR gene transcription is regulated by NRF2, FOXA2, SP1, HNF4α, EGR-1 and AP1/AP4. ALR's FAD-linked sulfhydryl oxidase activity is essential for protein folding in the mitochondrial intermembrane space. ALR's functions also include cytochrome c reductase and protein Fe/S maturation activities. ALR depletion from hepatocytes leads to increased oxidative stress, impaired ATP synthesis and apoptosis/necrosis. Loss of ALR's functions due to homozygous mutation causes severe mitochondrial defects and congenital progressive multiorgan failure, suggesting that individuals with one functional ALR allele might be susceptible to disorders involving compromised mitochondrial function. Genetic ablation of ALR from hepatocytes induces structural and functional mitochondrial abnormalities, dysregulation of lipid homeostasis and development of steatohepatitis. High-fat diet-fed ALR-deficient mice develop non-alcoholic steatohepatitis (NASH) and fibrosis, while hepatic and serum levels of ALR are lower than normal in human NASH and NASH-cirrhosis. Thus, ALR deficiency may be a critical predisposing factor in the pathogenesis and progression of NASH.


Asunto(s)
Regeneración Hepática , Enfermedad del Hígado Graso no Alcohólico , Adenosina Trifosfato/metabolismo , Animales , Citocromos c/metabolismo , Flavina-Adenina Dinucleótido/metabolismo , Humanos , Lípidos , Hígado/patología , Regeneración Hepática/fisiología , Ratones , Mitocondrias/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo
6.
Hepatology ; 72(1): 72-87, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31610032

RESUMEN

BACKGROUND AND AIMS: Yes-associated protein (YAP) plays an important role in hepatocarcinogenesis, although the potential role of YAP in non-neoplastic liver diseases remains largely unknown. We report herein that YAP in Kupffer cells (KCs) enhances the production of proinflammatory cytokines and promotes the development of nonalcoholic steatohepatitis (NASH). Our data show that the expression of YAP is significantly increased in KCs of wild-type mice fed a high-fat diet (HFD). APPROACH AND RESULTS: We generated mice with macrophage/monocyte-specific deletion of YAP (YAPϕKO ) or Toll-like receptor 4 (TLR4; TLR4ϕKO ), and animals were fed an HFD or treated with lipopolysaccharide (LPS). Our data showed that YAPϕKO mice fed an HFD exhibited lower serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) levels and less hepatic inflammation when compared to their littermate controls. LPS treatment induced accumulation of YAP in KCs in vitro and in mice, which was prevented by macrophage/monocyte-specific deletion of TLR4 (TLR4ϕKO ). LPS transcriptionally activates YAP through activator protein 1 in macrophages/KCs. LPS-induced YAP further enhances expression of proinflammatory cytokines (including monocyte chemoattractant protein 1, tumor necrosis factor alpha, and interleukin 6) through YAP association with the TEA domain-binding motif in the promoter region of inflammatory cytokines. Forced overexpression of active YAP (YAP5SA) in KCs enhanced the production of proinflammatory cytokines. Treatment of HFD-fed mice with verteporfin inhibited KC activation, reduced liver inflammation, and decreased serum ALT/AST levels. Analyses of liver tissues from NASH patients reveal that YAP is increased in KCs and that level of YAP in human liver tissues is positively correlated with expression of proinflammatory cytokines. CONCLUSIONS: This study describes an important role of YAP in KCs for regulation of liver inflammation in NASH. Our findings suggest that inhibition of YAP may represent an effective therapeutic strategy for NASH treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de Ciclo Celular/fisiología , Macrófagos del Hígado/metabolismo , Factores de Transcripción/fisiología , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Alanina Transaminasa/sangre , Animales , Aspartato Aminotransferasas/sangre , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/biosíntesis , Macrófagos del Hígado/química , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/análisis , Factores de Transcripción/biosíntesis , Proteínas Señalizadoras YAP
7.
Hepatology ; 72(5): 1800-1818, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32064648

RESUMEN

BACKGROUND AND AIMS: During liver injury, quiescent hepatic stellate cells (qHSCs) transdifferentiate into proliferative and fibrogenic activated myofibroblastic phenotype (activated hepatic stellate cell; aHSCs) expressing smooth muscle α-actin (αSMA) and platelet-derived growth factor beta receptor (PDGFßR). Their interactions with gut-derived bacterial lipopolysaccharide (LPS) are implicated in hepatic fibrogenesis. However, LPS can also attenuate fibrogenic characteristics of aHSCs. APPROACH AND RESULTS: We examined molecular mechanisms of antifibrogenic effects of LPS on aHSCs in vitro and in vivo. Culture-activated rat HSCs were exposed to 0-100 ng/mL of LPS or its active component, diphosphoryl-lipid A (DPLA), and parameters of fibrosis and inflammatory cytokines/chemokines were determined by qRT-PCR, western, and immunohistochemical analyses. In vivo, HSCs were activated by repeated CCl4 administration to rats every 3 days for 3 or 8 weeks, then challenged with LPS (5 mg/kg; IP). HSCs were isolated 24 hours later, and fibrogenic/inflammatory parameters were analyzed. LPS induced phenotypic changes in aHSCs (rounding, size reduction) and loss of proliferation. LPS down-regulated expression of αSMA, PDGFßR, transforming growth factor beta receptor 1 (TGFßR1), collagen 1α1 (Col1α1), and fibronectin while up-regulating tumor necrosis factor alpha, interleukin-6, and C-X-C motif chemokine ligand 1 expression. LPS did not increase peroxisome proliferation-activated receptor gamma expression or lipid accumulation typical of qHSCs. DPLA elicited the same effects as LPS on aHSCs, indicating specificity, and monophosphoryl lipid A down-regulated fibrogenic markers, but elicited very weak inflammatory response. LPS down-regulated the expression of cMyb, a transcription factor for αSMA, and up-regulated small mother against decapentaplegic (SMAD)7 and CCAAT/enhancer-binding protein (C/EBP)δ, the transcriptional inhibitors of Col1α1 expression. In vivo LPS treatment of aHSCs inhibited their proliferation, down-regulated PDGFßR, αSMA, TGFßR1, Col1α1, and cMyb expression, and increased expression of SMAD7, C/EBPα, and C/EBPδ. CONCLUSIONS: In conclusion, LPS induces a unique phenotype in aHSCs associated with down-regulation of key fibrogenic mechanisms and thus may have an important role in limiting fibrosis.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Células Estrelladas Hepáticas/inmunología , Lípido A/análogos & derivados , Cirrosis Hepática Experimental/inmunología , Hígado/patología , Animales , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Tetracloruro de Carbono/administración & dosificación , Tetracloruro de Carbono/toxicidad , Transdiferenciación Celular/inmunología , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Regulación hacia Abajo , Silenciador del Gen , Células Estrelladas Hepáticas/patología , Humanos , Lípido A/inmunología , Lípido A/metabolismo , Hígado/citología , Hígado/inmunología , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/patología , Ratones , Ratones Noqueados , Miofibroblastos/inmunología , Miofibroblastos/patología , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myb/metabolismo , Ratas , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteína smad7/genética , Proteína smad7/metabolismo , Regulación hacia Arriba/inmunología
8.
Hepatology ; 72(5): 1586-1604, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32031683

RESUMEN

BACKGROUND AND AIMS: The augmenter of liver regeneration (ALR) protein is critical for lipid homeostasis and mitochondrial function. We investigated high-fat/high-carbohydrate (HF/HC) diet-induced nonalcoholic fatty liver disease (NAFLD) in wild-type (WT), hepatocyte-specific ALR-knockout (ALR-H-KO), and ALR-heterozygous (ALR-H-HET) mice. ALR was measured in serum of human nonalcoholic steatohepatitis (NASH) and NASH-induced cirrhosis (serum and liver). APPROACH AND RESULTS: HF/HC feeding decreased ALR expression in all groups of mice. The otherwise normal ALR-H-HET mice gained more weight and steatosis than WT mice when challenged metabolically with the HF/HC diet; ALR-H-KO mice gained the least weight and had the least steatosis. These findings were consistent with correspondingly increased triglycerides and cholesterol and altered expression of carnitine palmitoyltransferase 1a, sterol regulatory element-binding protein, acetyl coenzyme A carboxylase, and fatty acid synthase. All HF/HC-fed mice developed insulin resistance, the magnitude being lower in ALR-H-KO mice. HF/HC-fed ALR-H-HET mice were more resistant to glucose challenge than WT or ALR-H-KO mice. The frequency of tumor necrosis factor alpha-producing, interleukin 6 (IL6)-producing, and IL17-producing cells was greater in ALR-H-KO than ALR-H-HET and lowest in WT mice. HF/HC feeding did not increase their number in ALR-H-KO mice, and the increase in ALR-H-HET was greater than that in WT mice except for IL17 cells. Cluster of differentiation 25-positive (CD25+ ) forkhead box P3-positive CD4+ regulatory T-cell frequency was lower in ALR-H-HET than WT mice and further reduced in ALR-H-KO mice; HF/HC reduced regulatory T-cell frequency only in WT mice. HF/HC-fed ALR-H-HET, but not WT, mice developed fibrosis; and ALR-H-KO mice progressed to cirrhosis. White adipose tissue of HF/HC-fed ALR-deficient mice developed strong inflammation, indicating bidirectional interactions with the liver. Hepatic and serum ALR levels were significantly reduced in patients with NASH-cirrhosis. Serum ALR was also significantly lower in patients with NASH. CONCLUSIONS: Hepatic ALR deficiency may be a critical predisposing factor for aggressive NAFLD progression.


Asunto(s)
Cirrosis Hepática/metabolismo , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/deficiencia , Adulto , Anciano , Animales , Biopsia , Colesterol/sangre , Colesterol/metabolismo , Dieta de Carga de Carbohidratos/efectos adversos , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Hepatectomía , Heterocigoto , Humanos , Resistencia a la Insulina , Hígado/cirugía , Cirrosis Hepática/sangre , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Regeneración Hepática , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/sangre , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Triglicéridos/sangre , Triglicéridos/metabolismo
9.
J Magn Reson Imaging ; 54(3): 739-749, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33738856

RESUMEN

BACKGROUND: An imaging method that allows quantitative fibrosis estimates is needed to facilitate the diagnosis of chronic liver disease. Amide proton transfer (APT) and tissue sodium concentration (TSC) estimates could meet this need. HYPOTHESIS: APT and TSC estimates correlate with fibrosis in a mouse model of chronic liver disease. STUDY TYPE: Prospective. PHANTOMS/ANIMAL MODEL: Male C57Bl/6 mice given CCl4 or vehicle (N = 8 each) twice weekly for 16 weeks. FIELD STRENGTH/SEQUENCE: Liver T1 (Look-Locker gradient recalled echo [GRE] sequence), T2 (multiecho spin echo sequence), T1rho (fast spin echo sequence with 500 Hz spin locking pulse), and APT (GRE sequence with off-resonance pulses) data were acquired at 7 T at 12 and 16 weeks. Liver sodium data (multiple echo GRE sequence) were acquired at 12 weeks at 9.4 T. ASSESSMENT: Liver proton T1 , T2 , T1rho , APT, sodium T2 *, and TSC were calculated. Histological measures included Sirius Red, hematoxylin and eosin, liver hydroxyproline content, and serum alanine transaminase (ALT). STATISTICAL TESTS: Welch's two-sided t-test was used to test for differences between control and CCl4 -treated groups for serum ALT, hydroxyproline, Sirius Red staining, T1 , T2 , T1rho , APT, TSC, and sodium T2 *. Pearson's correlations between liver T1 , APT, TSC, or sodium T2 * with Sirius Red staining and hydroxyproline levels were calculated. RESULTS: APT was significantly different (P < 0.05) between groups in the left liver lobe at 16 weeks (CCl4 : 8.0% ± 1.2%, controls: 6.2% ± 1.0%), as were average liver TSC at 12 weeks (CCl4 : 38 mM ± 5 mM, controls: 27 mM ± 2 mM), and average sodium liver T2 * at 12 weeks (CCl4 : 10 msec ± 1.0 msec, controls: 12 msec ± 1.9 msec). APT, TSC, and sodium T2 * correlated significantly (P < 0.05) with Sirius Red staining and hydroxyproline levels. DATA CONCLUSION: Liver TSC and APT significantly correlated with histopathologic markers of fibrosis in this mouse model. EVIDENCE LEVEL: 1 TECHNICAL EFFICACY: Stage 3.


Asunto(s)
Cirrosis Hepática , Imagen por Resonancia Magnética , Animales , Hígado/diagnóstico por imagen , Hígado/patología , Cirrosis Hepática/diagnóstico por imagen , Cirrosis Hepática/patología , Masculino , Ratones , Fantasmas de Imagen , Estudios Prospectivos
10.
FASEB J ; 33(3): 3825-3840, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30540918

RESUMEN

Levels of augmenter of liver regeneration (ALR), a multifunctional protein, are reduced in steatohepatitis. ALR depletion from ALR flox/flox/Alb-Cre [ALR-L-knockout (KO)] mouse causes robust steatosis and apoptosis of hepatocytes, and pericellular fibrosis between 1 and 2 wk postbirth. Steatosis regresses by 4 wk upon reappearance of ALR-expressing hepatocytes. We investigated mechanisms of ALR depletion-induced steatosis. ALR-L-KO mice (1-, 2-, and 4 wk old) and Adeno-Cre-transfected ALR flox/flox hepatocytes were used for in vivo and in vitro studies. ALR depletion from hepatocytes in vivo downregulated peroxisome proliferator-activated receptor (PPAR)-α, carnitine palmitoyl transferase I (CPT1)a, peroxisomal membrane protein 70 (PMP70) (modest down-regulation), and acyl-CoA oxidase 1 (ACOX1). The markedly up-regulated (20X) novel microRNA-540 (miR-540) was identified to target PPARα, PMP70, ACOX1, and CPT1a. ALR depletion from primary hepatocytes increased oxidative stress, miR-540 expression, and steatosis and down-regulated PPARα, ACOX1, PMP70, and CPT1a expression. Anti-miR-540 mitigated ALR depletion-induced steatosis and prevented loss of PPARα, ACOX1, PMP70, and CPT1a expression. Antioxidant N-acetylcysteine and recombinant ALR (rALR) both inhibited ALR depletion-induced miR-540 expression and lipid accumulation in hepatocytes. Finally, treatment of ALR-L-KO mice with rALR between 1 and 2 wk prevented miR-540 expression, and arrested steatosis and fibrosis. We conclude that ALR deficiency-mediated oxidative stress induces generation of miR-540, which promotes steatosis by dysregulating peroxisomal and mitochondrial lipid homeostasis.-Kumar, S., Rani, R., Karns, R., Gandhi, C. R. Augmenter of liver regeneration protein deficiency promotes hepatic steatosis by inducing oxidative stress and microRNA-540 expression.


Asunto(s)
Hígado Graso/genética , Regeneración Hepática/genética , MicroARNs/genética , Deficiencia de Proteína/genética , Animales , Apoptosis/genética , Retículo Endoplásmico/genética , Femenino , Hepatocitos/patología , Humanos , Hígado/patología , Ratones , Ratones Noqueados , Mitocondrias/genética , Estrés Oxidativo/genética , PPAR alfa/genética , Regulación hacia Arriba/genética
11.
J Biol Chem ; 293(48): 18466-18476, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30348900

RESUMEN

Mice depleted of hepatic stellate cells (HSCs) are protected from concanavalin A (ConA)-induced liver injury that is mediated by the activation of interferon regulatory factor 1 (IRF1). The aim of this study was to determine the mechanisms of ConA-mediated signaling and synthesis/release of mediators by HSCs that damage hepatocytes. Primary cultures of wildtype (WT) and IRF1-knockout (KO) HSCs and hepatocytes were used, and ConA-induced liver damage in interferon (IFN)αß receptor-deficient (IFNαßR-KO) mice was determined. Specific binding of ConA to HSCs induced rapid activation of JAK2 and STAT1. ConA-induced expression of IRF1, IFNß, tumor necrosis factor α, and CXCL1 was abrogated by selective inhibition of JAK2 and STAT1. Despite activating JAK2/STAT1, ConA failed to stimulate expression of inflammatory cytokines in HSCs from IRF1-KO mice. ConA-conditioned WT-HSC medium caused activation of JNK and caspase 3, and apoptosis of hepatocytes from WT but not from IRF1-KO or IFNαßR-KO mice. Conversely, ConA-conditioned medium of IRF1-KO HSCs failed to cause apoptosis of WT or IRF1-KO hepatocytes. IFNαßR-KO mice were protected from ConA-induced liver damage, and ConA-induced hepatic expression of IRF1 and pro-inflammatory cytokines and chemokines, and infiltration of neutrophils were significantly lower in IFNαßR-KO than in WT mice. These results demonstrate distinct roles of IRF1 in hepatic inflammation (HSCs) and injury (hepatocytes) and can be an important target for intervention in acute liver injury.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Concanavalina A/farmacología , Citocinas/biosíntesis , Células Estrelladas Hepáticas/efectos de los fármacos , Factor 1 Regulador del Interferón/fisiología , Animales , Células Cultivadas , Medios de Cultivo Condicionados , Citocinas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Factor 1 Regulador del Interferón/genética , Interferón gamma/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Transducción de Señal , Superóxido Dismutasa-1/metabolismo
12.
Am J Pathol ; 187(9): 2008-2019, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28710903

RESUMEN

Concanavalin A (ConA) causes immune cell-mediated liver damage, but the contribution of resident nonparenchymal cells (NPCs) is also evident. Hepatic stellate cells (HSCs) induce hepatic inflammation and immunological reactions; we therefore investigated their role in ConA-induced liver injury. ConA was administered i.v. to control or HSC-depleted mice; hepatic histopathology and cytokines/chemokines were determined after 6 hours. In vitro, effects of ConA-conditioned HSC medium on hepatocytes were determined. ConA induced inflammation, sinusoidal congestion, and extensive midzonal hepatocyte death in control mice, which were strongly minimized in HSC-depleted mice. CD4 and natural killer T cells and neutrophils were markedly reduced in ConA-treated HSC-depleted mice compared with control mice. The increase in cytokines/chemokines of hepatic injury was much higher in ConA-treated control mice than in HSC-depleted mice. ConA-treated HSCs showed increased expression of interferon-ß, tumor necrosis factor-α, and CXCL1, induced oxidative stress in hepatocytes, and caused hepatocyte apoptosis. ConA induced nuclear translocation of interferon-regulatory factor-1 (IRF1) in hepatocytes in vivo, and ConA/HSC induced a similar effect in cultured hepatocytes. IRF1-knockout mice were resistant to ConA-induced liver damage, and anti-interferon ß antibody mitigated ConA/HSC-induced injury. In HSC-NPC co-culture, ConA-induced expression of inflammatory cytokines/chemokines was significantly augmented compared with NPCs alone. HSCs play an essential role in ConA-induced liver injury directly via the interferon-ß/IRF1 axis, and by modulating properties of NPCs.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Concanavalina A/toxicidad , Células Estrelladas Hepáticas/patología , Hígado/patología , Animales , Apoptosis/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones
13.
Am J Pathol ; 187(6): 1356-1367, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28412299

RESUMEN

Gram-negative bacterial endotoxin lipopolysaccharide (LPS) is implicated in acute and chronic liver injury; its effects are mediated predominantly via the membrane receptor Toll-like receptor 4 (TLR4). However, TLR4-independent effects of LPS may play important role in hepatic pathophysiology. We investigated carbon tetrachloride (CCl4)-induced fibrosis and LPS-induced acute liver injury in wild-type (WT) and B6.B10ScN-Tlr4lps-del/JthJ [TLR4-knockout (KO)] mice. Effects of LPS on fibrogenic hepatic stellate cells (HSCs) from WT and TLR4-KO mice were assessed in vitro. CCl4 produced similar fibrosis and necroinflammation and increased the mRNA and protein expression of cytokines and chemokines in WT and TLR4-KO mice. However, circulating LPS concentration did not increase in CCl4-treated mice. Interestingly, LPS down-modulated α-smooth muscle actin (activated HSC marker) and collagen 1 in both WT and TLR4-KO HSCs. LPS induced similar activation of NF-κB, and stimulated the expression of cytokines and chemokines in WT and TLR4-KO HSCs. Finally, LPS caused similar inflammation and injury in previously untreated WT and TLR4-KO mice. The results provide evidence of the TLR4/LPS-independent mechanisms of liver fibrosis and also indicate that TLR4 is not entirely critical to LPS-induced acute liver injury. The results further indicate that LPS signaling in activated HSCs might be a mechanism of limiting liver fibrosis.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática Experimental/patología , Receptor Toll-Like 4/fisiología , Actinas/biosíntesis , Actinas/genética , Animales , Tetracloruro de Carbono , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Quimiocinas/biosíntesis , Quimiocinas/genética , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Citocinas/biosíntesis , Citocinas/genética , Regulación hacia Abajo/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Lipopolisacáridos/farmacología , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Receptor Toll-Like 4/deficiencia
14.
J Cell Physiol ; 231(1): 94-105, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26031389

RESUMEN

Bacterial lipopolysaccharide (LPS)-stimulated hepatic stellate cells (HSCs) produce many cytokines including IFNß, TNFα, and IL6, strongly inhibit DNA synthesis, but induce apoptosis of a small number of hepatocytes. In vivo administration of LPS (up to 10 mg/mL) causes modest inflammation and weight loss in rats but not mortality. We determined whether LPS-stimulated HSCs instigate mechanisms of hepatocyte survival. Rats received 10 mg/kg LPS (i.p.) and determinations were made at 6 h. In vitro, HSCs were treated with 100 ng/mL LPS till 24 h. The medium was transferred to hepatocytes, and determinations were made at 0-12 h. Controls were HSC-conditioned medium or medium-containing LPS. LPS treatment of rats caused autophagy in hepatocytes, a physiological process for clearance of undesirable material including injured or damaged organelles. This was accompanied by activation of c-Jun NH2 terminal kinase (JNK) and apoptosis of ~4-5% of hepatocytes. In vitro, LPS-conditioned HSC medium (LPS/HSC) induced autophagy in hepatocytes but apoptosis of only ~10% of hepatocytes. While LPS/HSC stimulated activation of JNK (associated with cell death), it also activated NFkB and ERK1/2 (associated with cell survival). LPS-stimulated HSCs produced IFNß, and LPS/HSC-induced autophagy in hepatocytes and their apoptosis were significantly inhibited by anti-IFNß antibody. Blockade of autophagy, on the other hand, strongly augmented hepatocyte apoptosis. While LPS-stimulated HSCs cause apoptosis of a subpopulation of hepatocytes by producing IFNß, they also induce cell survival mechanisms, which may be of critical importance in resistance to liver injury during endotoxemia.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Endotoxinas/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Células Estrelladas Hepáticas/citología , Hepatocitos/citología , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratas Sprague-Dawley
15.
Gastroenterology ; 148(2): 379-391.e4, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25448926

RESUMEN

BACKGROUND & AIMS: Augmenter of liver regeneration (ALR, encoded by GFER) is a widely distributed pleiotropic protein originally identified as a hepatic growth factor. However, little is known about its roles in hepatic physiology and pathology. We created mice with liver-specific deletion of ALR to study its function. METHODS: We developed mice with liver-specific deletion of ALR (ALR-L-KO) using the albumin-Cre/LoxP system. Liver tissues were collected from ALR-L-KO mice and ALR(floxed/floxed) mice (controls) and analyzed by histology, reverse-transcription polymerase chain reaction, immunohistochemistry, electron microscopy, and techniques to measure fibrosis and lipids. Liver tissues from patients with and without advanced liver disease were determined by immunoblot analysis. RESULTS: Two weeks after birth, livers of ALR-L-KO mice contained low levels of ALR and adenosine triphosphate (ATP); they had reduced mitochondrial respiratory function and increased oxidative stress, compared with livers from control mice, and had excessive steatosis, and hepatocyte apoptosis. Levels of carbamyl-palmitoyl transferase 1a and ATP synthase subunit ATP5G1 were reduced in livers of ALR-L-KO mice, indicating defects in mitochondrial fatty acid transport and ATP synthesis. Electron microscopy showed mitochondrial swelling with abnormalities in shapes and numbers of cristae. From weeks 2-4 after birth, levels of steatosis and apoptosis decreased in ALR-L-KO mice, and numbers of ALR-expressing cells increased, along with ATP levels. However, at weeks 4-8 after birth, livers became inflamed, with hepatocellular necrosis, ductular proliferation, and fibrosis; hepatocellular carcinoma developed by 1 year after birth in nearly 60% of the mice. Hepatic levels of ALR were also low in ob/ob mice and alcohol-fed mice with liver steatosis, compared with controls. Levels of ALR were lower in liver tissues from patients with advanced alcoholic liver disease and nonalcoholic steatohepatitis than in control liver tissues. CONCLUSIONS: We developed mice with liver-specific deletion of ALR, and showed that it is required for mitochondrial function and lipid homeostasis in the liver. ALR-L-KO mice provide a useful model for investigating the pathogenesis of steatohepatitis and its complications.


Asunto(s)
Carcinoma Hepatocelular/etiología , Hígado Graso/etiología , Neoplasias Hepáticas/etiología , Regeneración Hepática/fisiología , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/fisiología , Animales , Apoptosis , Reductasas del Citocromo/fisiología , Humanos , Metabolismo de los Lípidos , Cirrosis Hepática Experimental/etiología , Ratones , Ratones Noqueados , Mitocondrias/fisiología
16.
Hepatology ; 72(3): 1157-1158, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32219876
17.
Hepatol Res ; 45(3): 305-14, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24796378

RESUMEN

AIM: Liver fibrosis occurs as a result of several chronic liver diseases and leads to portal hypertension, cirrhosis and liver failure, often requiring liver transplantation. Activated hepatic stellate cells (HSC) are known to contribute to liver fibrosis, but currently there are no effective therapies for the treatment of established liver fibrosis. Activation of the acidic sphingomyelinase (ASM) has been shown to be involved in HSC activation. In the present study we investigated whether treatment with the ASM inhibitor, amitriptyline (TCA), could prevent and/or reverse fibrosis induced in mice by carbon tetrachloride (CCl4 ). METHODS: Mice were treated with CCl4 for 8 weeks to induce fibrosis. Concurrently, mice received drinking water with or without 180 mg/L TCA. RESULTS: Mice receiving TCA in the water had decreased hepatic collagen deposition and reduced liver mRNA expression of the fibrogenic mediators, transforming growth factor (TGF)-ß1, tissue inhibitor of matrix metalloproteinase-1, collagen and tumor necrosis factor-α. TCA treatment also reduced HSC activation determined by α-smooth muscle actin staining. In a separate set of experiments, mice were treated with CCl4 for 5 weeks prior to treatment with TCA, to test whether TCA had any effect on established fibrosis. Remarkably, in mice with established fibrosis, treatment with TCA significantly reduced collagen deposition, HSC activation, and prevented portal hypertension and improved hepatic architecture. Treatment of isolated HSC in vitro with TCA completely inhibited TGF-ß1-induced collagen expression and platelet-derived growth factor-ß-ß-induced proliferation. CONCLUSION: The data suggest that ASM is a critical signaling component in HSC for the development of liver fibrosis and represents an important therapeutic target.

18.
J Hepatol ; 60(2): 298-305, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24060854

RESUMEN

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) that express glial fibrillary acidic protein (GFAP) are located between the sinusoidal endothelial cells and hepatocytes. HSCs are activated during liver injury and cause hepatic fibrosis by producing excessive extracellular matrix. HSCs also produce many growth factors, chemokines and cytokines, and thus may play an important role in acute liver injury. However, this function has not been clarified due to unavailability of a model, in which HSCs are depleted from the normal liver. METHODS: We treated mice expressing HSV-thymidine kinase under the GFAP promoter (GFAP-Tg) with 3 consecutive (3 days apart) CCl4 (0.16 µl/g; ip) injections to stimulate HSCs to enter the cell cycle and proliferate. This was followed by 10-day ganciclovir (40 µg/g/day; ip) treatment, which is expected to eliminate actively proliferating HSCs. Mice were then subjected to hepatic ischemia/reperfusion (I/R) or endotoxin treatment. RESULTS: CCl4/ganciclovir treatment caused depletion of the majority of HSCs (about 64-72%), while the liver recovered from the initial CCl4-induced injury (confirmed by histology, serum ALT and neutrophil infiltration). The magnitude of hepatic injury due to I/R or endotoxemia (determined by histopathology and serum ALT) was lower in HSC-depleted mice. Their hepatic expression of TNF-α, neutrophil chemoattractant CXCL1 and endothelin-A receptor also was significantly lower than the control mice. CONCLUSIONS: HSCs play an important role both in I/R- and endotoxin-induced acute hepatocyte injury, with TNF-α and endothelin-1 as important mediators of these effects.


Asunto(s)
Células Estrelladas Hepáticas/patología , Células Estrelladas Hepáticas/fisiología , Hígado/lesiones , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Animales , Tetracloruro de Carbono/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Quimiocina CXCL1/genética , Modelos Animales de Enfermedad , Endotelina-1/genética , Ganciclovir/toxicidad , Expresión Génica , Proteína Ácida Fibrilar de la Glía , Células Estrelladas Hepáticas/efectos de los fármacos , Interleucina-6/genética , Lipopolisacáridos/toxicidad , Hígado/patología , Hígado/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Daño por Reperfusión/genética , Timidina Quinasa/genética , Factor de Necrosis Tumoral alfa/genética
20.
J Immunol ; 188(8): 3667-77, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22427640

RESUMEN

Hepatic stellate cells (HSCs) may play an important role in hepatic immune regulation by producing numerous cytokines/chemokines and expressing Ag-presenting and T cell coregulatory molecules. Due to disruption of the endothelial barrier during cold-ischemic storage and reperfusion of liver grafts, HSCs can interact directly with cells of the immune system. Endotoxin (LPS), levels of which increase in liver diseases and transplantation, stimulates the synthesis of many mediators by HSCs. We hypothesized that LPS-stimulated HSCs might promote hepatic tolerogenicity by influencing naturally occurring immunosuppressive CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs). Following their portal venous infusion, allogeneic CD4(+) T cells, including Tregs, were found closely associated with HSCs, and this association increased in LPS-treated livers. In vitro, both unstimulated and LPS-stimulated HSCs upregulated Fas (CD95) expression on conventional CD4(+) T cells and induced their apoptosis in a Fas/Fas ligand-dependent manner. By contrast, HSCs induced Treg proliferation, which required cell-cell contact and was MHC class II-dependent. This effect was augmented when HSCs were pretreated with LPS. LPS increased the expression of MHC class II, CD80, and CD86 and stimulated the production of IL-1α, IL-1ß, IL-6, IL-10 and TNF-α by HSCs. Interestingly, production of IL-1α, IL-1ß, IL-6, and TNF-α was strongly inhibited, but that of IL-10 enhanced in LPS-pretreated HSC/Treg cocultures. Adoptively transferred allogeneic HSCs migrated to the secondary lymphoid tissues and induced Treg expansion in lymph nodes. These data implicate endotoxin-stimulated HSCs as important immune regulators in liver transplantation by inducing selective expansion of tolerance-promoting Tregs and reducing inflammation and alloimmunity.


Asunto(s)
Comunicación Celular/inmunología , Endotoxinas/farmacología , Regulación de la Expresión Génica/inmunología , Trasplante de Hígado/inmunología , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante/inmunología , Traslado Adoptivo , Animales , Antígenos CD/biosíntesis , Antígenos CD/inmunología , Apoptosis , Comunicación Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Citocinas/inmunología , Genes MHC Clase II , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/inmunología , Linfocitos T Reguladores/citología , Factores de Transcripción/biosíntesis , Factores de Transcripción/inmunología , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA