Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.984
Filtrar
1.
J Gastroenterol Hepatol ; 39(2): 224-230, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37939704

RESUMO

Liver ischemia/reperfusion injury (IRI) is a major complication after partial hepatectomy and liver transplantation and during hypovolemic shock and hypoxia-related diseases. Liver IRI is a current research hotspot. The early stage of liver IRI is characterized by injury and dysfunction of liver sinusoidal endothelial cells (LSECs), which, along with hepatocytes, are the major cells involved in liver injury. In this review, we elaborate on the roles played by LSECs in liver IRI, including the pathological features of LSECs, LSECs exacerbation of the sterile inflammatory response, LSECs interactions with platelets and the promotion of liver regeneration, and the activation of LSECs autophagy. In addition, we discuss the study of LSECs as therapeutic targets for the treatment of liver IRI and the existing problems when applying LSECs in liver IRI research.


Assuntos
Células Endoteliais , Traumatismo por Reperfusão , Humanos , Células Endoteliais/fisiologia , Fígado/patologia , Hepatócitos/fisiologia , Traumatismo por Reperfusão/patologia , Isquemia/patologia
2.
J Hepatol ; 80(2): 309-321, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37918568

RESUMO

BACKGROUND & AIMS: Post-hepatectomy liver failure (PHLF) leads to poor prognosis in patients undergoing hepatectomy, with hepatic vascular reconstitution playing a critical role. However, the regulators of hepatic vascular reconstitution remain unclear. In this study, we aimed to investigate the regulatory mechanisms of hepatic vascular reconstitution and identify biomarkers predicting PHLF in patients undergoing hepatectomy. METHODS: Candidate genes that were associated with hepatic vascular reconstitution were screened using adeno-associated virus vectors in Alb-Cre-CRISPR/Cas9 mice subjected to partial hepatectomy. The biological activities of candidate genes were estimated using endothelial precursor transfusion and associating liver partition and portal vein ligation for staged hepatectomy (ALPPS) models. The level of candidates was detected in biopsies from patients undergoing ALPPS. Risk factors for PHLF were also screened using retrospective data. RESULTS: Downregulation of Gata3 and upregulation of Ramp2 in hepatocytes promoted the proliferation of liver sinusoidal endothelial cells and hepatic revascularization. Pigment epithelium-derived factor (PEDF) and vascular endothelial growth factor A (VEGFA) played opposite roles in regulating the migration of endothelial precursors from bone marrow and the formation of new sinusoids after hepatectomy. Gata3 restricted endothelial cell function in patient-derived hepatic organoids, which was abrogated by a Gata3 inhibitor. Moreover, overexpression of Gata3 led to higher mortality in ALPPS mice, which was improved by a PEDF-neutralizing antibody. The expression of Gata3/RAMP and PEDF/VEGFA tended to have a negative correlation in patients undergoing ALPPS. A nomogram incorporating multiple factors, such as serum PEDF/VEGF index, was constructed and could efficiently predict the risk of PHLF. CONCLUSIONS: The balance of Gata3 and Ramp2 in hepatocytes regulates the proliferation of liver sinusoidal endothelial cells and hepatic revascularization via changes in the expression of PEDF and VEGFA, revealing potential targets for the prevention and treatment of PHLF. IMPACT AND IMPLICATIONS: In this study, we show that the balance of Gata3 and Ramp2 in hepatocytes regulates hepatic vascular reconstitution by promoting a shift from pigment epithelium-derived factor (PEDF) to vascular endothelial growth factor A (VEGFA) expression during hepatectomy- or ALLPS (associating liver partition and portal vein ligation for staged hepatectomy)-induced liver regeneration. We also identified serum PEDF/VEGFA index as a potential predictor of post-hepatectomy liver failure in patients who underwent hepatectomy. This study improves our understanding of how hepatocytes contribute to liver regeneration and provides new targets for the prevention and treatment of post-hepatectomy liver failure.


Assuntos
Falência Hepática , Neoplasias Hepáticas , Humanos , Camundongos , Animais , Regeneração Hepática/fisiologia , Fator A de Crescimento do Endotélio Vascular , Estudos Retrospectivos , Células Endoteliais , Fígado/cirurgia , Hepatectomia/efeitos adversos , Hepatócitos/fisiologia , Veia Porta/cirurgia , Falência Hepática/etiologia , Ligadura , Fator de Transcrição GATA3 , Proteína 2 Modificadora da Atividade de Receptores
3.
Stem Cells Transl Med ; 13(3): 204-218, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38103170

RESUMO

Orthotopic liver transplantation (OLT) is the current standard of care for both chronic and acute terminal liver disease. However, a major limitation of this treatment is the shortage of healthy donor organs and the need for life-long immunosuppression to prevent graft rejection. Hepatocyte transplantation (HTx) has emerged as a promising, alternative therapeutic approach to either replace OLT or to act as a bridge until a donor liver becomes available thus reducing waiting list mortality. HTx involves the infusion and engraftment of human hepatocytes, typically isolated from organs unsuitable for OLT, into recipient liver parenchyma to carry out the missing hepatic function of the native cells. HTx is less invasive than OLT and can be performed repeatedly if required. The safety of clinical HTx has been shown and treatment results are promising, especially in patients with liver-based metabolic disorders. Nevertheless, HTx has failed to become the standard of care treatment for such disorders. This review aims to evaluate the progress that has been made within the field of HTx over the last 30 years and identify potential shortcomings within the approach which may be hindering its routine clinical application.


Assuntos
Hepatopatias , Transplante de Fígado , Humanos , Doadores Vivos , Hepatopatias/terapia , Hepatócitos/fisiologia
4.
Cell Prolif ; 56(5): e13467, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37199059

RESUMO

Ex vivo gene manipulation in human hepatocytes is a promising therapeutic strategy in the treatment of inherited liver diseases. However, a major limitation is the lack of a highly efficient and safe genetic manipulation system for transplantable primary human hepatocytes (PHHs). Here, we reported that proliferating human hepatocytes (ProliHHs) cultured in vitro showed high susceptibility to lentivirus-mediated genetic modification and maintained cellular phenotypes after lentiviral infection. Human factor VIII expression was introduced through F8-Lentivirus-mediated transduction of ProliHHs followed by xenotransplantation into immunocompromised haemophilia A mice. We demonstrated that these F8-modified ProliHHs could effectively repopulate the mouse liver, resulting in therapeutic benefits in mouse models. Furthermore, no genotoxicity was detected in F8-modified ProliHHs using lentiviral integration site analysis. Thus, this study demonstrated, for the first time, the feasibility and safety of lentiviral modification in ProliHHs to induce the expression of coagulation factor VIII in the treatment of haemophilia A.


Assuntos
Fator VIII , Hemofilia A , Humanos , Camundongos , Animais , Fator VIII/genética , Hemofilia A/genética , Hemofilia A/terapia , Vetores Genéticos , Hepatócitos/fisiologia , Fígado
5.
J Hepatol ; 79(2): 417-432, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37088309

RESUMO

BACKGROUND & AIMS: While normal human liver is thought to be generally quiescent, clonal hepatocyte expansions have been observed, though neither their cellular source nor their expansion dynamics have been determined. Knowing the hepatocyte cell of origin, and their subsequent dynamics and trajectory within the human liver will provide an important basis to understand disease-associated dysregulation. METHODS: Herein, we use in vivo lineage tracing and methylation sequence analysis to demonstrate normal human hepatocyte ancestry. We exploit next-generation mitochondrial sequencing to determine hepatocyte clonal expansion dynamics across spatially distinct areas of laser-captured, microdissected, clones, in tandem with computational modelling in morphologically normal human liver. RESULTS: Hepatocyte clones and rare SOX9+ hepatocyte progenitors commonly associate with portal tracts and we present evidence that clones can lineage-trace with cholangiocytes, indicating the presence of a bipotential common ancestor at this niche. Within clones, we demonstrate methylation CpG sequence diversity patterns indicative of periportal not pericentral ancestral origins, indicating a portal to central vein expansion trajectory. Using spatial analysis of mitochondrial DNA variants by next-generation sequencing coupled with mathematical modelling and Bayesian inference across the portal-central axis, we demonstrate that patterns of mitochondrial DNA variants reveal large numbers of spatially restricted mutations in conjunction with limited numbers of clonal mutations. CONCLUSIONS: These datasets support the existence of a periportal progenitor niche and indicate that clonal patches exhibit punctuated but slow growth, then quiesce, likely due to acute environmental stimuli. These findings crucially contribute to our understanding of hepatocyte dynamics in the normal human liver. IMPACT AND IMPLICATIONS: The liver is mainly composed of hepatocytes, but we know little regarding the source of these cells or how they multiply over time within the disease-free human liver. In this study, we determine a source of new hepatocytes by combining many different lab-based methods and computational predictions to show that hepatocytes share a common cell of origin with bile ducts. Both our experimental and computational data also demonstrate hepatocyte clones are likely to expand in slow waves across the liver in a specific trajectory, but often lie dormant for many years. These data show for the first time the expansion dynamics of hepatocytes in normal liver and their cell of origin enabling the accurate measurment of changes to their dynamics that may lead to liver disease. These findings are important for researchers determining cancer risk in human liver.


Assuntos
Hepatopatias , Nicho de Células-Tronco , Humanos , Teorema de Bayes , Diferenciação Celular , Hepatócitos/fisiologia , Fígado , DNA Mitocondrial
6.
Transplant Proc ; 55(4): 1012-1015, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37100736

RESUMO

BACKGROUND: Hepatocyte transplantation has been reported to be useful for metabolic diseases and acute liver failure. However, the shortage of donors limits its widespread use. The use of livers from donors after circulatory death, which are currently unavailable for liver transplantation, may alleviate donor shortage. In this study, we investigated the effects of mechanical perfusion on cardiac arrest hepatocytes in a rat model using cardiac arrest donor livers, and we evaluated the function of cardiac arrest hepatocytes. METHODS: F344 rat hepatocytes isolated from livers removed during cardiac pulsation were compared with those isolated from livers removed after 30 minutes of warm ischemia after cardiac arrest. We then compared hepatocytes isolated from livers removed after 30 minutes of warm ischemia with those isolated after 30 minutes of mechanical perfusion before isolation. The yield per liver weight, ammonia removal capacity, and adenosine diphosphate/adenosine triphosphate ratio were evaluated. RESULTS: Thirty minutes of warm inhibition reduced hepatocyte yield but did not alter ammonia removal capacity and energy status. Mechanical perfusion increased hepatocyte yield and improved the adenosine diphosphate/adenosine triphosphate ratio after 30 minutes of warm inhibition. CONCLUSION: Thirty minutes of warm ischemic time may decrease isolated hepatocyte yield without degrading their function. If increased yields are obtained, livers from donors dying of cardiac arrest could be used for hepatocyte transplantation. The results also suggest that mechanical perfusion may positively affect the energy status of hepatocytes.


Assuntos
Amônia , Parada Cardíaca , Ratos , Animais , Ratos Endogâmicos F344 , Hepatócitos/fisiologia , Fígado/metabolismo , Perfusão/métodos , Isquemia Quente/efeitos adversos , Trifosfato de Adenosina/metabolismo , Difosfato de Adenosina/metabolismo , Preservação de Órgãos/métodos
7.
Cells ; 12(7)2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-37048049

RESUMO

Acellular liver scaffolds (ALS) produced by decellularization have been successfully explored for distinct regenerative purposes. To date, it is unknown whether transplanted ALSs are affected by cirrhotic livers, either becoming cirrhotic themselves or instead remaining as a robust template for healthy cell growth after transplantation into cirrhotic rats. Moreover, little is known about the clinical course of recipient cirrhotic livers after ALS transplantation. To address these questions, we transplanted ALSs into cirrhotic rats previously treated with the granulocyte colony-stimulating factor. Here, we report successful cellular engraftment within the transplanted ALSs at 7, 15, and 30 days after transplantation. Recellularization was orchestrated by liver tissue cell activation, resident hepatocytes and bile duct proliferation, and an immune response mediated by the granulocyte components. Furthermore, we showed that transplanted ALSs ensured a pro-regenerative and anti-inflammatory microenvironment, attracted vessels from the host cirrhotic tissue, and promoted progenitor cell recruitment. ALS transplantation induced cirrhotic liver regeneration and extracellular matrix remodeling. Moreover, the transplanted ALS sustained blood circulation and attenuated alterations in the ultrasonographic and biochemical parameters in cirrhotic rats. Taken together, our results confirm that transplanted ALSs are not affected by cirrhotic livers and remain a robust template for healthy cell growth and stimulated cirrhotic liver regeneration.


Assuntos
Fator Estimulador de Colônias de Granulócitos , Cirrose Hepática , Tecidos Suporte , Animais , Ratos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hepatócitos/fisiologia , Cirrose Hepática/terapia
8.
Hepatology ; 78(1): 167-178, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-36724876

RESUMO

In cases of end-stage liver diseases, the proliferation of existing hepatocytes is compromised, a feature of human chronic liver disease, in which most hepatocytes are dysfunctional. So far, liver transplantation represents the only curative therapeutic solution for advanced liver diseases, and the shortage of donor organs leads to high morbidity and mortality worldwide. The promising treatment is to prompt the biliary epithelial cells (BECs) transdifferentiation. However, the critical factors governing the initiation of BEC-derived liver regeneration are largely unknown. The zebrafish has advantages in large-scale genetic screens to identify the critical factors involved in liver regeneration. Here, we combined N-ethyl-N-nitrosourea screen, positional cloning, transgenic lines, antibody staining, and in situ hybridization methods and identified a liver regeneration defect mutant ( lrd ) using the zebrafish extensive liver injury model. Through positional cloning and genomic sequencing, we mapped the mutation site to rngtt . Loss of rngtt leads to the defects of BEC dedifferentiation, bipotential progenitor cell activation, and cell proliferation in the initiation stage of liver regeneration. The transdifferentiation from BECs to hepatocytes did not occur even at the late stage of liver regeneration. Mechanically, Rngtt transcriptionally regulates the attachment of mRNA cap to mTOR complex 1 (mTORC1) components and dnmt1 to maintain the activation of mTORC1 and DNA methylation in BECs after severe liver injury and prompt BEC to hepatocyte conversion. Furthermore, rptor and dnmt1 mutants displayed the same liver regeneration defects as rngtt mutation. In conclusion, our results suggest Rngtt is a new factor that initiates BEC-derived liver regeneration.


Assuntos
Regeneração Hepática , Peixe-Zebra , Animais , Humanos , DNA (Citosina-5-)-Metiltransferase 1 , Células Epiteliais , Hepatócitos/fisiologia , Fígado , Regeneração Hepática/genética , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas de Peixe-Zebra/genética
9.
Biochim Biophys Acta Rev Cancer ; 1878(3): 188870, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36842766

RESUMO

Given the liver's remarkable and unique regenerative capacity, researchers have long focused on liver progenitor cells (LPCs) and liver cancer stem cells (LCSCs). LPCs can differentiate into both hepatocytes and cholangiocytes. However, the mechanism underlying cell conversion and its distinct contribution to liver homeostasis and tumorigenesis remain unclear. In this review, we discuss the complicated conversions involving LPCs and LCSCs. As the critical intermediate state in malignant transformation, LPCs play double-edged sword roles. LPCs are not only involved in hepatic wound-healing responses by supplementing liver cells and bile duct cells in the damaged liver but may transform into LCSCs under dysregulation of key signaling pathways, resulting in refractory malignant liver tumors. Because LPC lineages are temporally and spatially dynamic, we discuss crucial LPC subgroups and summarize regulatory factors correlating with the trajectories of LPCs and LCSCs in the liver tumor microenvironment. This review elaborates on the double-edged sword roles of LPCs to help understand the liver's regenerative potential and tumor heterogeneity. Understanding the sources and transformations of LPCs is essential in determining how to exploit their regenerative capacity in the future.


Assuntos
Hepatócitos , Neoplasias Hepáticas , Humanos , Diferenciação Celular , Hepatócitos/patologia , Hepatócitos/fisiologia , Neoplasias Hepáticas/patologia , Células-Tronco Neoplásicas/patologia , Transformação Celular Neoplásica , Microambiente Tumoral
10.
J Biol Chem ; 299(3): 102937, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36690274

RESUMO

Defective autophagy and lipotoxicity are the hallmarks of nonalcoholic fatty liver disease. However, the precise molecular mechanism for the defective autophagy in lipotoxic conditions is not fully known. In the current study, we elucidated that activation of the mammalian target of rapamycin complex 1 (mTORC1)-G9a-H3K9me2 axis in fatty acid-induced lipotoxicity blocks autophagy by repressing key autophagy genes. The fatty acid-treated cells show mTORC1 activation, increased histone methyltransferase G9a levels, and suppressed autophagy as indicated by increased accumulation of the key autophagic cargo SQSTM1/p62 and decreased levels of autophagy-related proteins LC3II, Beclin1, and Atg7. Our chromatin immunoprecipitation analysis showed that decrease in autophagy was associated with increased levels of the G9a-mediated repressive H3K9me2 mark and decreased RNA polymerase II occupancy at the promoter regions of Beclin1 and Atg7 in fatty acid-treated cells. Inhibition of mTORC1 in fatty acid-treated cells decreased G9a-mediated H3K9me2 occupancy and increased polymerase II occupancy at Beclin1 and Atg7 promoters. Furthermore, mTORC1 inhibition increased the expression of Beclin1 and Atg7 in fatty acid-treated cells and decreased the accumulation of SQSTM1/p62. Interestingly, the pharmacological inhibition of G9a alone in fatty acid-treated cells decreased the H3K9me2 mark at Atg7 and Beclin1 promoters and restored the expression of Atg7 and Beclin1. Taken together, our findings have identified the mTORC1-G9a-H3K9me2 axis as a negative regulator of the autophagy pathway in hepatocellular lipotoxicity and suggest that the G9a-mediated epigenetic repression is mechanistically a key step during the repression of autophagy in lipotoxic conditions.


Assuntos
Autofagia , Ácidos Graxos , Histona Metiltransferases , Histonas , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Histonas/metabolismo , Ácidos Graxos/toxicidade , Autofagia/fisiologia , Epigênese Genética , Histona Metiltransferases/metabolismo , Hepatócitos/fisiologia , Células Hep G2 , Regulação da Expressão Gênica/efeitos dos fármacos , Palmitatos/toxicidade , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Regiões Promotoras Genéticas , Autofagossomos/genética , Autofagossomos/metabolismo , Humanos
11.
Hepatology ; 78(1): 136-149, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-36631003

RESUMO

BACKGROUND AND AIMS: The liver has the unique ability of regeneration, which is extremely important for restoring homeostasis after liver injury. Although clinical observations have revealed an association between psychological stress and the liver, whether stress has a causal influence on the liver regeneration remains markedly less defined. APPROACH AND RESULTS: Rearing rodents in an enriched environment (EE) can induce eustress or positive psychological stress. Herein, EE-induced eustress was found to significantly enhance the ability of liver regeneration after partial hepatectomy or carbon tetrachloride-induced liver injury based on the more rapid restoration of liver/body weight ratio and the significantly increased number of proliferating hepatocytes in EE mice. Mechanistically, the cytokine array revealed that IL-22 was markedly increased in the regenerating liver in response to EE. Blockade of IL-22 signaling abrogated the enhanced liver regeneration induced by EE. Group 1 innate lymphoid cells (ILCs), including type 1 ILCs (ILC1s), have been identified as the major sources of IL-22 in the regenerating liver. EE housing led to a rapid accumulation of hepatic ILC1s after partial hepatectomy and the EE-induced enhancement of liver regeneration and elevation of IL-22 was nearly eliminated in ILC1-deficient Tbx21-/- mice. Chemical sympathectomy or blockade of ß-adrenergic signaling also abolished the effect of EE on ILC1s and attenuated the enhanced liver regeneration of EE-housed mice. CONCLUSION: The study findings support the brain-liver axis and suggest that environment-induced eustress promotes liver regeneration through the sympathetic nerve/ILC1/IL-22 axis.


Assuntos
Imunidade Inata , Regeneração Hepática , Camundongos , Animais , Regeneração Hepática/fisiologia , Linfócitos , Interleucinas , Hepatectomia , Fígado , Hepatócitos/fisiologia , Camundongos Endogâmicos C57BL
12.
J Cell Biol ; 222(4)2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36716168

RESUMO

Hepatocytes grow their apical surfaces anisotropically to generate a 3D network of bile canaliculi (BC). BC elongation is ensured by apical bulkheads, membrane extensions that traverse the lumen and connect juxtaposed hepatocytes. We hypothesize that apical bulkheads are mechanical elements that shape the BC lumen in liver development but also counteract elevated biliary pressure. Here, by resolving their structure using STED microscopy, we found that they are sealed by tight junction loops, connected by adherens junctions, and contain contractile actomyosin, characteristics of mechanical function. Apical bulkheads persist at high pressure upon microinjection of fluid into the BC lumen, and laser ablation demonstrated that they are under tension. A mechanical model based on ablation results revealed that apical bulkheads double the pressure BC can hold. Apical bulkhead frequency anticorrelates with BC connectivity during mouse liver development, consistent with predicted changes in biliary pressure. Our findings demonstrate that apical bulkheads are load-bearing mechanical elements that could protect the BC network against elevated pressure.


Assuntos
Canalículos Biliares , Bile , Hepatócitos , Animais , Camundongos , Junções Aderentes , Canalículos Biliares/fisiologia , Hepatócitos/fisiologia , Fígado , Junções Íntimas , Actomiosina , Pressão , Estresse Mecânico
13.
Methods Mol Biol ; 2575: 181-193, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36301476

RESUMO

Currently, the only feasible option for patients with progressive and/or end-stage organ degeneration is to undergo transplantation. Due to the growing unmatched demand of available organ donors and, as a consequence, the continuous growth of patients' waiting lists, the development of new tissue engineering technologies is a relevant need. In this chapter, we will focus on the liver as a model organ to discuss contemporary tissue engineering strategies. Induced pluripotent cells are an attractive alternative to serve as a cell source for tissue engineering applications due to their pluripotency, the potentiality to generate autologous transplantation, and for their high proliferation rate. Among the main liver tissue engineering technologies, 3D bioprinting, hepatic organoids, and decellularization/recellularization of biological matrixes have grown much attention as alternatives to derive functional liver grafts. Thus, this chapter will discuss how recent publications have demonstrated the use of induced pluripotent cells in the development of the aforementioned technologies. Bioprinting is an additive manufacturing biofabrication process where cells are dispersed within a matrix formulation (i.e., bioink) and extruded in a modified 3D-printer. Polymers within bioink can be cross-linked to increase stiffness. Hepatic spheroids showed greater viability and liver function, due to preserved epithelial phenotype over time. Organoid is multi-lineage tissue constructs derived from a stem cell that recapitulates the early stages of organogenesis. The influence of cellular composition of non-parenchymal cells using induced pluripotent-derived cells or primary adult cells for hepatic organoid formation was recently tested. Decellularization is a process where harvested tissues or organs are washed with a detergent-based solution, to lyse and remove all cellular components. The final product is an extracellular scaffold with preserved tissue vasculature and ultra-structure, which can be used for subsequent recellularization with recipient cells. This chapter sheds light on recent works on the use of induced pluripotent-derived cells for liver tissue engineering approaches and on how such technologies could potentially generate therapeutic alternatives for patients on waiting lists for liver transplantation.


Assuntos
Proliferação de Células , Hepatócitos , Células-Tronco , Engenharia Tecidual , Bioimpressão , Proliferação de Células/fisiologia , Hepatócitos/fisiologia , Transplante de Órgãos , Impressão Tridimensional , Células-Tronco/fisiologia , Tecnologia , Engenharia Tecidual/métodos
14.
Int J Mol Sci ; 23(19)2022 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-36232770

RESUMO

Hepatic glucose production (HGP) is an important component of glucose homeostasis, and deregulated HGP, particularly through gluconeogenesis, contributes to hyperglycemia and pathology of type-2 diabetes (T2D). It has been shown that the gluconeogenic gene expression is governed primarily by the transcription factor cAMP-response element (CRE)-binding protein (CREB) and its coactivator, CREB-regulated transcriptional coactivator 2 (CRTC2). Recently, we have discovered that Sam68, an adaptor protein and Src kinase substrate, potently promotes hepatic gluconeogenesis by promoting CRTC2 stability; however, the detailed mechanisms remain unclear. Here we show that in response to glucagon, Sam68 increases CREB/CRTC2 transactivity by interacting with CRTC2 in the CREB/CRTC2 complex and occupying the CRE motif of promoters, leading to gluconeogenic gene expression and glucose production. In hepatocytes, glucagon promotes Sam68 nuclear import, whereas insulin elicits its nuclear export. Furthermore, ablation of Sam68 in hepatocytes protects mice from high-fat diet (HFD)-induced hyperglycemia and significantly increased hepatic and peripheral insulin sensitivities. Thus, hepatic Sam68 potentiates CREB/CRTC2-mediated glucose production, contributes to the pathogenesis of insulin resistance, and may serve as a therapeutic target for T2D.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Diabetes Mellitus Tipo 2 , Gluconeogênese , Glucose , Hepatócitos , Resistência à Insulina , Proteínas de Ligação a RNA , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Expressão Gênica , Glucagon/metabolismo , Gluconeogênese/genética , Gluconeogênese/fisiologia , Glucose/metabolismo , Hepatócitos/metabolismo , Hepatócitos/fisiologia , Homeostase , Hiperglicemia/metabolismo , Insulina/metabolismo , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Fígado/metabolismo , Camundongos , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
15.
Acta Physiol (Oxf) ; 235(3): e13827, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35500155

RESUMO

AIM: Low-grade inflammation is the hallmark of non-alcoholic fatty liver diseases (NAFLD) and non-alcoholic steatohepatitis (NASH). The leakage of microbiota-derived products can contribute to liver inflammation during NAFLD/NASH development. Here, we assessed the roles of gut microbial DNA-containing extracellular vesicles (mEVs) in regulating liver cellular abnormalities in the course of NAFLD/NASH. METHODS: We performed studies with Vsig4-/- , C3-/- , cGAS-/- , and their wild-type littermate mice. Vsig4+ macrophage population and bacterial DNA abundance were examined in both mouse and human liver by either flow cytometric or immunohistochemistry analysis. Gut mEVs were adoptively transferred into Vsig4-/- , C3-/- , cGAS-/- , or littermate WT mice, and hepatocyte inflammation and HSC fibrogenic activation were measured in these mice. RESULTS: Non-alcoholic fatty liver diseases and non-alcoholic steatohepatitis development was concomitant with a diminished liver Vsig4+ macrophage population and a marked bacterial DNA enrichment in both hepatocytes and HSCs. In the absence of Vsig4+ macrophages, gut mEVs translocation led to microbial DNA accumulation in hepatocytes and HSCs, resulting elevated hepatocyte inflammation and HSC fibrogenic activation. In contrast, in lean WT mice, Vsig4+ macrophages remove gut mEVs from bloodstream through a C3-dependent opsonization mechanism and prevent the infiltration of gut mEVs into hepatic cells. Additionally, Vsig4-/- mice more quickly developed significant liver steatosis and fibrosis than WT mice after Western diet feeding. In vitro treatment with NASH mEVs triggered hepatocyte inflammation and HSC fibrogenic activation. Microbial DNAs are key cargo for the effects of gut mEVs by activating cGAS/STING. CONCLUSION: Accumulation of microbial DNAs fuels the development of NAFLD/NASH-associated liver abnormalities.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Animais , DNA Bacteriano , Modelos Animais de Doenças , Fibrose , Hepatócitos/patologia , Hepatócitos/fisiologia , Inflamação/patologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Nucleotidiltransferases
16.
Mol Biol Rep ; 49(7): 5843-5852, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35338439

RESUMO

BACKGROUND: Alcohol is mainly catabolized by class I alcohol dehydrogenase (ADH1) in liver. ADH deficiency can aggravate ethanol-induced tissue injury. Extracellular signal-regulated kinases 1/2 (ERK1/2) is involved in alcohol metabolism. However, the relationship between ERK1/2 and ADH1 remains unclear. METHODS AND RESULTS: To inhibit ERK1/2, HepG2 and BNL cells were treated with mitogen-activated protein kinases 1/2 (MEK1/2) inhibitors (U0126 and PD98059), and C57BL/6J mice were fed U0126. After treatment, the protein and mRNA expression of ADH1 were determined by Western blot and quantitative real time-PCR. The activity of ADH1 promoter was detected using luciferase assay. The results showed MEK1/2 inhibitors significantly increased ADH1 protein expression by inducing its transcription activity. Then we demonstrated a farnesoid X receptor (FXR) response element (FXRE) in ADH1 promoter by ChIP assay. To test whether FXR mediates the induction of MEK1/2 inhibitors on ADH1, HepG2 cells were transfected with FXR siRNA or ADH1 promoters with FXRE mutation. We found both FXR siRNA and FXRE mutation in ADH1 promoter abolished MEK1/2 inhibitors-induced ADH1 expression, indicating the activation of MEK1/2 inhibitors on ADH1 depends on FXR. CONCLUSIONS: Our findings revealed inhibition of ERK1/2 can significantly increase ADH1 expression, indicating MEK1/2 inhibitors may possess potential application in alcohol-related diseases.


Assuntos
Álcool Desidrogenase , Hepatócitos , Inibidores de Proteínas Quinases/farmacologia , Álcool Desidrogenase/genética , Animais , Hepatócitos/fisiologia , Fígado , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno
17.
Mol Biol Rep ; 49(5): 4089-4093, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35305226

RESUMO

BACKGROUND: The constitutive androstane receptor (CAR, NR1I3)-mediated mechanisms regulating hepatocyte proliferation and growth of the liver did not yet experience complete elucidation. We investigated whether STAT3 could be activated in vivo by NR1I3 signaling in mouse liver. METHODS AND RESULTS: Using Western blot analysis, immunofluorescence assays and real-time PCR we established the state of STAT3 activation when it comes to the mouse liver subsequent to treatment ofNR1I3 agonist,1,4-bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP). STAT3 nuclear relocation and hepatocyte growth were both induced by NR1I3-mediated phosphorylation of STAT3. Moreover, the NR1I3-STAT3 signaling pathway's proliferation impact was facilitated, partly, by cMyc and Cyclin D1 upregulation. CONCLUSIONS: This work's evidence demonstrates that NR1I3-pushed STAT3 activation contributes to TCPOBOP-induced liver growth and hepatocyte proliferation, at least in part, through its molecular targets cMyc and CyclinD1.


Assuntos
Fígado , Receptores Citoplasmáticos e Nucleares , Animais , Proliferação de Células , Hepatócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Citoplasmáticos e Nucleares/genética , Transdução de Sinais
18.
Biochem Biophys Res Commun ; 598: 40-46, 2022 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-35151202

RESUMO

Hepatocytes injury caused by cytolethal distending toxin (CDT) are major events during helicobacter hepaticus (H.hepaticus) infection. Recent study showed that pre-survival autophagy was promoted against CdtB subunit induced DNA damage. In the present study, we demonstrated that inflammatory cytokines IL-6, IL-1ß, TNF-α, IFN-α, IFN-γ expression and STAT phosphorylation were promoted by CdtB. Besides, CdtB decreased cell viability while promote apoptosis in mouse liver (AML12) cells. Especially, apoptotic protein caspase-9, caspase-3 and PARP were activated while the ratio of Bcl-2/Bax was decreased after CdtB treatment. Moreover, apoptosis induced by CdtB was inhibited due to Erk/p38 MAPK signaling pathway suppression performed with SB203580 or U0126. Meanwhile, we found that CdtB increased autophagic marker levels accompanied by Akt/mTOR/P70S6K signaling pathway in a dose dependent manner. To assess the correlation between autophagy and apoptosis induced by H.hepaticus, chloroquine (CQ, 50 µM) was employed to inhibit autophagy. The result showed that inhibition of autophagy with CQ treatment promoted apoptosis induced by CdtB. Altogether, all these results suggest that CdtB triggers apoptosis via MAPK/Erk/p38 signaling pathway in caspase dependent manner, which was prevented by autophagy in AML12 cells. Collectively, our findings provide new insights into the virulence potential of CdtB on the molecular pathogenesis throughout H.hepaticus infection.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Toxinas Bacterianas/toxicidade , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Animais , Apoptose/fisiologia , Autofagia/fisiologia , Caspases/genética , Caspases/metabolismo , Linhagem Celular , Citocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Helicobacter hepaticus/patogenicidade , Hepatócitos/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos
19.
Endocrinology ; 163(3)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35038735

RESUMO

CONTEXT: 3,5,3'-L-triiodothyronine (T3) is a potent inducer of hepatocyte proliferation via the Wnt/ß-catenin signaling pathway. Previous studies suggested the involvement of rapid noncanonical thyroid hormone receptor (TR) ß signaling, directly activating hepatic Wnt/ß-catenin signaling independent from TRß DNA binding. However, the mechanism by which T3 increases Wnt/ß-catenin signaling in hepatocytes has not yet been determined. OBJECTIVE: We aimed to determine whether DNA binding of TRß is required for stimulation of hepatocyte proliferation by T3. METHODS: Wild-type (WT) mice, TRß knockout mice (TRß KO), and TRß mutant mice with either specifically abrogated DNA binding (TRß GS) or abrogated direct phosphatidylinositol 3 kinase activation (TRß 147F) were treated with T3 for 6 hours or 7 days. Hepatocyte proliferation was assessed by Kiel-67 (Ki67) staining and apoptosis by terminal deoxynucleotidyl transferase dUTP nick-end labeling assay. Activation of ß-catenin signaling was measured in primary murine hepatocytes. Gene expression was analyzed by microarray, gene set enrichment analysis (GSEA), and quantitative reverse transcription polymerase chain reaction. RESULTS: T3 induced hepatocyte proliferation with an increased number of Ki67-positive cells in WT and TRß 147F mice (9.2% ±â€…6.5% and 10.1% ±â€…2.9%, respectively) compared to TRß KO and TRß GS mice (1.2% ±â€…1.1% and 1.5% ±â€…0.9%, respectively). Microarray analysis and GSEA showed that genes of the Wnt/ß-catenin pathway-among them, Fzd8 (frizzled receptor 8) and Ctnnb1 (ß-catenin)-were positively enriched only in T3-treated WT and TRß 147F mice while B-cell translocation gene anti-proliferation factor 2 was repressed. Consequently, expression of Ccnd1 (CyclinD1) was induced. CONCLUSIONS: Instead of directly activating Wnt signaling, T3 and TRß induce key genes of the Wnt/ß-catenin pathway, ultimately stimulating hepatocyte proliferation via CyclinD1. Thus, canonical transcriptional TRß action is necessary for T3-mediated stimulation of hepatocyte proliferation.


Assuntos
Proliferação de Células/fisiologia , Hepatócitos/fisiologia , Receptores beta dos Hormônios Tireóideos/fisiologia , Tri-Iodotironina/farmacologia , Animais , Sítios de Ligação/genética , Proliferação de Células/efeitos dos fármacos , Ciclina D1/fisiologia , DNA/metabolismo , Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hipotireoidismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mutação , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Receptores beta dos Hormônios Tireóideos/genética , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...