Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Molecules ; 26(6)2021 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-33810101

RESUMEN

To date, there is no effective treatment for alcoholic liver disease, despite its prevalence world-wide. Because alcohol consumption is associated with oxidative stress-induced liver injury and pro-inflammatory responses, naturally occurring antioxidants and/or anti-inflammatories may be potential therapeutics. Spermidine is an abundant, ubiquitous polyamine that has been found to display strong antioxidant and anti-inflammatory properties. To further investigate whether spermidine is an effective intervention for alcohol-induced liver disease, we examined its hepatoprotective properties using a two-hit, chronic ethanol and acute lipopolysaccharide (LPS)-induced mouse model of liver injury. We determined that spermidine administration prevented ethanol and LPS-induced increases in liver injury using plasma ALT as a readout. Furthermore, histological analysis of tissue from control and treated animals revealed that the pathology associated with ethanol and LPS treatment was prevented in mice additionally treated with spermidine. As predicted, spermidine also prevented ethanol and LPS-induced oxidative stress by decreasing the levels of both reactive oxygen species (ROS) and lipid peroxidation. We further determined that spermidine treatment prevented the nuclear translocation of nuclear factor κB (NFκB) by blocking the phosphorylation of the inhibitory protein, IκB, thereby preventing expression of pro-inflammatory cytokines. Finally, by measuring expression of known markers of hepatic stellate cell activation and monitoring collagen deposition, we observed that spermidine also prevented alcohol and LPS-induced hepatic fibrosis. Together, our results indicate that spermidine is an antioxidant thereby conferring anti-inflammatory and anti-fibrotic effects associated with alcoholic liver injury.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Etanol/toxicidad , Lipopolisacáridos/toxicidad , Hepatopatías Alcohólicas/prevención & control , Hígado/metabolismo , Espermidina/farmacología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Femenino , Hígado/patología , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/patología , Ratones
2.
Biochem Pharmacol ; 183: 114323, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33166508

RESUMEN

Alcoholic liver disease (ALD) is closely linked to oxidative stress induction. Antioxidant enzymes balance oxidative stress and function as intermediary signaling regulators. Nucleoredoxin (NXN), an antioxidant enzyme, regulates physiological processes through redox-sensitive interactions. NXN interacts with myeloid differentiation primary response gene-88 (MYD88) and flightless-I (FLII) to regulate toll-like receptor 4 (TLR4)/MYD88 pathway activation, but FLII also regulates key cell processes and is secreted into the bloodstream. However, the effects of chronic ethanol consumption recapitulated by either ethanol alone or in combination with lipopolysaccharides (LPS), as a two-hit ALD model, on FLII/NXN/MYD88 complex and FLII secretion have not been explored yet. In this study, we have demonstrated that ethanol feeding increased FLII protein levels, its nuclear translocation and plasma secretion, and modified its tissue distribution both in vivo and in vitro ALD models. Ethanol increased MYD88/FLII interaction ratio, and decreased NXN/MYD88 interaction ratio but this was partially reverted by two-hit model. While ethanol and two-hit model increased MYD88/TLR4 interaction ratio, two-hit model significantly decreased FLII nuclear translocation and its plasma secretion. Ethanol and LPS provoked similar effects in vitro; however, NXN overexpression partially reverted these alterations, and ethanol alone increased FLII secretion into culture medium. In summary, by analyzing the response of FLII/NXN/MYD88 complex during ALD early progression both in vivo and in vitro, we have discovered that the effects of chronic ethanol consumption disrupt this complex and identified FLII as a candidate non-invasive plasma biomarker for the early detection of ALD.


Asunto(s)
Etanol/toxicidad , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Hepatopatías Alcohólicas/metabolismo , Proteínas de Microfilamentos/metabolismo , Transactivadores/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Diagnóstico Precoz , Femenino , Humanos , Hepatopatías Alcohólicas/diagnóstico , Ratones , Ratones Endogámicos C57BL
3.
Basic Clin Pharmacol Toxicol ; 127(5): 389-404, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32524749

RESUMEN

Alcoholic liver disease (ALD) may be attributed to multiple hits driving several alterations. The aim of this work was to determine whether nucleoredoxin (NXN) interacts with flightless-I (FLII)/actin complex and how this ternary complex is altered during ALD progression induced by different ALD models. ALD was recapitulated in C57BL/6J female mice by the well-known ALD Lieber-DeCarli model, and by an in vitro human co-culture system overexpressing NXN. The effects of ethanol and low doses of lipopolysaccharides (LPS) and diethylnitrosamine (DEN) were also evaluated in vivo as a first approach of an ALD multi-hit protocol. We demonstrated that NXN interacts with FLII/actin complex. This complex was differentially altered in ALD in vivo and in vitro, and NXN overexpression partially reverted this alteration. We also showed that ethanol, LPS and DEN synergistically induced liver structural disarrangement, steatosis and inflammatory infiltration accompanied by increased levels of proliferation (Ki67), ethanol metabolism (CYP2E1), hepatocarcinogenesis (GSTP1) and LPS-inducible (MYD88 and TLR4) markers. In summary, we provide evidence showing that NXN/FLII/actin complex is involved in ALD progression and that NXN might be involved in the regulation of FLII/actin-dependent cellular functions. Moreover, we present a promising first approach of a multi-hit protocol to better recapitulate ALD pathogenesis.


Asunto(s)
Hígado Graso/metabolismo , Hígado Graso/patología , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/patología , Proteínas de Microfilamentos/metabolismo , Oxidorreductasas/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP2E1/metabolismo , Dietilnitrosamina/farmacología , Etanol , Femenino , Lipopolisacáridos/farmacología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL
4.
Cells ; 8(11)2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31671832

RESUMEN

Akt kinase isoforms (Akt1, Akt2, and Akt3) have generally been thought to play overlapping roles in phosphoinositide 3-kinase (PI3K)-mediated-signaling. However, recent studies have suggested that they display isoform-specific roles in muscle and fat. To determine whether such isoform-specificity is observed with respect to alcoholic liver disease (ALD) progression, we examined the role of Akt1, Akt2, and Akt3 in hepatic inflammation, and pro-fibrogenic proliferation and migration using Kupffer cells, hepatic stellate cells (HSC), and hepatocytes in an ethanol and lipopolysaccharide (LPS)-induced two-hit model in vitro and in vivo. We determined that siRNA-directed silencing of Akt2, but not Akt1, significantly suppressed cell inflammatory markers in HSC and Kupffer cells. Although both Akt1 and Akt2 inhibited cell proliferation in HSC, only Akt2 inhibited cell migration. Both Akt1 and Akt2, but not Akt3, inhibited fibrogenesis in hepatocytes and HSC. In addition, our in vivo results show that administration of chronic ethanol, binge ethanol and LPS (EBL) in wild-type C57BL/6 mice activated all three Akt isoforms with concomitant increases in activated forms of phosphoinositide dependent kinase-1 (PDK1), mammalian target-of-rapamycin complex 2 (mTORC2), and PI3K, resulting in upregulation in expression of inflammatory, proliferative, and fibrogenic genes. Moreover, pharmacological blocking of Akt2, but not Akt1, inhibited EBL-induced inflammation while blocking of both Akt1 and Akt2 inhibited pro-fibrogenic marker expression and progression of fibrosis. Our findings indicate that Akt isoforms play unique roles in inflammation, cell proliferation, migration, and fibrogenesis during EBL-induced liver injury. Thus, close attention must be paid when targeting all Akt isoforms as a therapeutic intervention.


Asunto(s)
Hepatitis/genética , Cirrosis Hepática/genética , Hepatopatías Alcohólicas/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , Animales , Células Cultivadas , Progresión de la Enfermedad , Etanol/farmacología , Femenino , Células Hep G2 , Hepatitis/etiología , Hepatitis/patología , Humanos , Isoenzimas/fisiología , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Hepatopatías Alcohólicas/complicaciones , Hepatopatías Alcohólicas/patología , Ratones , Ratones Endogámicos C57BL
5.
Toxicol Appl Pharmacol ; 378: 114611, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31176654

RESUMEN

Hepatocellular carcinoma (HCC) arises after a long period of exposition to etiological factors that might be either independent or collectively contributing. Several rodent models resemble human HCC; however, the major limitation of these models is the lack of chronic injury that reproducibly mimics the molecular alterations as it occurs in humans. Thus, we hypothesized that chronic administration of different DEN treatments identifies the best-fit dose to induce the HCC and/or to determine whether small DEN doses act synergistically with other known hepatotoxins to induce HCC in mice. C57BL/6 J male mice were intraperitoneally injected twice a week for 6 weeks with different DEN doses ranging from 2.5 to 40 mg/kg body weight; then, selected doses (2.5, 5 and 20 mg/kg) for 6, 10, 14, and 18 weeks. We demonstrated that DEN at 20 mg/kg promoted reactive oxygen species and 4-hydroxynonenal production, cell proliferation inflammatory infiltrate, and fibrosis, which in turn induced liver cancer by week 18. These parameters were established by evaluating histopathological changes, HCC markers such as glutathione S-transferase placental-1 (Gstp1), Cytokeratin-19 (Ck19) and prostaglandin reductase-1 (Ptgr1); that of Cyp2e1, a DEN metabolizing enzyme; and the expression of the proliferation marker Ki67. While DEN at 2.5 and 5 mg/kg increased Gstp1 and Ck19, DEN at 20 mg/kg decreased them and Cyp2e1 expression and activity. In summary, our results demonstrate that DEN chronically administrated at 20 mg/kg induces the HCC, while DEN at 2.5 and 5 mg/kg could be useful in elucidating its synergistic effect with other hepatotoxic agents in mice.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Dietilnitrosamina/administración & dosificación , Dietilnitrosamina/efectos adversos , Neoplasias Hepáticas/inducido químicamente , Hígado/efectos de los fármacos , Animales , Carcinogénesis/metabolismo , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/metabolismo , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Inflamación/metabolismo , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo
6.
J Appl Toxicol ; 39(8): 1118-1131, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30883860

RESUMEN

Rebaudioside A (Reb A) is a diterpenoid isolated from the leaves of Stevia rebaudiana (Bertoni) that has been shown to possess pharmacological activity, including anti-inflammatory and antioxidant properties. However, the ability of Reb A to prevent liver injury has not been evaluated. Therefore, we aimed to study the potential of Reb A (20 mg/kg; two times daily intraperitoneally) to prevent liver injury induced by thioacetamide (TAA) administration (200 mg/kg; three times per week intraperitoneally). In addition, cocultures were incubated with either lipopolysaccharide or ethanol. Antifibrotic, antioxidant and immunological responses were evaluated. Chronic TAA administration produced considerable liver damage and distorted the liver parenchyma with the presence of prominent thick bands of collagen. In addition, TAA upregulated the expression of α-smooth muscle actin, transforming growth factor-ß1, metalloproteinases 9, 2 and 13, and nuclear factor kappaB and downregulated nuclear erythroid factor 2. Reb A administration prevented all of these changes. In cocultured cells, Reb A prevented the upregulation of genes implicated in fibrotic and inflammatory processes when cells were exposed to ethanol and lipopolysaccharide. Altogether, our results suggest that Reb A prevents liver damage by blocking oxidative processes via upregulation of nuclear erythroid factor 2, exerts immunomodulatory effects by downregulating the nuclear factor-κB system and acts as an antifibrotic agent by maintaining collagen content.


Asunto(s)
Antioxidantes/uso terapéutico , Diterpenos de Tipo Kaurano/uso terapéutico , Cirrosis Hepática/prevención & control , Hígado/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Antioxidantes/aislamiento & purificación , Antioxidantes/farmacología , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Diterpenos de Tipo Kaurano/aislamiento & purificación , Diterpenos de Tipo Kaurano/farmacología , Expresión Génica/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/genética , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Estrés Oxidativo/genética , Ratas , Ratas Wistar , Stevia/química , Tioacetamida/toxicidad
7.
Life Sci ; 224: 187-196, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30890404

RESUMEN

AIMS: Stevioside is a diterpenoid obtained from the leaves of Stevia rebaudiana (Bertoni) that exhibits antioxidant, antifibrotic, antiglycemic and anticancer properties. Therefore, we aimed to study whether stevioside has beneficial effects in liver injury induced by long-term thioacetamide (TAA) administration and investigated the possible underlying molecular mechanism using in vivo, in vitro and in silico approaches. MAIN METHODS: Liver injury was induced in male Wistar rats by TAA administration (200 mg/kg), intraperitoneally, three times per week. Rats received saline or stevioside (20 mg/kg) twice daily intraperitoneally. In addition, cocultures were incubated with either lipopolysaccharide or ethanol. Liver injury, antioxidant and immunological responses were evaluated. KEY FINDINGS: Chronic TAA administration induced significant liver damage. In addition, TAA upregulated the protein expression of nuclear factor (NF)-κB, thus increasing the expression of proinflammatory cytokines and decreasing the antioxidant capacity of the liver through downregulation of nuclear erythroid factor 2 (Nrf2). Notably, stevioside administration prevented all of these changes. In vitro, stevioside prevented the upregulation of several genes implicated in liver inflammation when cocultured cells were incubated with lipopolysaccharide or ethanol. In silico assays using tumor necrosis factor receptor (TNFR)-1 and Toll-like receptor (TLR)-4-MD2 demonstrated that stevioside docks with TNFR1 and TLR4-MD2, thus promoting an antagonistic action against this proinflammatory mediator. SIGNIFICANCE: Collectively, these data suggest that stevioside prevented liver damage through antioxidant activity by upregulating Nrf2 and immunomodulatory activity by blocking NF-κB signaling.


Asunto(s)
Antioxidantes/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Diterpenos de Tipo Kaurano/farmacología , Glucósidos/farmacología , Factores Inmunológicos/farmacología , Edulcorantes/farmacología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Simulación por Computador , Técnicas In Vitro , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Tioacetamida/toxicidad
8.
Basic Clin Pharmacol Toxicol ; 124(6): 670-680, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30561898

RESUMEN

Liver cirrhosis is associated with increased morbidity and mortality with important health and social consequences; however, an effective treatment has not been found yet. Previous reports have shown some beneficial effects of stevioside (SVT) in different diseases, but the ability of SVT to inhibit liver cirrhosis has not been reported. Therefore, we studied the potential of this diterpenoid to inhibit liver cirrhosis induced by thioacetamide, a model that shares many similarities with the human disease, and investigated the possible underlying molecular mechanism using in vivo and in vitro approaches. Cirrhosis was induced in male Wistar rats by chronic thioacetamide administration (200 mg/kg) intraperitoneally three times per week. Rats received saline or SVT (20 mg/kg) two times daily intraperitoneally. In addition, co-cultures were incubated with either lipopolysaccharide or ethanol. Liver fibrosis, hepatic stellate cells activation, metalloproteinases activity, canonical and non-canonical Smads pathway and expression of several profibrogenic genes were evaluated. Thioacetamide activated hepatic stellate cells and distorted the liver parenchyma with the presence of abundant thick bands of collagen. In addition, thioacetamide up-regulated the protein expression of α-smooth muscle actin, transforming growth factor-ß1, metalloproteinases-9,-2 and -13 and overstimulate the canonical and non-canonical Smad pathways. SVT administration inhibited all of these changes. In vitro, SVT inhibited the up-regulation of several genes implicated in cirrhosis when cells were exposed to lipopolysaccharides or ethanol. We conclude that SVT inhibited liver damage by blocking hepatic stellate cells activation, down-regulating canonical and non-canonical profibrotic Smad pathways.


Asunto(s)
Diterpenos de Tipo Kaurano/farmacología , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Glucósidos/farmacología , Cirrosis Hepática/tratamiento farmacológico , Proteínas Smad/metabolismo , Actinas/metabolismo , Animales , Línea Celular , Colágeno Tipo I/metabolismo , Colagenasas , Nucleótidos de Desoxicitosina , Regulación hacia Abajo , Fibrosis/inducido químicamente , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Linfocinas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Tioacetamida/toxicidad , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba
9.
Expert Opin Biol Ther ; 18(sup1): 177-184, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30063851

RESUMEN

OBJECTIVES: Hepatic stellate cells (HSC) trans-differentiation is central to the development of liver fibrosis, marked by the expression of pro-fibrogenic genes and the proliferation and migration of activated HSC. Therefore, preventing and/or reverting the activation, proliferation, and migration of HSC may lead to new therapies for treating fibrosis/cirrhosis. Thymosin ß4 (Tß4) inhibits PDGF-BB-induced fibrogenesis, proliferation and migration of HSC by blocking Akt phosphorylation. Here, we utilized Tß4-derived peptides: amino-terminal-Ac-SDKPDMAEIEKFDKS (1-15aa) and actin-binding-LKKTETQ (17-23aa) to investigate the molecular mechanisms in the anti-fibrogenic actions of Tß4. METHODS: We used RT-PCR, Western blot, and proliferation and migration assays in early passages of human HSC cultures treated with PDGF-BB and/or Tß4 peptides. RESULTS: We showed that 17-23aa but not 1-15aa inhibited PDGF-BB-dependent up-regulation of PDGFß receptor, α-SMA, and collagen 1. It also blunted the phosphorylation of Akt at T 308 and S473, resulting in the inhibition of phosphorylation of PRAS40, and HSC proliferation and migration. Interestingly, 1-15aa blocked Akt phosphorylation at S473, but not T308 by inhibiting mTOR phosphorylation, thus, it did not have any effect on HSC proliferation and migration. CONCLUSION: These findings suggest that while 1-15aa has a minor effect on Akt phosphorylation, the anti-fibrogenic actions of Tß4 are exerted via 17-23aa.


Asunto(s)
Actinas/metabolismo , Becaplermina/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Timosina/farmacología , Animales , Células Cultivadas , Células Estrelladas Hepáticas/fisiología , Humanos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/prevención & control , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas/fisiología , Timosina/química , Timosina/metabolismo
10.
Oxid Med Cell Longev ; 2018: 9630175, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30116499

RESUMEN

Thymosin beta 4 (Tß4), an actin-sequestering protein, is involved in tissue development and regeneration. It prevents inflammation and fibrosis in several tissues. We investigated the role of Tß4 in chronic ethanol- and acute lipopolysaccharide- (LPS-) induced mouse liver injury. C57BL/6 mice were fed 5% ethanol in liquid diet for 4 weeks plus binge ethanol (5 g/kg, gavage) with or without LPS (2 mg/kg, intraperitoneal) for 6 hours. Tß4 (1 mg/kg, intraperitoneal) was administered for 1 week. We demonstrated that Tß4 prevented ethanol- and LPS-mediated increase in liver injury markers as well as changes in liver pathology. It also prevented ethanol- and LPS-mediated increase in oxidative stress by decreasing ROS and lipid peroxidation and increasing the antioxidants, reduced glutathione and manganese-dependent superoxide dismutase. It also prevented the activation of nuclear factor kappa B by blocking the phosphorylation of the inhibitory protein, IκB, thereby prevented proinflammatory cytokine production. Moreover, Tß4 prevented fibrogenesis by suppressing the epigenetic repressor, methyl-CpG-binding protein 2, that coordinately reversed the expression of peroxisome proliferator-activated receptor-γ and downregulated fibrogenic genes, platelet-derived growth factor-ß receptor, α-smooth muscle actin, collagen 1, and fibronectin, resulting in reduced fibrosis. Our data suggest that Tß4 has antioxidant, anti-inflammatory, and antifibrotic potential during alcoholic liver injury.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Etanol/efectos adversos , Fibrosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Lipopolisacáridos/efectos adversos , Proteínas de Microfilamentos/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Timosina/uso terapéutico , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/farmacología , Timosina/farmacología
11.
Biochem Pharmacol ; 156: 135-146, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30125555

RESUMEN

Nucleoredoxin (NXN) is a redox-regulating protein potentially targeted by reactive oxygen species (ROS). It regulates molecular pathways that participate in several key cellular processes. However, the role of NXN in the alcohol liver disease (ALD) redox regulation has not been fully understood. Here, we investigated the effects of ethanol and ethanol plus lipopolysaccharide, a two-hit liver injury model (Ethanol/LPS), on NXN/dishevelled (DVL) interaction and on DVL-dependent phosphoinositides production both in mouse liver and in a co-culture system consisting of human hepatic stellate cells (HSC) and ethanol metabolizing-VL17A human hepatocyte cells. Ethanol and two-hit model increased Nxn protein and mRNA expression, and 4-hydroxynonenal adducts. Two-hit model promoted Nxn nuclear translocation and Dvl/Phosphatidylinositol 4-kinase type-IIα (Pi4k2a) interaction ratio but surprisingly decreased Dvl protein and mRNA levels and reverted ethanol-induced Nxn/Dvl and Dvl/frizzled (Fzd) interaction ratios. Ethanol resulted in a significant increase of Dvl protein and mRNA expression, and decreased Nxn/Dvl interaction ratio but promoted the interaction of Dvl with Fzd and Pi4k2a; formation of this complex induced phosphatidylinositol 4-phosphate [PI(4)P] production. Ethanol and LPS treatments provoked similar alterations on NXN/DVL interaction and its downstream effect in HSC/VL17A co-culture system. Interestingly, ROS and glutathione levels as well as most of ethanol-induced alterations were modified by NXN overexpression in the co-culture system. In conclusion, two-hit model of ethanol exposure disrupts NXN/DVL homeostatic status to allow DVL/FZD/PI4K2A complex formation and stimulates PI(4)P production. These results provide a new mechanism showing that NXN also participates in the regulation of phosphoinositides production that is altered by ethanol during alcoholic liver disease progression.


Asunto(s)
Proteínas Dishevelled/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Nucleares/metabolismo , Oxidorreductasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Línea Celular , Técnicas de Cocultivo , Proteínas Dishevelled/genética , Etanol , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Oxidorreductasas/genética
13.
Oxid Med Cell Longev ; 2016: 5017460, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26881029

RESUMEN

Curcumin, an antioxidant compound found in Asian spices, was evaluated for its protective effects against ethanol-induced hepatosteatosis, liver injury, antiatherogenic markers, and antioxidant status in rats fed with Lieber-deCarli low menhaden (2.7% of total calories from ω-3 polyunsaturated fatty acids (PUFA)) and Lieber-deCarli high menhaden (13.8% of total calories from ω-3 PUFA) alcohol-liquid (5%) diets supplemented with or without curcumin (150 mg/kg/day) for 8 weeks. Treatment with curcumin protected against high ω-3 PUFA and ethanol-induced hepatosteatosis and increase in liver injury markers, alanine aminotransferase, and aspartate aminotransferase. Curcumin upregulated paraoxonase 1 (PON1) mRNA and caused significant increase in serum PON1 and homocysteine thiolactonase activities as compared to high ω-3 PUFA and ethanol group. Moreover, treatment with curcumin protected against ethanol-induced oxidative stress by increasing the antioxidant glutathione and decreasing the lipid peroxidation adduct 4-hydroxynonenal. These results strongly suggest that chronic ethanol in combination with high ω-3 PUFA exacerbated hepatosteatosis and liver injury and adversely decreases antiatherogenic markers due to increased oxidative stress and depletion of glutathione. Curcumin supplementation significantly prevented these deleterious actions of chronic ethanol and high ω-3 PUFA. Therefore, we conclude that curcumin may have therapeutic potential to protect against chronic alcohol-induced liver injury and atherosclerosis.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Curcumina/química , Dieta , Etanol/efectos adversos , Estrés Oxidativo , Aldehídos/química , Animales , Antioxidantes/química , Arildialquilfosfatasa/metabolismo , Aterosclerosis , Biomarcadores/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Ácidos Grasos Omega-3/química , Hígado Graso/tratamiento farmacológico , Hígado Graso/patología , Femenino , Glutatión/química , Metabolismo de los Lípidos , Peroxidación de Lípido/efectos de los fármacos , Ratas , Ratas Wistar
14.
Oxid Med Cell Longev ; 2016: 1840513, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28074114

RESUMEN

Chronic ethanol-induced downregulation of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) and upregulation of peroxisome proliferator-activated receptor gamma coactivator 1-beta (PGC1ß) affect hepatic lipid oxidation and lipogenesis, respectively, leading to fatty liver injury. Low-ω3 fatty acid (Low-ω3FA) that primarily regulates PGC1α and soy protein (SP) that seems to have its major regulatory effect on PGC1ß were evaluated for their protective effects against ethanol-induced hepatosteatosis in rats fed with Lieber-deCarli control or ethanol liquid diets with high or low ω3FA fish oil and soy protein. Low-ω3FA and SP opposed the actions of chronic ethanol by reducing serum and liver lipids with concomitant decreased fatty liver. They also prevented the downregulation of hepatic Sirtuin 1 (SIRT1) and PGC1α and their target fatty acid oxidation pathway genes and attenuated the upregulation of hepatic PGC1ß and sterol regulatory element-binding protein 1c (SREBP1c) and their target lipogenic pathway genes via the phosphorylation of 5' adenosine monophosphate-activated protein kinase (AMPK). Thus, these two novel modulators attenuate ethanol-induced hepatosteatosis and consequent liver injury potentially by regulating the two opposing lipid oxidation and lipogenic pathways.


Asunto(s)
Ácidos Grasos Omega-3/farmacología , Hígado Graso Alcohólico/patología , Peroxidación de Lípido/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas de Soja/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Etanol/toxicidad , Ácidos Grasos Omega-3/uso terapéutico , Hígado Graso Alcohólico/prevención & control , Femenino , Lípidos/análisis , Lípidos/sangre , Lipoproteínas HDL/sangre , Hígado/metabolismo , Coactivadores de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Ratas , Ratas Wistar , Sirtuina 1/genética , Sirtuina 1/metabolismo , Proteínas de Soja/uso terapéutico , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
15.
Clin Res Hepatol Gastroenterol ; 39 Suppl 1: S29-34, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26189985

RESUMEN

Fatty liver (hepatosteatosis) is the earliest abnormality in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD) due either to metabolic risk factors associated with insulin resistance and/or metabolic syndrome in the absence of alcohol consumption or to chronic alcohol abuse. When unchecked, both NAFLD and AFLD lead to steatohepatitis, fibrosis, cirrhosis, hepatocellular carcinoma (HCC) and eventual death. A number of common mechanisms contribute to the above various stages of hepatocyte injury, including lipotoxicity, endotoxin release, oxidative and ER stress leading to increased pro-inflammatory cytokines that stimulate hepatic fibrogenesis and cirrhosis by activating the quiescent hepatic stellate cells (HSC) into myofibroblasts. Significantly, patients with either NAFLD or AFLD respond favorably to early treatment modalities to reduce hepatic fat accumulation and consequent increased inflammatory signalling and activation of hepatic stellate cells. Although the pathogenic pathways associated with NAFLD and AFLD are seemingly similar, differentiation of the molecular mechanism/s of the pathogenesis of these liver diseases is critical in identifying the unique molecular signatures, especially in the early diagnosis of NAFLD and AFLD. Current clinical practice requires the invasive biopsy procedure for the conclusive diagnosis of NAFLD and AFLD. Micro RNAs (miRNAs) are ∼22 nucleotide non-coding sequences that bind to the 3'-untranslated region of target transcripts and regulate gene expression by degradation of target mRNAs or inhibition of translation. Emerging studies may prove to establish miRNAs as excellent non-invasive tools for the early diagnosis of various stages of liver diseases.


Asunto(s)
Hígado Graso/metabolismo , Biomarcadores/metabolismo , Citocinas/metabolismo , Humanos , Resistencia a la Insulina , Metabolismo de los Lípidos , MicroARNs/metabolismo , Estrés Oxidativo
16.
Am J Pathol ; 184(7): 2035-44, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24814604

RESUMEN

Because scavenger receptor class B type 1 is the cholesterol uptake liver receptor, whereas peroxisome proliferator-activated receptor γ coactivator-1ß (PGC-1ß) and PGC-1α are critical for lipid synthesis and degradation, we investigated the roles of these signaling molecules in the actions of ethanol-polyunsaturated fatty acids and betaine on hepatosteatosis and steatohepatitis. Ethanol-polyunsaturated fatty acid treatment caused the following: i) hepatosteatosis, as evidenced by increased liver cholesterol and triglycerides, lipid score, and decreased serum adiponectin; ii) marked inhibition of scavenger receptor class B type 1 glycosylation, its plasma membrane localization, and its hepatic cholesterol uptake function; and iii) moderate steatohepatitis, as evidenced by histopathological characteristics, increased liver tumor necrosis factor α and IL-6, decreased glutathione, and elevated serum alanine aminotransferase. These actions of ethanol involved up-regulated PGC-1ß, sterol regulatory element-binding proteins 1c and 2, acetyl-CoA carboxylase, and HMG-CoA reductase mRNAs/proteins and inactive non-phosphorylated AMP kinase; and down-regulated silence regulator gene 1 and PGC-1α mRNA/proteins and hepatic fatty acid oxidation. Betaine markedly blunted all these actions of ethanol on hepatosteatosis and steatohepatitis. Therefore, we conclude that ethanol-mediated impaired post-translational modification, trafficking, and function of scavenger receptor class B type 1 may account for alcoholic hyperlipidemia. Up-regulation of PGC-1ß and lipid synthetic genes and down-regulation of silence regulator gene 1, PGC-1α, adiponectin, and lipid degradation genes account for alcoholic hepatosteatosis. Induction of proinflammatory cytokines and depletion of endogenous antioxidant, glutathione, account for alcoholic steatohepatitis. We suggest betaine as a potential therapeutic agent because it effectively protects against adverse actions of ethanol.


Asunto(s)
Betaína/farmacología , Hígado Graso Alcohólico/metabolismo , Hígado Graso/metabolismo , Receptores Depuradores de Clase B/metabolismo , Factores de Transcripción/metabolismo , Adiponectina/sangre , Alanina Transaminasa/sangre , Animales , Dieta Alta en Grasa , Etanol/efectos adversos , Hígado Graso/patología , Hígado Graso Alcohólico/patología , Femenino , Interleucina-6/metabolismo , Hígado/metabolismo , Hígado/fisiopatología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Procesamiento Proteico-Postraduccional , Ratas Wistar , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
17.
Am J Pathol ; 184(5): 1458-67, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24641900

RESUMEN

Alcohol-induced liver fibrosis and eventually cirrhosis is a leading cause of death. Acetaldehyde, the first metabolite of ethanol, up-regulates expression of the human α2(I) collagen gene (COL1A2). Early acetaldehyde-mediated effects involve phosphorylation and nuclear translocation of SMAD3/4-containing complexes that bind to COL1A2 promoter to induce fibrogenesis. We used human and mouse hepatic stellate cells to elucidate the mechanisms whereby acetaldehyde up-regulates COL1A2 by modulating the role of Ski and the expression of SMADs 3, 4, and 7. Acetaldehyde induced up-regulation of COL1A2 by 3.5-fold, with concomitant increases in the mRNA (threefold) and protein (4.2- and 3.5-fold) levels of SMAD3 and SMAD4, respectively. It also caused a 60% decrease in SMAD7 expression. Ski, a member of the Ski/Sno oncogene family, is colocalized in the nucleus with SMAD4. Acetaldehyde induces translocation of Ski and SMAD4 to the cytoplasm, where Ski undergoes proteasomal degradation, as confirmed by the ability of the proteasomal inhibitor lactacystin to blunt up-regulation of acetaldehyde-dependent COL1A2, but not of the nonspecific fibronectin gene (FN1). We conclude that acetaldehyde up-regulates COL1A2 by enhancing expression of the transactivators SMAD3 and SMAD4 while inhibiting the repressor SMAD7, along with promoting Ski translocation from the nucleus to cytoplasm. We speculate that drugs that prevent proteasomal degradation of repressors targeting COL1A2 may have antifibrogenic properties.


Asunto(s)
Acetaldehído/farmacología , Colágeno Tipo II/genética , Proteínas de Unión al ADN/metabolismo , Células Estrelladas Hepáticas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Smad/metabolismo , Regulación hacia Arriba/genética , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Colágeno Tipo II/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Fibronectinas/genética , Fibronectinas/metabolismo , Genes Reporteros , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Proteínas Smad/genética , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína smad7/genética , Proteína smad7/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba/efectos de los fármacos
18.
Hepatol Int ; 8 Suppl 2: 413-20, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26201319

RESUMEN

Alcoholic steatosis, instead of being innocuous, plays a critical role in liver inflammation and fibrogenesis. The severity of fatty liver is governed by the concerted balance between lipid transport, synthesis, and degradation. Whereas scavenger receptor class B, type I (SR-B1) is critical for reverse cholesterol uptake by the liver, peroxisome proliferator-activated receptor-gamma (PPARγ) coactivator-1α and -ß (PGC1α and PGC1ß) are critical for lipid degradation and synthesis, respectively. Because betaine is a lipotropic agent, we have evaluated its effects on alcoholic steatosis. Betaine effectively prevented chronic alcohol-mediated (i) impaired SR-B1 glycosylation, plasma membrane localization, and consequent impaired cholesterol transport; and (ii) up regulation of PGC-1ß, sterol regulatory element-binding protein 1c and downstream lipogenic genes with concomitant increased liver cholesterol, triglycerides and hepatic lipid score. Similarly, because of its anti-inflammatory and anti-fibrotic effects in other organs, we evaluated the protective effects of thymosin ß4 (Tß4) against carbon tetrachloride (CCl4)-induced hepatotoxicity in rat. Tß4 prevented CCl4-induced (i) necrosis, inflammatory infiltration and up-regulation of α1(2)collagen, alpha-smooth muscle actin (α-SMA), platelet derived growth factor beta (PDGF-ß) receptor and fibronectin mRNA expression; (ii) down-regulation of adipogenic gene, PPARγ and the up-regulation of epigenetic repressor gene, methyl CpG binding protein 2 (MeCP2) mRNA levels, suggesting that the anti-fibrogenic actions of Tß4 involve the prevention of trans-differentiation of quiescent hepatic stellate cells into myo-fibroblasts largely by up-regulating PPARγ and by down-regulating MeCP2 genes. We therefore conclude that betaine and Tß4 can effectively protect against alcoholic hepatosteatosis and hepatic fibrogenesis, respectively.

19.
Alcohol Clin Exp Res ; 37(11): 1838-48, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23895226

RESUMEN

BACKGROUND: Transforming growth factor beta 1 (TGF-ß1) is a pleiotropic cytokine that activates hepatic stellate cell (HSC) proliferation, but inhibits parenchymal cell proliferation. Therefore, we hypothesize that TGF-ß1 regulates HSC proliferation and elucidated its molecular action. METHODS: In order to elucidate the molecular mechanism whereby TGF-ß1 up-regulates platelet derived growth factor beta (PDGF-ß) receptor mRNA and induces a delayed proliferation of HSC, we used proliferation and apoptosis assays as well as RT-PCR, Western blot analysis, immunostaining, and flow cytometry in mouse and rat HSC. RESULTS: We show that TGF-ß1 markedly induces the proliferation of mouse HSC in culture with concomitant 2.1-fold (p < 0.001) stimulation in [(3) H]-thymidine incorporation into cellular DNA. This induction is maximal between 24 and 36 hours postcytokine exposure that is triggered by 7.6-fold (p < 0.001) up-regulation of PDGF-ß receptor mRNA and associated increase in PDGF-ß receptor protein after 48 hours. TGF-ß1-dependent HSC proliferation is mimicked by H2 O2 that is inhibited by catalase, implying that TGF-ß1 action is mediated via reactive oxygen species. HSC proliferation is blunted by PDGF-ß receptor-neutralizing antibody as well as by specific inhibitors of PI3 kinase (PI3K), AKT, and p70(S6K) , indicating that the action of TGF-ß1 involves the activation of PDGF-ß receptor via the PI3K/AKT/p70(S6K) signaling pathway. TGF-ß1 also induces a reorganization of actin and myosin filaments and cell morphology leading to the formation of palisades although their myosin and actin contents remained constant. These findings suggest that TGF-ß1-mediated oxidative stress causes the transdifferentiation of HSC and primes them for extracellular matrix (ECM) deposition and scar contraction. CONCLUSIONS: We conclude that liver injury up-regulates TGF-ß1 that inhibits parenchymal cell proliferation, but stimulates HSC proliferation leading to the production of ECM and type I collagen resulting in fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Becaplermina , Proliferación Celular , Tamaño de la Célula , Células Cultivadas , Células Estrelladas Hepáticas/citología , Peróxido de Hidrógeno/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-sis/metabolismo , Ratas , Transducción de Señal
20.
Free Radic Biol Med ; 65: 1487-1496, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23880292

RESUMEN

We investigated whether the fibrogenic actions of acetaldehyde, the immediate oxidation product of ethanol, are mediated via Wingless (WNT) and/or ß-catenin pathways in human hepatic stellate cells (HSC). First, we show that both ß-catenin small inhibitory RNA and a dominant negative-MYC expression vector markedly down-regulated the expressions of fibrogenic genes in freshly isolated HSC. We further show that acetaldehyde up-regulated platelet-derived growth factor receptor beta mRNA and protein expressions ranging from 4.0- to 7.2-fold (P<0.001). Acetaldehyde induced MYC and collagen type-1 alpha-2 mRNA and protein expressions were WNT independent because DKK1, an antagonist of the canonical WNT/ß-catenin pathway, completely failed to block these inductions. Acetaldehyde increased phospho-glycogen synthase kinase-3 beta (GSK3B) protein by 31% (P<0.01), whereas phospho-ß-catenin protein decreased by 50% (P ≤ 0.01). Significantly, in contrast to 43% (P<0.01) inhibition of ß-catenin nuclear translocation in nucleoredoxin (NXN)-overexpressed HSC, acetaldehyde profoundly stimulated ß-catenin nuclear translocation by 51%, (P<0.01). Acetaldehyde also increased the cellular reactive oxygen species level 2-fold (P<0.001) with a concomitant 2-fold (P<0.001) increase in 4-hydroxynonenal adducts. Conversely, there was a 44% decrease (P<0.001) in glutathione levels with a concomitant 76% (P<0.001) decrease in the level of NXN/ disheveled (DVL) complex. Based on these findings, we conclude that actions of acetaldehyde are mediated by a mechanism that inactivates NXN by releasing DVL, leading to the inactivation of GSK3B, and thereby blocks ß-catenin phosphorylation and degradation. Thus, the stabilized ß-catenin translocates to the nucleus where it up-regulates the fibrogenic pathway genes. This novel mechanism of action of acetaldehyde has the potential for therapeutic interventions in liver fibrosis induced by alcohol.


Asunto(s)
Acetaldehído/farmacología , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática Alcohólica/patología , Proteínas Nucleares/antagonistas & inhibidores , Oxidorreductasas/antagonistas & inhibidores , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Aldehídos/metabolismo , Células Cultivadas , Colágeno Tipo I/genética , Proteínas Dishevelled , Etanol/química , Etanol/metabolismo , Genes myc/genética , Glutatión/biosíntesis , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/biosíntesis , Glucógeno Sintasa Quinasa 3 beta , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteínas Nucleares/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Oxidorreductasas/biosíntesis , Fosfoproteínas/biosíntesis , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Especies Reactivas de Oxígeno/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Wnt/metabolismo , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA