Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cancers (Basel) ; 13(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638423

RESUMEN

Maraviroc (MVC), a CCR5 antagonist, reduces liver fibrosis, injury and tumour burden in mice fed a hepatocarcinogenic diet, suggesting it has potential as a cancer therapeutic. We investigated the effect of MVC on liver progenitor cells (LPCs) and macrophages as both have a role in hepatocarcinogenesis. Mice were fed the hepatocarcinogenic choline-deficient, ethionine-supplemented diet (CDE) ± MVC, and immunohistochemistry, RNA and protein expression were used to determine LPC and macrophage abundance, migration and related molecular mechanisms. MVC reduced LPC numbers in CDE mice by 54%, with a smaller reduction seen in macrophages. Transcript and protein abundance of LPC-associated markers correlated with this reduction. The CDE diet activated phosphorylation of AKT and STAT3 and was inhibited by MVC. LPCs did not express Ccr5 in our model; in contrast, macrophages expressed high levels of this receptor, suggesting the effect of MVC is mediated by targeting macrophages. MVC reduced CD45+ cells and macrophage migration in liver and blocked the CDE-induced transition of liver macrophages from an M1- to M2-tumour-associated macrophage (TAM) phenotype. These findings suggest MVC has potential as a re-purposed therapeutic agent for treating chronic liver diseases where M2-TAM and LPC numbers are increased, and the incidence of HCC is enhanced.

2.
Dis Model Mech ; 8(12): 1635-41, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26496771

RESUMEN

The choline-deficient, ethionine-supplemented (CDE) dietary model induces chronic liver damage, and stimulates liver progenitor cell (LPC)-mediated repair. Long-term CDE administration leads to hepatocellular carcinoma in rodents and lineage-tracing studies show that LPCs differentiate into functional hepatocytes in this model. The CDE diet was first modified for mice by our laboratory by separately administering choline-deficient chow and ethionine in the drinking water (CD+E diet). Although this CD+E diet is widely used, concerns with variability in weight loss, morbidity, mortality and LPC response have been raised by researchers who have adopted this model. We propose that these inconsistencies are due to differential consumption of chow and ethionine in the drinking water, and that incorporating ethionine in the choline-deficient chow, and altering the strength, will achieve better outcomes. Therefore, C57Bl/6 mice, 5 and 6 weeks of age, were fed an all-inclusive CDE diet of various strengths (67% to 100%) for 3 weeks. The LPC response was quantitated and cell lines were derived. We found that animal survival, LPC response and liver damage are correlated with CDE diet strength. The 67% and 75% CDE diet administered to mice older than 5 weeks and greater than 18 g provides a consistent and acceptable level of animal welfare and induces a substantial LPC response, permitting their isolation and establishment of cell lines. This study shows that an all-inclusive CDE diet for mice reproducibly induces an LPC response conducive to in vivo studies and isolation, whilst minimizing morbidity and mortality.


Asunto(s)
Colina/farmacología , Dieta , Etionina/farmacología , Hígado/citología , Morbilidad , Células Madre/citología , Envejecimiento , Alanina Transaminasa/sangre , Animales , Conductos Biliares/citología , Biomarcadores/metabolismo , Peso Corporal , Diferenciación Celular , Línea Celular , Linaje de la Célula , Hepatocitos/citología , Inflamación/patología , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Análisis de Supervivencia
3.
Hepatology ; 62(4): 1272-84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26173184

RESUMEN

UNLABELLED: Liver progenitor cells (LPCs) are necessary for repair in chronic liver disease because the remaining hepatocytes cannot replicate. However, LPC numbers also correlate with disease severity and hepatocellular carcinoma risk. Thus, the progenitor cell response in diseased liver may be regulated to optimize liver regeneration and minimize the likelihood of tumorigenesis. How this is achieved is currently unknown. Human and mouse diseased liver contain two subpopulations of macrophages with different ontogenetic origins: prenatal yolk sac-derived Kupffer cells and peripheral blood monocyte-derived macrophages. We examined the individual role(s) of Kupffer cells and monocyte-derived macrophages in the induction of LPC proliferation using clodronate liposome deletion of Kupffer cells and adoptive transfer of monocytes, respectively, in the choline-deficient, ethionine-supplemented diet model of liver injury and regeneration. Clodronate liposome treatment reduced initial liver monocyte numbers together with the induction of injury and LPC proliferation. Adoptive transfer of monocytes increased the induction of liver injury, LPC proliferation, and tumor necrosis factor-α production. CONCLUSION: Kupffer cells control the initial accumulation of monocyte-derived macrophages. These infiltrating monocytes are in turn responsible for the induction of liver injury, the increase in tumor necrosis factor-α, and the subsequent proliferation of LPCs.


Asunto(s)
Comunicación Celular/fisiología , Macrófagos del Hígado/fisiología , Regeneración Hepática/fisiología , Hígado/citología , Monocitos/fisiología , Células Madre/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Hepatology ; 52(1): 291-302, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20578156

RESUMEN

UNLABELLED: Liver progenitor cells (LPCs) represent the cell compartment facilitating hepatic regeneration during chronic injury while hepatocyte-mediated repair mechanisms are compromised. LPC proliferation is frequently observed in human chronic liver diseases such as hereditary hemochromatosis, fatty liver disease, and chronic hepatitis. In vivo studies have suggested that a tumor necrosis factor family member, tumor necrosis factor-like weak inducer of apoptosis (TWEAK), is promitotic for LPCs; whether it acts directly is not known. In our murine choline-deficient, ethionine-supplemented (CDE) model of chronic liver injury, TWEAK receptor [fibroblast growth factor-inducible 14 (Fn14)] expression in the whole liver is massively upregulated. We therefore set out to investigate whether TWEAK/Fn14 signaling promotes the regenerative response in CDE-induced chronic liver injury by mitotic stimulation of LPCs. Fn14 knockout (KO) mice showed significantly reduced LPC numbers and attenuated inflammation and cytokine production after 2 weeks of CDE feeding. The close association between LPC proliferation and activation of hepatic stellate cells in chronic liver injury prompted us to investigate whether fibrogenesis was also modulated in Fn14 KO animals. Collagen deposition and expression of key fibrogenesis mediators were reduced after 2 weeks of injury, and this correlated with LPC numbers. Furthermore, the injection of 2-week-CDE-treated wildtype animals with TWEAK led to increased proliferation of nonparenchymal pan cytokeratin-positive cells. Stimulation of an Fn14-positive LPC line with TWEAK led to nuclear factor kappa light chain enhancer of activated B cells (NFkappaB) activation and dose-dependent proliferation, which was diminished after targeting of the p50 NFkappaB subunit by RNA interference. CONCLUSION: TWEAK acts directly and stimulates LPC mitosis in an Fn14-dependent and NFkappaB-dependent fashion, and signaling via this pathway mediates the LPC response to CDE-induced injury and regeneration.


Asunto(s)
Regeneración Hepática , Mitosis , Células Madre/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Proliferación Celular , Deficiencia de Colina/metabolismo , Colágeno/metabolismo , Citocina TWEAK , Etionina/farmacología , Hígado/efectos de los fármacos , Hígado/lesiones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitógenos/farmacología , FN-kappa B/agonistas , FN-kappa B/genética , FN-kappa B/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Proteínas Recombinantes/farmacología , Células Madre/efectos de los fármacos , Receptor de TWEAK , Factores de Necrosis Tumoral/farmacología
5.
Carcinogenesis ; 29(6): 1124-33, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18413365

RESUMEN

A hallmark of cancer cells is their ability to evade apoptosis and mitochondria play a critical role in this process. Delineating mitochondrial differences between normal and cancer cells has proven challenging due to the lack of matched cell lines. Here, we compare two matched liver progenitor cell (LPC) lines, one non-tumorigenic [p53-immortalized liver (PIL) 4] and the other tumorigenic (PIL2). Analysis of these cell lines and a p53 wild-type non-tumorigenic cell line [bipotential murine oval liver (BMOL)] revealed an increase in expression of genes encoding the antiapoptotic proteins cellular inhibitor of apoptosis protein (cIAP) 1 and yes associate protein in the PIL2 cells, which resulted in an increase in the protein encoded by these genes. PIL2 cells have higher mitochondrial membrane potential (Deltapsi(m)) compared with PIL4 and BMOL and had greater levels of reactive oxygen species, despite the fact that the mitochondrial antioxidant enzyme, manganese superoxide disumutase, was elevated at transcript and protein levels. Taken together, these results may account for the observed resistance of PIL2 cells to apoptotic stimuli compared with PIL4. We tested a new gold compound to show that hyperpolarized Deltapsi(m) led to its increased accumulation in mitochondria of PIL2 cells. This compound selectively induces apoptosis in PIL2 cells but not in PIL4 or BMOL. The gold compound depolarized the Deltapsi(m), depleted the adenosine triphosphate pool and activated caspase-3 and caspase-9, suggesting that apoptosis was mediated via mitochondria. This investigation shows that the non-tumorigenic and tumorigenic LPCs are useful models to delineate the role of mitochondrial dysfunction in tumorigenesis and for the future development of mitochondria-targeted chemotherapeutics that selectively target tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Metabolismo Energético , Compuestos de Oro/farmacología , Hepatocitos/metabolismo , Mitocondrias/efectos de los fármacos , Células Madre/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Hepatocitos/citología , Immunoblotting , Proteínas Inhibidoras de la Apoptosis/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Análisis por Micromatrices , Mitocondrias/metabolismo , Fosfoproteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos , Proteínas Señalizadoras YAP
6.
Stem Cell Res ; 1(3): 195-204, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19383400

RESUMEN

Liver progenitor (oval) cells have enormous potential in the treatment of patients with liver disease using a cell therapy approach, but their use is limited by their scarcity and the number of donor livers from which they can be derived. Bone marrow may be a suitable source. Previously the derivation of oval cells from bone marrow was examined in rodents using hepatotoxins and partial hepatectomy to create liver damage. These protocols induce oval cell proliferation; however, they do not produce the disease conditions that occur in humans. In this study we have used the choline-deficient, ethionine-supplemented (CDE) diet (which causes fatty liver) and viral hepatitis as models of chronic injury to evaluate the contribution of bone marrow cells to oval cells under conditions that closely mimic human liver disease pathophysiology. Following transplantation of lacZ-transgenic bone marrow cells into congenic mice, liver injury was induced and the movement of bone marrow cells to the liver monitored. Bone marrow-derived oval cells were observed in response to the CDE diet and viral injury but represented a minor fraction (0-1.6%) of the oval cell compartment, regardless of injury severity. In all situations only rare, individual bone marrow-derived oval cells were observed. We hypothesized that the bone marrow cells may replenish oval cells that are expended by protracted liver injury and regeneration; however, experiments involving a subsequent episode of chronic liver injury failed to induce proliferation of the bone marrow-derived oval cells that appeared as a result of the first episode. Bone marrow-derived hepatocytes were also observed in all injury models and controls at a frequency unrelated to that of oval cells. We conclude that during viral-and steatosis-induced liver disease the contribution of bone marrow cells to hepatocytes, either via oval cells or by independent mechanisms, is minimal and that the majority of oval cells responding to this injury are sourced from the liver.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Regeneración Hepática , Animales , Células de la Médula Ósea/citología , Deficiencia de Colina , Dieta , Modelos Animales de Enfermedad , Etionina/administración & dosificación , Etionina/efectos adversos , Hígado/citología , Hepatopatías/etiología , Ratones , Ratones Congénicos , Resultado del Tratamiento
7.
J Hepatol ; 47(6): 826-33, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17923165

RESUMEN

BACKGROUND/AIMS: Several previous studies have suggested that interferon gamma (IFNgamma) may play a key role during hepatic progenitor cell (HPC) mediated liver regeneration. However to date, no studies have directly tested the ability of IFNgamma to mediate the HPC response in an in vivo model. METHODS/RESULTS: Administration of IFNgamma to mice receiving a choline deficient, ethionine (CDE) supplemented diet to induce chronic injury resulted in an augmented HPC response. This was accompanied by increased inflammation, altered cytokine expression and hepatic fibrosis. Serum alanine aminotransferase activity, hepatocyte apoptosis and Bak staining were significantly increased in IFNgamma-treated, CDE-fed mice, demonstrating that liver damage was exacerbated in these animals. Administration of IFNgamma to control diet fed mice did not induce liver damage, however it did stimulate hepatic inflammation. CONCLUSIONS: Our results suggest that IFNgamma increases the HPC response to injury by stimulating hepatic inflammation and aggravating liver damage. This is accompanied by an increase in hepatic fibrogenesis, supporting previous reports which suggest that the HPC response may drive fibrogenesis during chronic liver injury.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Interferón gamma/efectos adversos , Cirrosis Hepática/inducido químicamente , Células Madre/efectos de los fármacos , Animales , Enfermedad Crónica , Dieta , Modelos Animales de Enfermedad , Fibrinógeno/biosíntesis , Hepatocitos/citología , Inflamación/inducido químicamente , Interferón gamma/administración & dosificación , Ratones , Proteínas Recombinantes
8.
Int J Biochem Cell Biol ; 39(12): 2226-39, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17693121

RESUMEN

Oval cells have great potential for use in cell therapy to treat liver disease, however this cannot be achieved until the factors which govern their proliferation and differentiation are better understood. We describe a method to establish primary cultures of murine oval cells, and the derivation of two novel lines from these. Primary cultures from the livers of wildtype or TAT-GRE lacZ transgenic mice subjected to a choline-deficient, ethionine-supplemented diet comprised up to 80% oval cells at day 7 based on A6 or CK19 staining. Cell lines were clonally derived, which underwent spontaneous immortalisation following prolonged maintenance in culture. Immunostaining and RT-PCR demonstrated they express hepatocytic and biliary markers and they were therefore termed "bipotential murine oval liver" (BMOL) cells. Under proliferating culture conditions, BMOL or BMOL-TAT cells abundantly expressed oval cell and biliary markers, whereas mature hepatocytic markers were upregulated when the growth conditions were changed to facilitate differentiation. Hepatic differentiation of BMOL-TAT cells could be traced by measuring the expression of their lacZ transgene, which is driven by a promoter element from tyrosine aminotransferase (TAT), a marker of adult hepatocytes. Interestingly, haematopoietic markers were upregulated in superconfluent cultures, indicating a possible multipotentiality. None of the cell lines grew in semi-solid agar, nor did they form tumours in nude mice, suggesting they are non-tumourigenic. These novel murine oval cell lines, together with a reliable method for isolation and culture of primary oval cells, will provide a useful tool for investigating the contribution of oval cells to liver regeneration.


Asunto(s)
Etionina/administración & dosificación , Alimentos Formulados , Hígado/citología , Células Madre/citología , Animales , Antígenos de Diferenciación/análisis , Antígenos de Diferenciación/metabolismo , Cadherinas/análisis , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Línea Celular Transformada , Separación Celular/métodos , Colina/administración & dosificación , Conexinas/genética , Factor de Transcripción GATA2/genética , Expresión Génica/efectos de los fármacos , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Inmunohistoquímica , Queratina-19/análisis , Queratina-19/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Antígenos Thy-1/genética , Tirosina Transaminasa/genética , Tirosina Transaminasa/metabolismo
9.
Hepatology ; 45(2): 486-94, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17256754

RESUMEN

UNLABELLED: Gp130-mediated IL-6 signaling may play a role in oval cell proliferation in vivo. Levels of IL-6 are elevated in livers of mice treated with a choline-deficient ethionine-supplemented (CDE) diet that induces oval cells, and there is a reduction of oval cells in IL-6 knockout mice. The CDE diet recapitulates characteristics of chronic liver injury in humans. In this study, we determined the impact of IL-6 signaling on oval cell-mediated liver regeneration in vivo. Signaling pathways downstream of gp130 activation were also dissected. Numbers of A6(+ve) liver progenitor oval cells (LPCs) in CDE-treated murine liver were detected by immunohistochemistry and quantified. Levels of oval cell migration and proliferation were compared in CDE-treated mouse strains that depict models of gp130-mediated hyperactive ERK-1/2 signaling (gp130(deltaSTAT)), hyperactive STAT-3 signaling (gp130(Y757F) and Socs-3(-/deltaAlb)) or active ERK-1/2 as well as active STAT-3 signaling (wild-type). The A6(+ve) LPC numbers were increased with IL-6 treatment in vivo. The gp130(Y757F) mice displayed increased A6(+ve) LPCs numbers compared with wild-type and gp130(deltaSTAT) mice. Numbers of A6(+ve) LPCs were also increased in the livers of CDE treated Socs-3(-/deltaAlb) mice compared with their control counterparts. Lastly, inhibition of ERK-1/2 activation in cultured oval cells increased hyper IL-6-induced cell growth. For the first time, we have dissected the gp130-mediated signaling pathways, which influence liver progenitor oval cell proliferation. CONCLUSION: Hyperactive STAT-3 signaling results in enhanced oval cell numbers, whereas ERK-1/2 activation suppresses oval cell proliferation.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular , Receptor gp130 de Citocinas/fisiología , Interleucina-6/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor de Transcripción STAT3/fisiología , Transducción de Señal/fisiología , Células Madre/fisiología , Animales , Línea Celular , Humanos , Hígado/citología , Hígado/metabolismo , Regeneración Hepática/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
10.
Hepatology ; 45(1): 31-41, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17187411

RESUMEN

UNLABELLED: Oval cells are hepatocytic precursors that proliferate in late-stage cirrhosis and that give rise to a subset of human hepatocellular carcinomas. Although liver regeneration typically occurs through replication of existing hepatocytes, oval cells proliferate only when hepatocyte proliferation is inhibited. Transforming growth factor-beta (TGF-beta) is a key inhibitory cytokine for hepatocytes, both in vitro and in vivo. Because TGF-beta levels are elevated in chronic liver injury when oval cells arise, we hypothesized that oval cells may be less responsive to the growth inhibitory effects of this cytokine. To examine TGF-beta signaling in vivo in oval cells, we analyzed livers of rats fed a choline-deficient, ethionine-supplemented (CDE) diet for phospho-Smad2. Phospho-Smad2 was detected in more than 80% of hepatocytes, but staining was substantially reduced in oval cells. Ki67 staining, in contrast, was significantly more common in oval cells than hepatocytes. To understand the inverse relationship between TGF-beta signaling and proliferation in oval cells and hepatocytes, we examined TGF-beta signaling in vitro. TGF-beta caused marked growth inhibition in primary hepatocytes and the AML12 hepatocyte cell line. Two oval cell lines, LE/2 and LE/6, were less responsive. The greater sensitivity of the hepatocytes to TGF-beta-induced growth inhibition may result from the absence of Smad6 in these cells. CONCLUSION: Our results indicate that oval cells, both in vivo and in vitro, are less sensitive to TGF-beta-induced growth inhibition than hepatocytes. These findings further suggest an underlying mechanism for the proliferation of oval cells in an environment inhibitory to hepatocytic proliferation.


Asunto(s)
Proliferación Celular , Hepatocitos/citología , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Apoptosis , Células Cultivadas , Ciclinas/genética , Ciclinas/metabolismo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Ratones , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Proteínas Smad/genética , Proteínas Smad/metabolismo , Células Madre/citología
11.
J Hepatol ; 46(1): 134-41, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17112626

RESUMEN

BACKGROUND/AIMS: Liver regeneration following chronic injury is associated with inflammation, the proliferation of liver progenitor (oval) cells and fibrosis. Previous studies identified interferon-gamma as a key mediator of oval cell proliferation. Interferon-gamma is known to regulate Th1 cell activities during immune challenge. Therefore, we hypothesised that progenitor cell-mediated regeneration is associated with a Th1 immune response. METHODS: C57Bl/6 (normal Th1 response) and BALB/c mice (deficient in Th1 signalling) were placed on a carcinogenic diet to induce liver injury, progenitor cell proliferation and fibrosis. RESULTS: Serum transaminases and mortality were elevated in BALB/c mice fed the diet. Proliferation of liver progenitor cells was significantly attenuated in BALB/c animals. The pattern of cytokine expression and inflammation differed between strains. Liver fibrosis and hepatic stellate cell activation were significantly inhibited in BALB/c mice compared to C57Bl/6. In addition, interferon-gamma knockout mice also showed reduced fibrosis compared to wild type. These findings are in contrast to published results, in which interferon-gamma is shown to be anti-fibrogenic. CONCLUSIONS: Our data demonstrate that the hepatic progenitor cell response to a CDE diet is inhibited in mice lacking Th1 immune signalling and further show that this inhibition is associated with reduced liver fibrosis.


Asunto(s)
Deficiencia de Colina/metabolismo , Etionina/administración & dosificación , Cirrosis Hepática/prevención & control , Animales , Apoptosis , Proliferación Celular , Deficiencia de Colina/patología , Citocinas/biosíntesis , Suplementos Dietéticos , Hepatocitos/metabolismo , Hepatocitos/patología , Interferón gamma/deficiencia , Interferón gamma/genética , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Regeneración Hepática/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de la Especie , Células Madre/metabolismo , Células Madre/patología , Células TH1/fisiología
12.
Hepatology ; 43(5): 1074-83, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16628647

RESUMEN

Hepatic progenitor cells (called oval cells in rodents) proliferate during chronic liver injury. They have been suggested as targets of malignant transformation in chronic liver diseases, including chronic hepatitis C. Interferon alpha therapy reduces the risk of hepatocellular carcinoma (HCC) in chronic hepatitis C regardless of viral clearance. The aim of this study was to determine whether interferon alpha could reduce the risk of HCC by modifying preneoplastic events in the hepatic progenitor cell population. Pre- and post-treatment liver biopsies were evaluated for changes in t he hepaticprogenitor cell population in 16 patients with non-responding chronic hepatitis C Interferon alpha-based treatment significantly reduced the numbers of c-kit-positive hepatic progenitor cells by 50%. To determine the mechanism of cell number reduction, the effects of interferon alpha on murinehepatic progenitor cells were studied in vitro. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) proliferation assay and proliferating cell nuclear antigen staining showed that interferon alpha had a dose-dependent, anti-proliferative effect Interferon alpha stimulated hepatocytic and biliary differentiation of the oval cell lines reflected by increased expression of albumin and cytokeratin19 accompanied by decreased expression of alphafetoprotein and Thy-1. To validatethese results in vivo, mice were placed on the choline-deficient, ethionine-supplemented diet to induce liver injury and oval cell proliferation and treated with pegylated interferon alpha 2b for 2 weeks. This resulted in a significant four-fold reduction in the number of oval cells (P < .05). In conclusion, interferon alpha-based treatment reduced the number of hepatic progenitor cells in chronic liver injury by modulating apoptosis, proliferation, and differentiation. Supplementay material for this article can


Asunto(s)
Antivirales/farmacología , Proliferación Celular/efectos de los fármacos , Interferón-alfa/farmacología , Hígado/citología , Hígado/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Células Cultivadas , Hepatitis C Crónica/tratamiento farmacológico , Humanos , Interferón alfa-2 , Interferón-alfa/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Polietilenglicoles , Proteínas Recombinantes
13.
Carcinogenesis ; 27(8): 1607-16, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16497703

RESUMEN

Oval cell proliferation precedes neoplasia in many rodent models of hepatocellular carcinoma and prevention of this proliferative response can reduce the risk of subsequent carcinoma. This study aimed to determine whether a selective cyclo-oxygenase-2 (COX-2) inhibitor, SC-236, affects (i) the oval cell response to liver injury in a mouse model of hepatocarcinogenesis and (ii) an oval cell line. Four-week-old mice were fed either normal chow or a choline deficient, ethionine supplemented (CDE) diet in the presence or absence of SC-236. Liver histology and oval cell numbers were determined after 2, 4, 12 and 52 weeks of treatment. Oval cells were scored using morphological criteria and positive immuno-staining for the M(2)-isozyme of pyruvate kinase (M2PK) or A6. An immortalized oval cell line (PIL-2) was used to study the in vitro effects of SC-236 on oval cell proliferation, apoptosis and Akt phosphorylation. The percentage of M2PK-positive oval cells and COX-2-positive cells was reduced by 80% and 45%, respectively, in CDE-fed mice receiving SC-236 compared with CDE-fed animals not receiving SC-236. Some M2PK-positive oval cells were also COX-2 positive. The percentage of A6-positive cells was not affected by SC-236 administration to CDE-fed mice. Administration of SC-236 increased apoptosis as evidenced by a 73% increase in the number of TUNEL-positive cells at 2 weeks in CDE-fed mice. Primary oval cells and PIL-2 cells expressed COX-2. In vitro treatment of PIL-2 cells with SC-236 resulted in a dose-dependent preferential death of A6-negative cells. Administration of 25 and 50 microM Prostaglandin E(2) partially attenuated SC-236 induced cell death by 25%. In vitro oval cell death was associated with apoptosis and a 70% reduction in Akt phosphorylation. These results suggest that the SC-236 induced reduction of M2PK-positive oval cell numbers may be due to COX-2 dependent inhibition of Akt phosphorylation and induction of apoptosis.


Asunto(s)
Antimetabolitos/administración & dosificación , Deficiencia de Colina , Inhibidores de la Ciclooxigenasa 2/farmacología , Modelos Animales de Enfermedad , Etionina/administración & dosificación , Hepatopatías/prevención & control , Hígado/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/química , Ciclooxigenasa 2/metabolismo , Dieta , Suplementos Dietéticos , Hígado/lesiones , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Piruvato Quinasa/metabolismo , Células Madre/metabolismo , Sulfonamidas/farmacología
14.
Exp Cell Res ; 306(1): 252-63, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15878349

RESUMEN

Following acute injury, the liver regenerates through hepatocyte division. If this pathway is impaired, liver repair depends on the recruitment of adult liver progenitor (oval) cells. Mice fed a choline deficient, ethionine supplemented (CDE) diet possess substantial numbers of oval cells, which can be isolated, or examined in vivo. Oncostatin M (OSM) has been shown to induce maturation of murine fetal hepatoblasts into hepatocytes. We recently confirmed this in human fetal liver cultures. Here, we show that liver OSM expression increases in mice fed a CDE diet and CDE-derived oval cell isolates express OSM and its receptor (OSMR). Oval cell lines (PIL cells), as well as primary oval cell cultures, displayed STAT-3 phosphorylation following OSM stimulation. OSM had no effect on the growth of primary oval cells, but it was pro-apoptotic to PIL cells, suggesting that the two cell models are not directly comparable. Expression of PCNA and cyclin D1 was not affected by OSM treatment. No evidence was obtained to suggest an effect on oval cell maturation with OSM treatment. However, decreased albumin production, accompanied by increased expression of haptoglobin and fibrinogen, suggests that OSM induced an acute phase reaction in cultured oval cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hígado/citología , Péptidos/farmacología , Células Madre/efectos de los fármacos , Reacción de Fase Aguda/metabolismo , Animales , Comunicación Autocrina/genética , Ciclo Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colina/metabolismo , Proteínas de Unión al ADN/metabolismo , Etionina/farmacología , Fibrinógeno/genética , Expresión Génica/efectos de los fármacos , Haptoglobinas/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Oncostatina M , Péptidos/genética , Péptidos/metabolismo , Fosforilación , Receptores de Citocinas/genética , Receptores de Oncostatina M , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3 , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Células Madre/metabolismo , Transactivadores/metabolismo
15.
Carcinogenesis ; 26(10): 1782-92, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15917308

RESUMEN

Multifaceted evidence links the development of liver tumours to the activation and proliferation of adult liver progenitor (oval) cells during the early stages of chronic liver injury. The aim of this study was to examine the role of the peroxisome proliferator activated receptors (PPARs): PPARalpha, delta and gamma, in mediating the behaviour of liver progenitor cells during pre-neoplastic disease and to investigate their potential as therapeutic targets for the treatment of chronic liver injury. We observed increased liver expression of PPARalpha and gamma in concert with expanding oval cell numbers during the first 21 days following commencement of the choline deficient, ethionine supplemented (CDE) dietary model of carcinogenic liver injury in mice. Both primary and immortalized liver progenitor cells were found to express PPARalpha, delta and gamma, but not gamma2, the alternate splice form of PPARgamma. WY14643 (PPARalpha agonist), GW501516 (PPARdelta agonist) and ciglitazone (PPARgamma agonist) were tested for their ability to modulate the behaviour of p53-immortalized liver (PIL) progenitor cell lines in vitro. Both PPARdelta and gamma agonists induced dose-dependent growth inhibition and apoptosis of PIL cells. In contrast, the PPARalpha agonist had no effect on PIL cell growth. None of the drugs affected the maturation of PIL cells along either the hepatocytic or biliary lineages, as judged by their patterns of hepatic gene expression prior to and following treatment. Administration of the PPARgamma agonist ciglitazone to mice fed with the CDE diet for 14 days resulted in a significantly diminished oval cell response and decreased fibrosis compared with those receiving placebo. In contrast, GW501516 did not affect oval cell numbers or liver fibrosis, but inhibited CDE-induced hepatic steatosis. In summary, PPARgamma agonists reduce oval cell proliferation and fibrosis during chronic liver injury and may be useful in the prevention of hepatocellular carcinoma.


Asunto(s)
Etionina/toxicidad , Hígado/citología , PPAR alfa/fisiología , PPAR gamma/agonistas , Receptores Citoplasmáticos y Nucleares/fisiología , Células Madre/citología , Animales , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR alfa/agonistas , PPAR alfa/genética , PPAR gamma/genética , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética , Tiazoles/farmacología , Tiazolidinedionas/farmacología
16.
Hepatology ; 41(2): 327-35, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15660390

RESUMEN

The liver regenerates after acute injury via hepatocyte cell division; during chronic injury, when hepatocyte replication is impaired or blocked, liver progenitor oval cells mediate liver regeneration. If both regeneration options are blocked in animal models, then liver failure and death ensues. The mechanisms underlying oval cell induction, proliferation, and subsequent liver regeneration remain poorly characterized. In particular, cell-signaling pathways that distinguish the alternative pathways are unknown. This study shows that in a mouse model, hepatic expression of lymphotoxin-beta (LTbeta) and interferon gamma (IFNgamma) transcripts is increased in response to the choline-deficient, ethionine-supplemented (CDE) diet, which induces oval cell-mediated liver regeneration. Oval cells express LTbeta and IFNgamma transcripts, contributing to the increased expression in the liver of mice fed the CDE diet. An attenuated oval cell response to such a diet was observed in LTbeta receptor-, LTbeta-, and IFNgamma-gene targeted mice. Loss of LTbeta and LTbeta receptor signaling reduced the number of oval cells expressing A6 and muscle pyruvate kinase. The lack of IFNgamma signaling reduced muscle pyruvate kinase(+), but not A6(+), oval cells. In contrast, partial hepatectomy suppressed LTbeta and IFNgamma transcripts. We also show that IFNgamma induces STAT-3 phosphorylation in an oval cell line. In conclusion, LTbeta, LTbeta receptor, and IFNgamma are involved in oval cell-mediated, but not hepatocyte-mediated, liver regeneration, and the absence of these pathways impairs the oval cell-dependent regenerative response.


Asunto(s)
Interferón gamma/metabolismo , Regeneración Hepática , Hígado/lesiones , Linfotoxina-alfa/metabolismo , Proteínas de la Membrana/metabolismo , Transducción de Señal , Enfermedad Aguda , Animales , Línea Celular , Deficiencia de Colina , Enfermedad Crónica , Proteínas de Unión al ADN/metabolismo , Dieta/efectos adversos , Relación Dosis-Respuesta a Droga , Etionina/administración & dosificación , Hepatectomía/métodos , Interferón gamma/genética , Interferón gamma/farmacología , Hígado/metabolismo , Hígado/patología , Linfotoxina-alfa/genética , Linfotoxina beta , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación/efectos de los fármacos , ARN Mensajero/metabolismo , Factor de Transcripción STAT3 , Células Madre/metabolismo , Transactivadores/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Heridas y Lesiones/fisiopatología
17.
Cell Tissue Res ; 319(1): 61-70, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15592751

RESUMEN

Following acute liver injury, hepatocytes divide to facilitate regeneration. However, during chronic injury, hepatocyte proliferation is typically blocked and repair is mediated through liver progenitor (oval) cells. Signalling of the p55 tumour necrosis factor (TNF) receptor is central to these processes. Two ligands for p55 are known: TNF and lymphotoxin-alpha (LTalpha). However, one study suggests that another exists that mediates liver injury following viral challenge. We have therefore investigated whether ligands other than TNF and LTalpha are required for liver regeneration following either acute or chronic injury. Wild-type and double TNF/LTalpha knockout (TNF-/-LTalpha-/-) mice were subjected to either partial hepatectomy (PHx) or a choline-deficient ethionine-supplemented (CDE) diet. Proliferating hepatocytes, oval cells and inflammatory cells were identified and quantified in liver sections by immunohistochemistry. Liver inflammatory cells were characterised by cell surface antigen expression. Liver damage and mortality were monitored. Both hepatocyte and oval cell proliferation was reduced in TNF-/-LTalpha-/- mice. Lymphocyte clusters were evident in all TNF-/-LTalpha-/- livers and were heterogeneous, comprising B and T lymphocytes. PHx evoked liver inflammation in TNF-/-LTalpha-/- but not wild-type mice, whereas no difference was apparent between genotypes in CDE experiments. Thus, TNF/LTalpha signalling mediates liver regeneration involving both hepatocytes and progenitor cells. The hyper-inflammatory response following PHx in TNF-/-LTalpha-/- animals, which is absent following CDE-induced injury, demonstrates that the two forms of liver injury evoke discrete inflammatory responses and provides a model in which such differences can be examined further.


Asunto(s)
Hepatitis/patología , Hepatocitos/inmunología , Regeneración Hepática , Hígado/patología , Enfermedad Aguda , Animales , Antígenos de Superficie/biosíntesis , Diferenciación Celular , Proliferación Celular , Enfermedad Crónica , Dieta , Etionina/administración & dosificación , Hepatectomía , Hepatitis/etiología , Hepatitis/inmunología , Hepatocitos/patología , Inmunohistoquímica , Linfocitos/patología , Linfotoxina-alfa/genética , Ratones , Ratones Noqueados , Células Madre/inmunología , Células Madre/patología , Factores de Necrosis Tumoral/genética
18.
Wound Repair Regen ; 12(6): 650-6, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15555057

RESUMEN

Following acute injury, liver is usually regenerated from hepatocytes by a process that is dependent on interleukin (IL)-6. If this pathway is impaired, restoration of the liver mass and ultimately the survival of the animal are dependent on recruitment of cells from a precursor cell population, either a stem cell or an oval cell. Importantly, oval cells are also implicated in tumorigenesis. A carcinogenic choline-deficient ethionine supplemented (CDE) diet is capable of inducing substantial numbers of oval cells that we can isolate and utilize to identify cytokines, which affect oval cell proliferation and differentiation. Currently, a putative role of IL-6 in oval cell biology is suggested by the elevation of IL-6 in liver and serum of mice treated with a CDE diet and knockout mouse studies. Also, when IL-6 is injected into the peritoneal cavity of mice on the CDE diet, oval cell numbers are increased compared to mice on the CDE diet alone. We investigated the role of human IL-6 on p53 null immortalized murine oval cell lines (PIL), finding that they express transcripts for the IL-6 receptor and gp 130, STAT-3 is phosphorylated upon IL-6 stimulation, IL-6 induces IL-6 production, and proliferation is induced by IL-6. In addition, we show that mouse primary oval cells also express IL-6 receptor and gp 130 mRNA. These findings suggest that IL-6 directly stimulates oval cells and an autocrine mechanism may sustain oval cell proliferation.


Asunto(s)
Interleucina-6/farmacología , Hígado/citología , Células Madre/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Deficiencia de Colina , Ensayo de Inmunoadsorción Enzimática , Etionina/farmacología , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
19.
Carcinogenesis ; 23(10): 1685-93, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12376478

RESUMEN

To examine the effect of ethanol on hepatocarcinogenesis induced by a choline-deficient, ethionine-supplemented (CDE) diet, rats were fed either an ethanol-supplemented diet or ethanol-free, isocaloric diet for 2 months, followed by a CDE diet or control diet for up to 8 months. Changes to cellular composition and pattern of gene expression in the liver were determined at 0 and 3 days, and 1, 2 and 3 weeks after commencing the CDE diet, using histological/immunochemical techniques and northern analysis. Oval cells in the liver were identified morphologically and by expression of pi-glutathione S-transferase (pi-GST), alpha-fetoprotein (AFP) and the embryonic isoform of pyruvate kinase (M2-PK). Oval cell numbers and changes in the pattern of gene expression induced by the CDE diet were accelerated by pre-treatment with ethanol. At all stages, the proportion of oval cells in the test group exceeded that in controls. After 1 week, oval cells had spread sufficiently from the periportal region to be observed pericentrally in test animals and by 3 weeks, extensive formation of ductal structures was apparent, which were absent in controls. Additionally, M2-PK and AFP mRNA were detected earlier, and in greater abundance in animals pre-treated with ethanol. After 8 months of CDE treatment, one or two small hepatic foci (<10 hepatocytes), strongly positive for pi-GST, were detected in the liver of ethanol-pre-treated animals. These foci were absent in CDE-treated animals; however, animals pre-treated with ethanol followed by chronic CDE treatment showed increased size (>40 hepatocytes) and numbers of foci, correlating with the extent of liver damage and varying from 5 to 50% of the liver section. Our data suggest that ethanol pre-treatment potentiates the short-term effects of the CDE diet by enhancing oval cell proliferation, while chronic CDE administration enhances the appearance of pre-malignant hepatic foci that are observed with ethanol pre-treatment alone.


Asunto(s)
Deficiencia de Colina , Etanol/farmacología , Etionina/farmacología , Hígado/patología , Lesiones Precancerosas/inducido químicamente , Animales , Suplementos Dietéticos , Etionina/administración & dosificación , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glutatión Transferasa/genética , Isoenzimas/genética , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Lesiones Precancerosas/patología , Piruvato Quinasa/genética , Ratas , Ratas Wistar , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , alfa-Fetoproteínas/genética
20.
Carcinogenesis ; 23(3): 435-45, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11895858

RESUMEN

Oval cells are bipotential liver stem cells able to differentiate into hepatocytes and bile duct epithelia. In normal adult liver oval cells are quiescent, existing in low numbers around the periportal region, and proliferate following severe, prolonged liver trauma. There is evidence implicating oval cells in the development of hepatocellular carcinoma, and hence the availability of an immortalized oval cell line would be invaluable for the study of liver cell lineage differentiation and carcinogenesis. A novel approach in the generation of cell lines is the use of the p53 knockout mouse. Absence of p53 allows a cell to cycle past the normal Hayflick limit, rendering it immortalized, although subsequent genetic alterations are thought necessary for transformation. p53 knockout mice were fed a choline-deficient, ethionine-supplemented diet, previously shown to increase oval cell numbers in wild-type mice. The oval cells were isolated by centrifugal elutriation and maintained in culture. Colonies of hepatic cells were isolated and characterized with respect to phenotype, growth characteristics and tumorigenicity. Analysis of gene expression by Northern blotting and immunocytochemistry suggests they are oval-like cells by virtue of albumin and transferrin expression, as well as the oval cell markers alpha fetoprotein, M(2)-pyruvate kinase and A6. Injection into athymic nude mice shows the cell lines are capable of forming tumors which phenotypically resemble hepatocellular carcinoma. Thus, the use of p53 null hepatic cells successfully generated immortalized and tumorigenic hepatic stem cell lines. The results presented support the idea that deleting p53 allows immortalization and contributes to the transformation of the oval-like cell lines. Further, the tumorigenic status of the cell lines is direct evidence for the participation of oval cells in the formation of hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Eliminación de Gen , Hepatocitos/patología , Células Madre/patología , Proteína p53 Supresora de Tumor/genética , Animales , Carcinoma Hepatocelular/metabolismo , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Transformada , Tamaño de la Célula/efectos de los fármacos , Transformación Celular Neoplásica , Etionina/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Inmunohistoquímica , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Ratones Desnudos , Especificidad de Órganos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA