Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Pathol ; 189(3): 590-603, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30610845

RESUMO

Exposure of mice to a diet containing 3,5-diethoxycarbonyl-1, 4-dihydrocollidine (DDC) induces porphyrin accumulation, cholestasis, immune response, and hepatobiliary damage mimicking hepatic porphyria and sclerosing cholangitis. Although ß-catenin signaling promotes hepatocyte proliferation, and macrophages are a source of Wnts, the role of macrophage-derived Wnts in modulating hepatobiliary injury/repair remains unresolved. We investigated the effect of macrophage-specific deletion of Wntless, a cargo protein critical for cellular Wnt secretion, by feeding macrophage-Wntless-knockout (Mac-KO) and wild-type littermates a DDC diet for 14 days. DDC exposure induced Wnt11 up-regulation in macrophages. Mac-KO mice on DDC showed increased serum alkaline phosphatase, aspartate aminotransferase, direct bilirubin, and histologic evidence of more cell death, inflammation, and ductular reaction. There was impaired hepatocyte proliferation evidenced by Ki-67 immunostaining, which was associated with decreased hepatocyte ß-catenin activation and cyclin-D1 in Mac-KO. Mac-KO also showed increased CD45, F4/80, and neutrophil infiltration after DDC diet, along with increased expression of several proinflammatory cytokines and chemokines. Gene expression analyses of bone marrow-derived macrophages from Mac-KO mice and F4/80+ macrophages isolated from DDC-fed Mac-KO livers showed proinflammatory M1 polarization. In conclusion, this study shows that a lack of macrophage Wnt secretion leads to more DDC-induced hepatic injury due to impaired hepatocyte proliferation and increased M1 macrophages, which promotes immune-mediated cell injury.


Assuntos
Colangite Esclerosante/metabolismo , Colestase/metabolismo , Dieta/efeitos adversos , Hepatócitos/metabolismo , Macrófagos/metabolismo , Piridinas/toxicidade , Proteínas Wnt/biossíntese , Animais , Colangite Esclerosante/induzido quimicamente , Colangite Esclerosante/genética , Colangite Esclerosante/patologia , Colestase/induzido quimicamente , Colestase/genética , Colestase/patologia , Hepatócitos/patologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Regulação para Cima/efeitos dos fármacos , Proteínas Wnt/genética
2.
Am J Pathol ; 189(5): 1091-1104, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30794807

RESUMO

Hepatoblastoma (HB), the most common pediatric primary liver neoplasm, shows nuclear localization of ß-catenin and yes-associated protein 1 (YAP1) in almost 80% of the cases. Co-expression of constitutively active S127A-YAP1 and ΔN90 deletion-mutant ß-catenin (YAP1-ΔN90-ß-catenin) causes HB in mice. Because heterogeneity in downstream signaling is being identified owing to mutational differences even in the ß-catenin gene alone, we investigated if co-expression of point mutants of ß-catenin (S33Y or S45Y) with S127A-YAP1 led to similar tumors as YAP1-ΔN90-ß-catenin. Co-expression of S33Y/S45Y-ß-catenin and S127A-YAP1 led to activation of Yap and Wnt signaling and development of HB, with 100% mortality by 13 to 14 weeks. Co-expression with YAP1-S45Y/S33Y-ß-catenin of the dominant-negative T-cell factor 4 or dominant-negative transcriptional enhanced associate domain 2, the respective surrogate transcription factors, prevented HB development. Although histologically similar, HB in YAP1-S45Y/S33Y-ß-catenin, unlike YAP1-ΔN90-ß-catenin HB, was glutamine synthetase (GS) positive. However, both ΔN90-ß-catenin and point-mutant ß-catenin comparably induced GS-luciferase reporter in vitro. Finally, using a previously reported 16-gene signature, it was shown that YAP1-ΔN90-ß-catenin HB tumors exhibited genetic similarities with more proliferative, less differentiated, GS-negative HB patient tumors, whereas YAP1-S33Y/S45Y-ß-catenin HB exhibited heterogeneity and clustered with both well-differentiated GS-positive and proliferative GS-negative patient tumors. Thus, we demonstrate that ß-catenin point mutants can also collaborate with YAP1 in HB development, albeit with a distinct molecular profile from the deletion mutant, which may have implications in both biology and therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Mutação , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Prognóstico , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Proteínas de Sinalização YAP , beta Catenina/genética
3.
Am J Pathol ; 188(8): 1895-1909, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29920228

RESUMO

Hepatoblastoma (HB) is the most common pediatric liver malignant tumor. Previously, we reported co-activation of ß-catenin and Yes-associated protein-1 (YAP1) in 80% of HB. Hepatic co-expression of active ß-catenin and YAP1 via sleeping beauty transposon/transposase and hydrodynamic tail vein injection led to HB development in mice. Here, we identify lipocalin 2 (Lcn2) as a target of ß-catenin and YAP1 in HB and show that serum Lcn2 values positively correlated with tumor burden. Lcn2 was strongly expressed in HB tumor cells in our mouse model. A tissue array of 62 HB cases showed highest LCN2 expression in embryonal and lowest in fetal, blastemal, and small cell undifferentiated forms of HB. Knockdown of LCN2 in HB cells had no effect on cell proliferation but reduced NF-κB reporter activity. Next, liver-specific Lcn2 knockout (KO) mice were generated. No difference in tumor burden was observed between Lcn2 KO mice and wild-type littermate controls after sleeping beauty transposon/transposase and hydrodynamic tail vein injection delivery of active YAP1 and ß-catenin, although Lcn2 KO mice with HB lacked any serum Lcn2 elevation, demonstrating that transformed hepatocytes are the source of serum Lcn2. More blastemal areas and inflammation were observed within HB in Lcn2 KO compared with wild-type tumors. In conclusion, Lcn2 expressed in hepatocytes appears to be dispensable for the pathogenesis of HB. However, transformed hepatocytes secrete serum Lcn2, making Lcn2 a valuable biomarker for HB.


Assuntos
Biomarcadores Tumorais/sangue , Hepatoblastoma/patologia , Hepatócitos/patologia , Lipocalina-2/sangue , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Proliferação de Células , Hepatoblastoma/sangue , Hepatócitos/metabolismo , Humanos , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição , Carga Tumoral , Proteínas de Sinalização YAP
4.
Gene Expr ; 18(3): 209-219, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-29519268

RESUMO

Activation of the Wnt/ß-catenin signaling is reported in large subsets of hepatocellular carcinoma (HCC). Upregulation of Wnt genes is one contributing mechanism. In the current study, we sought to address the role of hepatocyte-derived Wnts in a model of hepatic injury, fibrosis, and carcinogenesis. We subjected hepatocyte-specific Wntless knockout mice (HP-KO), unable to secrete Wnts from hepatocytes, and littermate controls (HP-CON) to diethylnitrosamine and carbon tetrachloride (DEN/CCl4) and harvested at 3, 5, and 6 months for histological and molecular analysis. Analysis at 5 months displayed increased hepatic expression of several Wnts and upregulation of some, but not all, ß-catenin targets, without mutations in Ctnnb1. At 5 months, HP-CON and HP-KO had comparable tumor burden and injury; however, HP-KO uniquely showed small CK19+ foci within tumors. At 6 months, both groups were moribund with comparable tumor burden and CK19 positivity. While HCC histology was indistinguishable between the groups, HP-KO exhibited increased active ß-catenin and decreased c-Myc, Brd4, E-cadherin, and others. Hepatic injury, inflammation, and fibrosis were also indistinguishable at 3 months between both groups. Thus, lack of Wnt secretion from hepatocytes did not affect overall injury, fibrosis, or HCC burden, although there were protein expression differences in the tumors occurring in the two groups.


Assuntos
Carcinoma Hepatocelular/metabolismo , Hepatócitos/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Wnt/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Tetracloreto de Carbono/toxicidade , Carcinoma Hepatocelular/etiologia , Dietilnitrosamina/toxicidade , Neoplasias Hepáticas/etiologia , Masculino , Camundongos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
5.
Semin Liver Dis ; 37(2): 141-151, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28564722

RESUMO

Liver possesses many critical functions such as synthesis, detoxification, and metabolism. It continually receives nutrient-rich blood from gut, which incidentally is also toxin-rich. That may be why liver is uniquely bestowed with a capacity to regenerate. A commonly studied procedure to understand the cellular and molecular basis of liver regeneration is that of surgical resection. Removal of two-thirds of the liver in rodents or patients instigates alterations in hepatic homeostasis, which are sensed by the deficient organ to drive the restoration process. Although the exact mechanisms that initiate regeneration are unknown, alterations in hemodynamics and metabolism have been suspected as important effectors. Key signaling pathways are activated that drive cell proliferation in various hepatic cell types through autocrine and paracrine mechanisms. Once the prehepatectomy mass is regained, the process of regeneration is adequately terminated. This review highlights recent discoveries in the cellular and molecular basis of liver regeneration.


Assuntos
Proliferação de Células , Hepatectomia , Regeneração Hepática , Fígado/fisiopatologia , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/cirurgia , Transdução de Sinais
6.
J Hepatol ; 67(2): 360-369, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28341391

RESUMO

BACKGROUND & AIMS: Iron overload disorders such as hereditary hemochromatosis and iron loading anemias are a common cause of morbidity from liver diseases and increase risk of hepatic fibrosis and hepatocellular carcinoma (HCC). Treatment options for iron-induced damage are limited, partly because there is lack of animal models of human disease. Therefore, we investigated the effect of iron overload in liver-specific ß-catenin knockout mice (KO), which are susceptible to injury, fibrosis and tumorigenesis following chemical carcinogen exposure. METHODS: Iron overload diet was administered to KO and littermate control (CON) mice for various times. To ameliorate an oxidant-mediated component of tissue injury, N-Acetyl-L-(+)-cysteine (NAC) was added to drinking water of mice on iron overload diet. RESULTS: KO on iron diet (KO +Fe) exhibited remarkable inflammation, followed by steatosis, oxidative stress, fibrosis, regenerating nodules and occurrence of occasional HCC. Increased injury in KO +Fe was associated with activated protein kinase B (AKT), ERK, and NF-κB, along with reappearance of ß-catenin and target gene Cyp2e1, which promoted lipid peroxidation and hepatic damage. Addition of NAC to drinking water protected KO +Fe from hepatic steatosis, injury and fibrosis, and prevented activation of AKT, ERK, NF-κB and reappearance of ß-catenin. CONCLUSIONS: The absence of hepatic ß-catenin predisposes mice to hepatic injury and fibrosis following iron overload, which was reminiscent of hemochromatosis and associated with enhanced steatohepatitis and fibrosis. Disease progression was notably alleviated by antioxidant therapy, which supports its chemopreventive role in the management of chronic iron overload disorders. LAY SUMMARY: Lack of animal models for iron overload disorders makes it hard to study the disease process for improving therapies. Feeding high iron diet to mice that lack the ß-catenin gene in liver cells led to increased inflammation followed by fat accumulation, cell death and wound healing that mimicked human disease. Administration of an antioxidant prevented hepatic injury in this model.


Assuntos
Fígado Gorduroso/etiologia , Fígado Gorduroso/metabolismo , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/metabolismo , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , beta Catenina/deficiência , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Modelos Animais de Doenças , Fígado Gorduroso/prevenção & controle , Feminino , Hemocromatose/complicações , Hemocromatose/metabolismo , Humanos , Sobrecarga de Ferro/tratamento farmacológico , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/prevenção & controle , Masculino , Camundongos , Camundongos Knockout , Estresse Oxidativo , Transdução de Sinais , beta Catenina/genética
7.
Gene Expr ; 17(3): 219-235, 2017 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-28474571

RESUMO

An important role for ß-catenin in regulating p65 (a subunit of NF-κB) during acute liver injury has recently been elucidated through use of conditional ß-catenin knockout mice, which show protection from apoptosis through increased activation of p65. Thus, we hypothesized that the p65/ß-catenin complex may play a role in regulating processes such as cell proliferation during liver regeneration. We show through in vitro and in vivo studies that the p65/ß-catenin complex is regulated through the TNF-α pathway and not through Wnt signaling. However, this complex is unchanged after partial hepatectomy (PH), despite increased p65 and ß-catenin nuclear translocation as well as cyclin D1 activation. We demonstrate through both in vitro silencing experiments and chromatin immunoprecipitation after PH that ß-catenin, and not p65, regulates cyclin D1 expression. Conversely, using reporter mice we show p65 is activated exclusively in the nonparenchymal (NPC) compartment during liver regeneration. Furthermore, stimulation of macrophages by TNF-α induces activation of NF-κB and subsequent secretion of Wnts essential for ß-catenin activation in hepatocytes. Thus, we show that ß-catenin and p65 are activated in separate cellular compartments during liver regeneration, with p65 activity in NPCs contributing to the activation of hepatocyte ß-catenin, cyclin D1 expression, and subsequent proliferation.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Regulação da Expressão Gênica , Regeneração Hepática , Fígado/metabolismo , Fator de Transcrição RelA/metabolismo , beta Catenina/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/metabolismo , Hepatectomia , Hepatócitos/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Células RAW 264.7 , Fator de Transcrição RelA/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/genética
8.
Am J Pathol ; 185(8): 2194-205, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26100214

RESUMO

Activation of Wnt/ß-catenin signaling during liver regeneration (LR) after partial hepatectomy (PH) is observed in several species. However, how this pathway is turned off when hepatocyte proliferation is no longer required is unknown. We assessed LR in liver-specific knockouts of Wntless (Wls-LKO), a protein required for Wnt secretion from a cell. When subjected to PH, Wls-LKO showed prolongation of hepatocyte proliferation for up to 4 days compared with littermate controls. This coincided with increased ß-catenin-T-cell factor 4 interaction and cyclin-D1 expression. Wls-LKO showed decreased expression and secretion of inhibitory Wnt5a during LR. Wnt5a expression increased between 24 and 48 hours, and Frizzled-2 between 24 and 72 hours, after PH in normal mice. Treatment of primary mouse hepatocytes and liver tumor cells with Wnt5a led to a notable decrease in ß-catenin-T-cell factor activity, cyclin-D1 expression, and cell proliferation. Intriguingly, Wnt5a-LKO did not display any prolongation of LR because of compensation by other cells. In addition, Wnt5a-LKO hepatocytes failed to respond to exogenous Wnt5a treatment in culture because of a compensatory decrease in Frizzled-2 expression. In conclusion, we demonstrate Wnt5a to be, by default, a negative regulator of ß-catenin signaling and hepatocyte proliferation, both in vitro and in vivo. We also provide evidence that the Wnt5a/Frizzled-2 axis suppresses ß-catenin signaling in hepatocytes in an autocrine manner, thereby contributing to timely conclusion of the LR process.


Assuntos
Proliferação de Células/fisiologia , Hepatócitos/metabolismo , Regeneração Hepática/fisiologia , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Regeneração Hepática/efeitos dos fármacos , Camundongos , Camundongos Knockout , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt-5a
9.
Gene Expr ; 17(1): 19-34, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27226410

RESUMO

Triiodothyronine (T3) induces hepatocyte proliferation in rodents. Recent work has shown molecular mechanism for T3's mitogenic effect to be through activation of ß-catenin signaling. Since systemic side effects of T3 may preclude its clinical use, and hepatocytes mostly express T3 hormone receptor ß (TRß), we investigated if selective TRß agonists like GC-1 may also have ß-catenin-dependent hepatocyte mitogenic effects. Here we studied the effect of GC-1 and T3 in conditional knockouts of various Wnt pathway components. We also assessed any regenerative advantage of T3 or GC-1 when given prior to partial hepatectomy in mice. Mice administered GC-1 showed increased pSer675-ß-catenin, cyclin D1, BrdU incorporation, and PCNA. No abnormalities in liver function tests were noted. GC-1-injected liver-specific ß-catenin knockouts (ß-catenin LKO) showed decreased proliferation when compared to wild-type littermates. To address if Wnt signaling was required for T3- or GC-1-mediated hepatocyte proliferation, we used LRP5-6-LKO, which lacks the two redundant Wnt coreceptors. Surprisingly, decreased hepatocyte proliferation was also evident in LRP5-6-LKO in response to T3 and GC-1, despite increased pSer675-ß-catenin. Further, increased levels of active ß-catenin (hypophosphorylated at Ser33, Ser37, and Thr41) were evident after T3 and GC-1 treatment. Finally, mice pretreated with T3 or GC-1 for 7 days followed by partial hepatectomy showed a significant increase in hepatocyte proliferation both at the time (T0) and 24 h after surgery. In conclusion, like T3, TRß-selective agonists induce hepatocyte proliferation through ß-catenin activation via both PKA- and Wnt-dependent mechanisms and confer a regenerative advantage following surgical resection. Hence, these agents may be useful regenerative therapies in liver transplantation or other surgical settings.


Assuntos
Acetatos/farmacologia , Proliferação de Células/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Regeneração Hepática/efeitos dos fármacos , Fenóis/farmacologia , Receptores beta dos Hormônios Tireóideos/agonistas , Tri-Iodotironina/farmacologia , beta Catenina/metabolismo , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hepatectomia/métodos , Hepatócitos/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores beta dos Hormônios Tireóideos/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
10.
J Hepatol ; 62(2): 380-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25457204

RESUMO

BACKGROUND & AIMS: Hepatocellular cancer (HCC) remains a disease of poor prognosis, highlighting the relevance of elucidating key molecular aberrations that may be targeted for novel therapies. Wnt signalling activation, chiefly due to mutations in CTNNB1, have been identified in a major subset of HCC patients. While several in vitro proof of concept studies show the relevance of suppressing Wnt/ß-catenin signalling in HCC cells or tumour xenograft models, no study has addressed the impact of ß-catenin inhibition in a relevant murine HCC model driven by Ctnnb1 mutations. METHODS: We studied the in vivo impact of ß-catenin suppression by locked nucleic acid (LNA) antisense treatment, after establishing Ctnnb1 mutation-driven HCC by diethylnitrosamine and phenobarbital (DEN/PB) administration. RESULTS: The efficacy of LNA directed against ß-catenin vs. scrambled on Wnt signalling was demonstrated in vitro in HCC cells and in vivo in normal mice. The DEN/PB model leads to HCC with Ctnnb1 mutations. A complete therapeutic response in the form of abrogation of HCC was observed after ten treatments of tumour-bearing mice with ß-catenin LNA every 48h as compared to the scrambled control. A decrease in ß-catenin activity, cell proliferation and increased cell death was evident after ß-catenin suppression. No effect of ß-catenin suppression was evident in non-Ctnnb1 mutated HCC, observed after DEN-only administration. CONCLUSIONS: Thus, we provide the in vivo proof of concept that ß-catenin suppression in HCC will be of significant therapeutic benefit, provided the tumours display Wnt activation via mechanisms like CTNNB1 mutations.


Assuntos
Carcinogênese/genética , Carcinoma Hepatocelular/genética , DNA de Neoplasias/genética , Neoplasias Hepáticas Experimentais/genética , Mutação , Oligonucleotídeos/metabolismo , beta Catenina/genética , Alquilantes/uso terapêutico , Animais , Western Blotting , Carcinogênese/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Análise Mutacional de DNA , Dietilnitrosamina/farmacologia , Ensaio de Imunoadsorção Enzimática , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Imuno-Histoquímica , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Fenobarbital/farmacologia , Células Tumorais Cultivadas , beta Catenina/efeitos dos fármacos , beta Catenina/metabolismo
12.
Gene Expr ; 19(2): 121-136, 2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30236172

RESUMO

Alterations in the Wnt signaling pathway including those impacting hepatic stellate cells (HSCs) have been implicated in liver fibrosis. In the current study, we first examined the expression of Wnt genes in human HSC (HHSCs) after treatment with a profibrogenic factor TGF-ß1. Next, we generated HSC-specific Wntless (Wls) knockout (KO) using the Lrat-cre and Wls-floxed mice. KO and littermate controls (CON) were characterized for any basal phenotype and subjected to two liver fibrosis protocols. In vitro, TGF-ß1 induced expression of Wnt2, 5a and 9a while decreasing Wnt2b, 3a, 4, and 11 in HHSC. In vivo, KO and CON mice were born at normal Mendelian ratio and lacked any overt phenotype. Loss of Wnt secretion from HSCs had no effect on liver weight and did not impact ß-catenin activation in the pericentral hepatocytes. After 7 days of bile duct ligation (BDL), KO and CON showed comparable levels of serum alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, total and direct bilirubin. Comparable histology, Sirius red staining, and immunohistochemistry for α-SMA, desmin, Ki-67, F4/80, and CD45 indicated similar proliferation, inflammation, and portal fibrosis in both groups. Biweekly administration of carbon tetrachloride for 4 or 8 weeks also led to comparable serum biochemistry, inflammation, and fibrosis in KO and CON. Specific Wnt genes were altered in HHSCs in response to TGF-ß1; however, eliminating Wnt secretion from HSC did not impact basal ß-catenin activation in normal liver nor did it alter the injury-repair response during development of liver fibrosis.


Assuntos
Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Sistema Biliar/lesões , Sistema Biliar/metabolismo , Células Cultivadas , Feminino , Células Estreladas do Fígado/efeitos dos fármacos , Humanos , Fígado/lesões , Fígado/patologia , Cirrose Hepática/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Crescimento Transformador beta1/farmacologia , Proteínas Wnt/genética , beta Catenina/metabolismo
13.
Hepatol Commun ; 2(7): 845-860, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30027142

RESUMO

ß-Catenin in hepatocytes, under the control of Wnts, regulates pericentral gene expression. It also contributes to liver regeneration (LR) after partial hepatectomy (PH) by regulating cyclin-D1 gene expression as shown in the ß-catenin and Wnt coreceptors low-density lipoprotein receptor-related protein 5/6 conditional knockouts (KO). However, conditional deletion of Wntless (Wls), required for Wnt secretion, in hepatocytes, cholangiocytes, or macrophages lacked any impact on zonation, while Wls deletion in macrophages only marginally affected LR. Here, we address the contribution of hepatic endothelial cells (ECs) in zonation and LR by characterizing EC-Wls-KO generated by interbreeding Wls-floxed and lymphatic vessel endothelial hyaluronan receptor (Lyve1)-cre mice. These mice were also used to study LR after PH. While Lyve1 expression in adult liver is limited to sinusoidal ECs only, Lyve1-cre mice bred to ROSA26-Stopflox/flox-enhanced yellow fluorescent protein (EYFP) mice showed EYFP labeling in sinusoidal and central vein ECs. EC-Wls-KO mice showed decreased liver weights; lacked glutamine synthetase, cytochrome P450 2e1, and cytochrome P450 1a2; and were resistant to acetaminophen-induced liver injury. After PH, EC-Wls-KO showed quantitative and qualitative differences in cyclin-D1 expression at 24-72 hours, which led to a lower hepatocyte proliferation at 40 hours but a rebound increase by 72 hours. ECs and macrophages isolated from regenerating livers at 12 hours showed significant up-regulation of Wnt2 and Wnt9b messenger RNA; these are the same two Wnts involved in baseline ß-catenin activity in pericentral hepatocytes. Conclusion: At baseline, ECs secrete Wnt proteins essential for ß-catenin activation in pericentral hepatocytes. During LR, sinusoidal and central vein ECs and secondarily macrophages secrete Wnt2, while predominantly central vein ECs and secondarily macrophages are the likely source of Wnt9b. This process spatiotemporally regulates ß-catenin activation in hepatocytes to induce cell proliferation. (Hepatology Communications 2018;2:845-860).

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA