Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Lab Invest ; 97(5): 577-590, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28218739

RESUMEN

During progression to type 1 diabetes, insulin-producing ß-cells are lost through an autoimmune attack resulting in unrestrained glucagon expression and secretion, activation of glycogenolysis, and escalating hyperglycemia. We recently identified a protein, designated islet homeostasis protein (IHoP), which specifically co-localizes within glucagon-positive α-cells and is overexpressed in the islets of both post-onset non-obese diabetic (NOD) mice and type 1 diabetes patients. Here we report that in the αTC1.9 mouse α-cell line, IHoP was released in response to high-glucose challenge and was found to regulate secretion of glucagon. We also show that in NOD mice with diabetes, major histocompatibility complex class II was upregulated in islets. In addition hyperglycemia was modulated in NOD mice via suppression of IHoP utilizing small interfering RNA (IHoP-siRNA) constructs/approaches. Suppression of IHoP in the pre-diabetes setting maintained normoglycemia, glyconeolysis, and fostered ß-cell restoration in NOD mice 35 weeks post treatment. Furthermore, we performed adoptive transfer experiments using splenocytes from IHoP-siRNA-treated NOD/ShiLtJ mice, which thwarted the development of hyperglycemia and the extent of insulitis seen in recipient mice. Last, IHoP can be detected in the serum of human type 1 diabetes patients and could potentially serve as an early novel biomarker for type 1 diabetes in patients.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas/metabolismo , Animales , Línea Celular , Femenino , Glucagón/análisis , Glucagón/metabolismo , Antígenos HLA-D/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Hiperglucemia/metabolismo , Islotes Pancreáticos/química , Masculino , Ratones , Ratones Endogámicos NOD , Proteínas/análisis , Proteínas/antagonistas & inhibidores , Transactivadores/metabolismo
2.
Am J Pathol ; 186(5): 1092-102, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26945106

RESUMEN

miRNAs are involved in liver regeneration, and their expression is dysregulated in hepatocellular carcinoma (HCC). Connective tissue growth factor (CTGF), a direct target of miR-133b, is crucial in the ductular reaction (DR)/oval cell (OC) response for generating new hepatocyte lineages during liver injury in the context of hepatotoxin-inhibited hepatocyte proliferation. Herein, we investigate whether miR-133b regulation of CTGF influences HCC cell proliferation and migration, and DR/OC response. We analyzed miR-133b expression and found it to be down-regulated in HCC patient samples and induced in the rat DR/OC activation model of 2-acetylaminofluorene with partial hepatectomy. Furthermore, overexpression of miR-133b via adenoviral system in vitro led to decreased CTGF expression and reduced proliferation and Transwell migration of both HepG2 HCC cells and WBF-344 rat OCs. In vivo, overexpression of miR-133b in DR/OC activation models of 2-acetylaminofluorene with partial hepatectomy in rats, and 3,5-diethoxycarbonyl-1,4-dihydrocollidine in mice, led to down-regulation of CTGF expression and OC proliferation. Collectively, these results show that miR-133b regulation of CTGF is a novel mechanism critical for the proliferation and migration of HCC cells and OC response.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , MicroARNs/fisiología , 2-Acetilaminofluoreno/farmacología , Adenoviridae/genética , Anciano , Animales , Carcinógenos/farmacología , Carcinoma Hepatocelular/fisiopatología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo/fisiología , Femenino , Vectores Genéticos , Células HEK293 , Células Hep G2 , Humanos , Neoplasias Hepáticas/fisiopatología , Masculino , Ratones , MicroARNs/metabolismo , Persona de Mediana Edad , Ratas , Transfección
3.
Am J Pathol ; 185(6): 1552-63, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25843683

RESUMEN

Hepatic progenitor/oval cell (OC) activation occurs when hepatocyte proliferation is inhibited and is tightly associated with the fibrogenic response during severe liver damage. Connective tissue growth factor (CTGF) is important for OC activation and contributes to the pathogenesis of liver fibrosis. By using the Yeast Two-Hybrid approach, we identified a disintegrin and metalloproteinase with thrombospondin repeat 7 (ADAMTS7) as a CTGF binding protein. In vitro characterization demonstrated CTGF binding and processing by ADAMTS7. Moreover, Adamts7 mRNA was induced during OC activation, after the implantation of 2-acetylaminofluorene with partial hepatectomy in rats or on feeding a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet in mice. X-Gal staining showed Adamts7 expression in hepatocyte nuclear factor 4α(+) hepatocytes and desmin(+) myofibroblasts surrounding reactive ducts in DDC-treated Adamts7(-/-) mice carrying a knocked-in LacZ gene. Adamts7 deficiency was associated with higher transcriptional levels of Ctgf and OC markers and enhanced OC proliferation compared to Adamts7(+/+) controls during DDC-induced liver injury. We also observed increased α-smooth muscle actin and procollagen type I mRNAs, large fibrotic areas in α-smooth muscle actin and Sirius red staining, and increased production of hepatic collagen by hydroxyproline measurement. These results suggest that ADAMTS7 is a new protease for CTGF protein and a novel regulator in the OC compartment, where its absence causes CTGF accumulation, leading to increased OC activation and biliary fibrosis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Desintegrinas/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Nicho de Células Madre/fisiología , Trombospondinas/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS7 , Animales , Factor de Crecimiento del Tejido Conjuntivo/genética , Desintegrinas/genética , Hígado/patología , Cirrosis Hepática/patología , Regeneración Hepática/fisiología , Ratones , Ratones Noqueados , Trombospondinas/genética
4.
Hepatology ; 61(2): 678-91, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25203810

RESUMEN

UNLABELLED: Connective tissue growth factor (CTGF) is a matricellular protein that mediates cell-matrix interaction through various subtypes of integrin receptors. This study investigated the role of CTGF and integrin αvß6 in hepatic progenitor/oval cell activation, which often occurs in the form of ductular reactions (DRs) when hepatocyte proliferation is inhibited during severe liver injury. CTGF and integrin αvß6 proteins were highly expressed in DRs of human cirrhotic livers and cholangiocarcinoma. Confocal microscopy analysis of livers from Ctgf promoter-driven green fluorescent protein reporter mice suggested that oval cells and cholangiocytes were the main sources of CTGF and integrin αvß6 during liver injury induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC). Deletion of exon 4 of the Ctgf gene using tamoxifen-inducible Cre-loxP system down-regulated integrin αvß6 in DDC-damaged livers of knockout mice. Ctgf deficiency or inhibition of integrin αvß6, by administrating the neutralizing antibody, 6.3G9 (10 mg/kg body weight), caused low levels of epithelial cell adhesion molecule and cytokeratin 19 gene messenger RNAs. Also, there were smaller oval cell areas, fewer proliferating ductular epithelial cells, and lower cholestasis serum markers within 2 weeks after DDC treatment. Associated fibrosis was attenuated, as indicated by reduced expression of fibrosis-related genes, smaller areas of alpha-smooth muscle actin staining, and low collagen production based on hydroxyproline content and Sirius Red staining. Finally, integrin αvß6 could bind to CTGF mediating oval cell adhesion to CTGF and fibronection substrata and promoting transforming growth factor (TGF)-ß1 activation in vitro. CONCLUSIONS: CTGF and integrin αvß6 regulate oval cell activation and fibrosis, probably through interacting with their common matrix and signal partners, fibronectin and TGF-ß1. CTGF and integrin αvß6 are potential therapeutic targets to control DRs and fibrosis in related liver disease.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Integrinas/metabolismo , Cirrosis Hepática/metabolismo , Células Madre Adultas/metabolismo , Animales , Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares Intrahepáticos , Adhesión Celular , Colangiocarcinoma/metabolismo , Femenino , Fibronectinas/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Piridinas , Conejos , Ratas , Factor de Crecimiento Transformador beta1/metabolismo
5.
Am J Pathol ; 183(2): 558-65, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23747949

RESUMEN

Liver disease affects millions of patients each year. The field of regenerative medicine promises alternative therapeutic approaches, including the potential to bioengineer replacement hepatic tissue. One approach combines cells with acellular scaffolds derived from animal tissue. The goal of this study was to scale up our rodent liver decellularization method to livers of a clinically relevant size. Porcine livers were cannulated via the hepatic artery, then perfused with PBS, followed by successive Triton X-100 and SDS solutions in saline buffer. After several days of rinsing, decellularized liver samples were histologically analyzed. In addition, biopsy specimens of decellularized scaffolds were seeded with hepatoblastoma cells for cytotoxicity testing or implanted s.c. into rodents to investigate scaffold immunogenicity. Histological staining confirmed cellular clearance from pig livers, with removal of nuclei and cytoskeletal components and widespread preservation of structural extracellular molecules. Scanning electron microscopy confirmed preservation of an intact liver capsule, a porous acellular lattice structure with intact vessels and striated basement membrane. Liver scaffolds supported cells over 21 days, and no increased immune response was seen with either allogeneic (rat-into-rat) or xenogeneic (pig-into-rat) transplants over 28 days, compared with sham-operated on controls. These studies demonstrate that successful decellularization of the porcine liver could be achieved with protocols developed for rat livers, yielding nonimmunogenic scaffolds for future hepatic bioengineering studies.


Asunto(s)
Hígado/citología , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Hígado/inmunología , Trasplante de Hígado/inmunología , Masculino , Ratas , Ratas Endogámicas F344 , Sus scrofa , Porcinos , Trasplante Heterólogo
6.
FASEB J ; 26(8): 3365-79, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22611085

RESUMEN

Blood vessels are formed during development and tissue repair through a plethora of modifiers that coordinate efficient vessel assembly in various cellular settings. Here we used the yeast 2-hybrid approach and demonstrated a broad affinity of connective tissue growth factor (CCN2/CTGF) to C-terminal cystine knot motifs present in key angiogenic regulators Slit3, von Willebrand factor, platelet-derived growth factor-B, and VEGF-A. Biochemical characterization and histological analysis showed close association of CCN2/CTGF with these regulators in murine angiogenesis models: normal retinal development, oxygen-induced retinopathy (OIR), and Lewis lung carcinomas. CCN2/CTGF and Slit3 proteins worked in concert to promote in vitro angiogenesis and downstream Cdc42 activation. A fragment corresponding to the first three modules of CCN2/CTGF retained this broad binding ability and gained a dominant-negative function. Intravitreal injection of this mutant caused a significant reduction in vascular obliteration and retinal neovascularization vs. saline injection in the OIR model. Knocking down CCN2/CTGF expression by short-hairpin RNA or ectopic expression of this mutant greatly decreased tumorigenesis and angiogenesis. These results provided mechanistic insight into the angiogenic action of CCN2/CTGF and demonstrated the therapeutic potential of dominant-negative CCN2/CTGF mutants for antiangiogenesis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/fisiología , Motivos Nodales de Cisteina/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Animales , Carcinoma Pulmonar de Lewis/inducido químicamente , Motivos Nodales de Cisteina/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas de la Membrana/fisiología , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Vasos Retinianos/crecimiento & desarrollo , Técnicas del Sistema de Dos Híbridos
7.
Liver Int ; 32(2): 312-20, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22098068

RESUMEN

BACKGROUND: Somatostatin is a pleiotropic peptide, exerting a variety of effects through its receptor subtypes. Recently, somatostatin has been shown to act as a chemoattractant for haematopoietic progenitor cells and hepatic oval cells (HOC) via receptor subtype 2 and subtype 4 (SSTR4) respectively. AIMS: We investigated the in vivo effect of somatostatin/SSTR4 on HOC migration in the injured liver model of rats and the type of signalling molecules associated with the chemotactic function. METHODS: Migration assay, HOC transplantation and phosphatidylinositol-3-kinase (PI3K) signalling were assessed with or without somatostatin and an analogue of somatostatin (TT232) that specifically binds to SSTR4. RESULTS: TT232 was shown to have an antimigratory action on HOC induced by somatostatin in vitro. In HOC transplantation experiments, a lower number of donor-derived cells were detected in TT232-treated animals, as compared with control animals. Activation of PI3K was observed in HOC exposed to somatostatin, and this activation was suppressed by either SSTR4 antibody or TT232-pretreatment. In addition, a PI3K inhibitor abrogated the motility of HOC. CONCLUSION: Together, these data suggest that somatostatin stimulates the migration of HOC within injured liver through SSTR4, and this action appears to be mediated by the PI3K pathway.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hígado/efectos de los fármacos , Péptidos Cíclicos/farmacología , Somatostatina/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Trasplante de Células , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Hepatocitos/patología , Hígado/patología , Masculino , Péptidos Cíclicos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores de Somatostatina/metabolismo , Transducción de Señal/efectos de los fármacos , Somatostatina/metabolismo
8.
Am J Pathol ; 176(6): 2732-42, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20413689

RESUMEN

To date the molecular signals regulating activation, proliferation, and differentiation of hepatic oval cells are not fully understood. The Wnt family is essential in hepatic embryogenesis and implicated in hepatic carcinogenesis. This study elucidates novel findings implicating Wnt1 in directing oval cell differentiation during the rat 2-acetylaminofluorene (2AAF) and 2/3 partial hepatectomy (PHx) liver regeneration model. Proteins of Wnt family members were predominantly localized in pericentral hepatocytes during liver injury, oval cell activation, and hepatocyte regeneration. In addition, Wnt message increased coinciding with the rise in oval cell number, whereas protein levels peaked immediately after the height of oval cell proliferation. Immunohistochemical analysis demonstrated nuclear translocation of beta-catenin within oval cells throughout the 2AAF/PHx protocol. Furthermore, RNA interference was used in vivo to confirm the physiological requirement of Wnt1 during the oval cell induction. Ultimately, inhibition of Wnt1 resulted in failure of oval cells to differentiate into hepatocytes and alternatively induced atypical ductular hyperplasia. Taken together, these data indicate that in vivo exposure to Wnt1 shRNA inhibited rat oval cell liver regeneration. In the absence of Wnt1 signaling, oval cells failed to differentiate into hepatocytes and underwent atypical ductular hyperplasia, exhibiting epithelial metaplasia and mucin production. Furthermore, changes in Wnt1 levels are required for the efficient regeneration of the liver by oval cells during massive hepatic injury.


Asunto(s)
Diferenciación Celular/fisiología , Regeneración Hepática/fisiología , Hígado/citología , Células Madre/fisiología , Proteína Wnt1/metabolismo , 2-Acetilaminofluoreno/farmacología , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Carcinógenos/farmacología , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Hepatectomía , Hígado/efectos de los fármacos , Hígado/patología , Hígado/cirugía , Masculino , Tamaño de los Órganos , Interferencia de ARN , Ratas , Ratas Endogámicas F344 , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Transducción de Señal/fisiología , Células Madre/citología , Proteína Wnt1/genética , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
10.
Lab Invest ; 90(8): 1199-208, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20440274

RESUMEN

Earlier studies conducted by our laboratory have shown that suppression of transforming growth factor-beta (TGFbeta)-mediated upregulation of connective tissue growth factor (CTGF) by iloprost resulted in a greatly diminished oval cell response to 2-acetylaminofluorene/partial hepatectomy (2AAF/PH) in rats. We hypothesized that this effect is due to decreased activation of hepatic stellate cells. To test this hypothesis, we maintained rats on a diet supplemented with 2% L-cysteine as a means of inhibiting stellate cell activation during the oval cell response to 2AAF/PH. In vitro experiments show that L-cysteine did, indeed, prevent the activation of stellate cells while exerting no direct effect on oval cells. Desmin immunostaining of liver sections from 2AAF/PH animals indicated that maintenance on the L-cysteine diet resulted in an 11.1-fold decrease in the number of activated stellate cells within the periportal zones. The total number of cells proliferating in the periportal zones of livers from animals treated with L-cysteine was drastically reduced. Further analyses showed a greater than fourfold decrease in the magnitude of the oval cell response in animals maintained on the L-cysteine diet as determined by immunostaining for both OV6 and alpha-fetoprotein (AFP). Global liver expression of AFP as measured by real-time PCR was shown to be decreased 4.7-fold in the L-cysteine-treated animals. These data indicate that the activation of hepatic stellate cells is required for an appropriate oval cell response to 2AAF/PH.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Regeneración Hepática/fisiología , Células Madre/metabolismo , 2-Acetilaminofluoreno/metabolismo , 2-Acetilaminofluoreno/farmacología , Animales , Factor de Crecimiento del Tejido Conjuntivo , Cisteína/metabolismo , Cisteína/farmacología , Hepatectomía , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hepatopatías/metabolismo , Regeneración Hepática/efectos de los fármacos , Masculino , Ratas , Ratas Endogámicas F344 , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , alfa-Fetoproteínas/metabolismo , alfa-Fetoproteínas/farmacología
11.
Lab Invest ; 89(9): 1032-42, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19581879

RESUMEN

Glycogen storage disease type Ia (GSDIa) is caused by a genetic defect in the hepatic enzyme glucose-6-phosphatase (G6Pase-alpha), which manifests as life-threatening hypoglycemia with related metabolic complications. A G6Pase-alpha knockout (KO) mouse model was generated to study potential therapies for correcting this disorder. Since then, gene therapy studies have produced promising results, showing long-term improvement in liver histology and glycogen metabolism. Under existing protocols, however, untreated KO pups seldom survived weaning. Here, we present a thorough characterization of the G6Pase-alpha KO mouse, as well as the husbandry protocol for rearing this strain to adulthood. These mice were raised with only palliative care, and characterized from birth through 6 months of age. Once KO mice have survived the very frail weaning period, their size, agility, serum lipids and glycemic control improve dramatically, reaching levels approaching their wild-type littermates. In addition, our data reveal that adult mice lacking G6Pase-alpha are able to mate and produce viable offspring. However, liver histology and glycogen accumulation do not improve with age. Overall, the reliable production of mature KO mice could provide a critical tool for advancing the GSDIa field, as the availability of a robust enzyme-deficient adult offers a new spectrum of treatment avenues that would not be tolerated by the frail pups. Most importantly, our detailed characterization of the adult KO mouse provides a crucial baseline for accurately gauging the efficacy of experimental therapies in this important model.


Asunto(s)
Glucosa-6-Fosfatasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I , Hipoglucemia/patología , Hígado/patología , Crianza de Animales Domésticos/métodos , Animales , Animales Recién Nacidos , Animales Lactantes , Análisis Químico de la Sangre , Peso Corporal/fisiología , Modelos Animales de Enfermedad , Femenino , Fibrosis , GTP Fosfohidrolasas/metabolismo , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo I/patología , Hipoglucemia/genética , Hipoglucemia/metabolismo , Riñón/enzimología , Riñón/patología , Hígado/enzimología , Glucógeno Hepático/análisis , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/enzimología , Tamaño de los Órganos/efectos de los fármacos , Reproducción/fisiología , Destete
12.
J Hepatol ; 51(1): 77-92, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19446912

RESUMEN

BACKGROUND/AIMS: Oval cells (OCs), putative hepatic stem cells, may give rise to liver cancers. We developed a carcinogenesis regimen, based upon induction of OC proliferation prior to carcinogen exposure. In our model, rats subjected to 2-acetylaminofluorene/ partial-hepatectomy followed by aflatoxin injection (APA regimen) developed well-differentiated hepatocholangiocarcinomas. The aim of this study was to establish and characterize cancer cell lines from this animal model. METHODS: Cancer cells were cultured from animals sacrificed eight months after treatment, and single clones were selected. The established cell lines, named LCSCs, were characterized, and their tumorigenicity was assessed in vivo. The roles of granulocyte-colony stimulating factor (G-CSF) and hepatocyte growth factor (HGF) in LCSC growth, survival and motility were also investigated. RESULTS: From primary tumors, six cell lines were developed. LCSCs shared with the primary tumors the expression of various OC-associated markers, including cMet and G-CSF receptor. In vitro, HGF conferred protection from death by serum withdrawal. Stimulation with G-CSF increased LCSC growth and motility, while the blockage of its receptor inhibited LCSC proliferation and migration. CONCLUSIONS: Six cancer cell lines were established from our model of hepatocholangiocarcinoma. HGF modulated LCSC resistance to apoptosis, while G-CSF acted on LCSCs as a proliferative and chemotactic agent.


Asunto(s)
Carcinoma Hepatocelular/patología , Colangiocarcinoma/patología , Factor Estimulante de Colonias de Granulocitos/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Neoplasias Hepáticas/patología , 2-Acetilaminofluoreno/toxicidad , Aflatoxina B1/toxicidad , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Hepatectomía , Masculino , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-met/fisiología , Ratas , Ratas Endogámicas F344 , Receptores de Factor Estimulante de Colonias de Granulocito/fisiología
13.
Hepatology ; 47(3): 996-1004, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18167060

RESUMEN

UNLABELLED: Oval cell activation, as part of the regenerative process after liver injury, involves considerable cell-matrix interaction. The matricellular protein, connective tissue growth factor (CTGF), has been shown to be critical for oval cell activation during liver regeneration following N-2-acetylaminofluorene/partial hepatectomy. To understand the mode of action of CTGF during this process, N-terminal CTGF was used as bait to screen a yeast two-hybrid complementary DNA library specific for regenerating livers with massive oval cell presence. Fibronectin (FN), a prominent component of hepatic extracellular matrix (ECM), was found to specifically bind to a new site on CTGF. In addition to module IV, this study showed that module I of CTGF was sufficient for binding to FN in both solid-phase in vitro binding assays and immunoprecipitation. Immunofluorescent staining revealed a dynamic ECM remodeling characterized by an FN-concentrated provisional matrix during oval cell-aided liver regeneration. Abundant CTGF protein was colocalized with FN in the provisional matrix. When expressed as recombinant proteins and immobilized on plastic surfaces, modules I and IV of CTGF were selectively adhesive to thymus cell antigen 1-positive (Thy1(+)) oval cells, stellate cells, and sinusoidal endothelial cells but not to hepatocytes. The adhesion of these two modules on Thy1(+) oval cells required heparan sulfate proteoglycan and integrin alpha(5)beta(1). Recombinant CTGF promoted an integrin alpha(5)beta(1)-dependent migration but not proliferation on Thy1(+) oval cells. CONCLUSION: Modules I and IV enabled the linkage of CTGF to FN and activated hepatic cells. Through these bindings, CTGF on the FN-concentrated provisional matrix promoted cell adhesion and migration, thereby facilitating oval cell activation.


Asunto(s)
Adhesión Celular , Movimiento Celular , Fibronectinas/metabolismo , Hepatocitos/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Regeneración Hepática , Animales , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo , Matriz Extracelular/metabolismo , Fibronectinas/análisis , Proteoglicanos de Heparán Sulfato/metabolismo , Hepatocitos/efectos de los fármacos , Proteínas Inmediatas-Precoces/análisis , Proteínas Inmediatas-Precoces/genética , Integrina alfa5beta1/metabolismo , Péptidos y Proteínas de Señalización Intercelular/análisis , Péptidos y Proteínas de Señalización Intercelular/genética , Regeneración Hepática/efectos de los fármacos , Masculino , Ratas , Ratas Endogámicas F344 , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Antígenos Thy-1/análisis , Técnicas del Sistema de Dos Híbridos
14.
Oncol Res ; 17(8): 339-45, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19544970

RESUMEN

Metaplastic tubular complexes (MTC) have been proposed as precursor lesions for pancreatic adenocarcinoma (PDAC). In this study, we investigated the potential role of bone marrow-derived progenitor cells (BMPC) in the formation of MTC and PDAC in a rat model. F344 rats defective for CD26 (dipeptidyl peptidase IV, DPPIV) expression were sublethally irradiated and received rescue bone marrow cells from wild-type F344 rats that express CD26. After confirming engraftment, recipient animals received dimethylbenzanthracene (DMBA) implantation in their pancreas. Animals were sacrificed monthly from 3 to 7 months. We observed both MTC and tumors in animals that received DMBA. These MTC were ductal complexes because they stained positive for cytokeratin but were negative for chymotrypsin and chromogranin A. Cells that expressed both CD26 and cytokeratin were rarely observed in the MTC. Cells expressing either both CD26 and CD45 or CD26 and smooth muscle actin were also found near the MTC. However, no CD26 signal was detected in the tumors. Within this model, there appeared to be no evidence supporting that BMPC turned into tumor cells directly. BMPC could modulate pancreatic cancer growth through tumor microenvironment.


Asunto(s)
Adenocarcinoma/patología , Células de la Médula Ósea/patología , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/patología , Lesiones Precancerosas/patología , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Adenocarcinoma/inducido químicamente , Adenocarcinoma/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Carcinógenos/toxicidad , Dipeptidil Peptidasa 4/metabolismo , Modelos Animales de Enfermedad , Inmunohistoquímica , Antígenos Comunes de Leucocito/metabolismo , Masculino , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/inducido químicamente , Neoplasias Pancreáticas/metabolismo , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/metabolismo , Ratas , Ratas Endogámicas F344
15.
Methods Mol Biol ; 482: 387-405, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19089369

RESUMEN

The pace of research on the potential therapeutic uses of liver stem cells or "oval cells" has accelerated significantly in recent years. Concurrent advancements in techniques for the isolation and characterization of these cells have helped fuel this research. Several models now exist for the induction of oval cell proliferation in rodents. Protocols for the isolation and culture of these cells have evolved to the point that they may be set up in any laboratory equipped for cell culture. The advent of magnetic cell sorting has eliminated reliance on expensive flow cytometric sorting equipment to generate highly enriched populations of oval cells. Our laboratory has had much success in using the oval cell surface marker Thy-1 in combination with magnetic sorting to produce material suitable for testing the influence of a myriad of chemical signaling molecules on the oval cell phenotype. This chapter will describe our basic strategy for oval cell induction and isolation. Additionally, two in vitro procedures are described which the reader may find useful in the early stages of developing an oval cell research project.


Asunto(s)
Separación Celular/métodos , Hígado/citología , Células Madre/citología , Animales , Movimiento Celular , Colagenasas/metabolismo , Citometría de Flujo , Gravitación , Hepatectomía , Separación Inmunomagnética , Perfusión , Ratas
16.
Liver Res ; 1(2): 81-87, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29276644

RESUMEN

The liver possesses an extraordinary ability to regenerate after injury. Hepatocyte-driven liver regeneration is the default pathway in response to mild-to-moderate acute liver damage. When replication of mature hepatocytes is blocked, facultative hepatic progenitor cells (HPCs), also referred to as oval cells (OCs) in rodents, are activated. HPC/OCs have the ability to proliferate clonogenically and differentiate into several lineages including hepatocytes and bile ductal epithelia. This is a conserved liver injury response that has been studied in many species ranging from mammals (rat, mouse, and human) to fish. In addition, improper HPC/OC activation is closely associated with fibrotic responses, characterized by myofibroblast activation and extracellular matrix production, in many chronic liver diseases. Matrix remodeling and metalloprotease activities play an important role in the regulation of HPC/OC proliferation and fibrosis progression. Thus, understanding molecular mechanisms underlying HPC/OC activation has therapeutic implications for rational design of anti-fibrotic therapies.

17.
J Histochem Cytochem ; 54(9): 1051-9, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16709727

RESUMEN

Breast cancer resistance protein (BCRP/ABCG2) is an ATP-binding cassette transport protein that is expressed in several organs including the liver. Previous studies have shown that ABC transport proteins play an important pathophysiological role in several liver diseases. However, to date, expression pattern and possible role of BCRP in human liver diseases and animal models have not been studied in detail. Here we investigated the expression pattern of BCRP in normal liver, chronic parenchymal and biliary human liver diseases, and parallel in different rat models of liver diseases. Expression was studied by immunohistochemistry and additionally by RT-PCR analysis in Thy-1-positive rat oval cells. Bile ducts, hepatic progenitor cells, reactive bile ductules, and blood vessel endothelium were immunoreactive for BCRP in normal liver and all types of human liver diseases and in rat models. BCRP was expressed by the canalicular membrane of hepatocytes in normal and diseased human liver, but never in rat liver. Remarkably, there was also expression of BCRP at the basolateral pole of human hepatocytes, and this was most pronounced in chronic biliary diseases. In conclusion, BCRP positivity in the progenitor cells/reactive ductules could contribute to the resistance of these cells to cytotoxic agents and xenotoxins. Basolateral hepatocytic expression in chronic biliary diseases may be an adaptive mechanism to pump bile constituents back into the sinusoidal blood. Strong differences between human and rat liver must be taken into account in future studies with animal models.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/biosíntesis , Conductos Biliares/metabolismo , Hepatocitos/metabolismo , Hepatopatías/metabolismo , Hígado/metabolismo , Proteínas de Neoplasias/biosíntesis , Células Madre/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Animales , Enfermedades de los Conductos Biliares/metabolismo , Enfermedades de los Conductos Biliares/patología , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Humanos , Inmunohistoquímica , Hígado/irrigación sanguínea , Hígado/citología , Hepatopatías/etiología , Hepatopatías/patología , Ratas , Especificidad de la Especie
18.
Cell Transplant ; 14(1): 41-7, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15789661

RESUMEN

Retrorsine has been used extensively to inhibit proliferation of resident hepatocytes in various transplantation models. Here we report a successful alternative to currently unavailable retrorsine that can be used in cellular transplantation models. Based on structural and molecular similarities, we investigate the use of monocrotaline (MCT) in cell transplantation studies in rodents. In this study, MCT was given to rats intraperitoneally in two injections 2 weeks apart. Two weeks after the final injection, a partial hepatectomy followed by splenic hepatocyte transplantation was performed. The results indicate that MCT, at two doses of 30 mg/kg, highly enhances liver repopulation by donor hepatocytes following partial hepatectomy and produces 15.3 +/- 4.9% liver repopulation within the first 6 weeks following transplantation. Additionally, we tested the effectiveness of MCT in a murine model. Using two injections of 50 mg/kg each, given 2 weeks apart, hepatocyte proliferation in the native liver was inhibited and subsequent oval cell transplants engrafted at 18 +/- 21.3% after 16 weeks posttransplantation. In conclusion, MCT can be used as an effective selective pressure for donor hepatocytes in cell transplantation to the liver in rodents.


Asunto(s)
Trasplante de Células/métodos , Hepatocitos/trasplante , Monocrotalina/farmacología , Alcaloides de Pirrolicidina/farmacología , Animales , Animales Modificados Genéticamente , Antineoplásicos Fitogénicos/farmacología , Proliferación Celular/efectos de los fármacos , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Glucosa-6-Fosfatasa/metabolismo , Glucógeno/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hepatectomía , Hepatocitos/efectos de los fármacos , Hígado/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ratas , Ratas Endogámicas F344
19.
Stem Cell Rev ; 1(3): 261-4, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-17142863

RESUMEN

The cellular origin of tumors remains as one of the unanswered, fundamental questions of cancer biology. The notion that tumors may arise from tissue stem cells is supported by phenotypic similarities between these two cell types, such as proliferative potential and expression of onco-fetal proteins. Liver stem cells, or oval cells, have been put forth as a possible target for hepatocarcinogens. Genetically modified and in vitro transformed oval cells have been shown to form tumors in transplantation to animals. Chemical carcinogenesis models in the liver demonstrate varying degrees of oval cell proliferation. There is also preliminary evidence that hepatocellular carcinoma may maintain a bipotential phenotype consistent with an oval cell origin. Whereas definitive proof of an oval cell origin of hepatocellular has yet to be presented, the current circumstantial evidence justifies continued research on this subject.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/biosíntesis , Células Madre Neoplásicas/metabolismo , Animales , Carcinógenos/toxicidad , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Proteínas de Neoplasias/genética , Trasplante de Neoplasias/métodos , Células Madre Neoplásicas/patología , Fenotipo
20.
World J Biol Chem ; 6(4): 379-88, 2015 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-26629321

RESUMEN

AIM: To study the binding of connective tissue growth factor (CTGF) to cystine knot-containing ligands and how this impacts platelet-derived growth factor (PDGF)-B signaling. METHODS: The binding strengths of CTGF to cystine knot-containing growth factors including vascular endothelial growth factor (VEGF)-A, PDGF-B, bone morphogenetic protein (BMP)-4, and transforming growth factor (TGF)-ß1 were compared using the LexA-based yeast two-hybrid system. EYG48 reporter strain that carried a wild-type LEU2 gene under the control of LexA operators and a lacZ reporter plasmid (p80p-lacZ) containing eight high affinity LexA binding sites were used in the yeast two-hybrid analysis. Interactions between CTGF and the tested growth factors were evaluated based on growth of transformed yeast cells on selective media and colorimetric detection in a liquid ß-galactosidase activity assay. Dissociation constants of CTGF to VEGF-A isoform 165 or PDGF-BB homo-dimer were measured in surface plasma resonance (SPR) analysis. CTGF regulation in PDGF-B presentation to the PDGF receptor ß (PDGFRß) was also quantitatively assessed by the SPR analysis. Combinational effects of CTGF protein and PDGF-BB on activation of PDGFRß and downstream signaling molecules ERK1/2 and AKT were assessed in rabbit corneal fibroblast cells by Western analysis. RESULTS: In the LexA-based yeast two-hybrid system, cystine knot motifs of tested growth factors were fused to the activation domain of the transcriptional factor GAL4 while CTGF was fused to the DNA binding domain of the bacterial repressor protein LexA. Yeast co-transformants containing corresponding fusion proteins for CTGF and all four tested cystine knot motifs survived on selective medium containing galactose and raffinose but lacking histidine, tryptophan, and uracil. In liquid ß-galactosidase assays, CTGF expressing cells that were co-transformed with the cystine knot of VEGF-A had the highest activity, at 29.88 ± 0.91 fold above controls (P < 0.01). Cells containing the cystine knot of BMP-4 expressed the second most activity, with a 24.77 ± 0.47 fold increase (P < 0.01). Cells that contained the cystine knot of TGF-ß1 had a 3.80 ± 0.66 fold increase (P < 0.05) and the ones with the cystine knot of PDGF-B had a 2.64 ± 0.33 fold increase of ß-galactosidase activity (P < 0.01). Further SPR analysis showed that the association rate between VEGF-A 165 and CTGF was faster than PDGF-BB and CTGF. The calculated dissociation constant (KD) of CTGF to VEGF165 and PDGF-BB was 1.8 and 43 nmol/L respectively. PDGF-BB ligand and PDGFRß receptor formed a stable complex with a low dissociation constant 1.4 nmol/L. Increasing the concentration of CTGF up to 263.2 nmol/L significantly the ligand/receptor binding. In addition, CTGF potentiated phosphorylation of PDGFRß and AKT in rabbit corneal fibroblast cells stimulated by PDGF-BB in tissue culture condition. In contrast, CTGF did not affect PDGF-B induced phosphorylation of ERK1/2. CONCLUSION: CTGF has a differential binding affinity to VEGF-A, PDGF-B, BMP-4, and TGF-ß. Its weak association with PDGF-B may represent a novel mechanism to enhance PDGF-B signaling.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA