Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
iScience ; 25(12): 105503, 2022 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-36404924

RESUMEN

Advances in cellular engineering, as well as gene, and cell therapy, may be used to produce human tissues with programmable genetically enhanced functions designed to model and/or treat specific diseases. Fabrication of synthetic human liver tissue with these programmable functions has not been described. By generating human iPSCs with target gene expression controlled by a guide RNA-directed CRISPR-Cas9 synergistic-activation-mediator, we produced synthetic human liver tissues with programmable functions. Such iPSCs were guide-RNA-treated to enhance expression of the clinically relevant CYP3A4 and UGT1A1 genes, and after hepatocyte-directed differentiation, cells demonstrated enhanced functions compared to those found in primary human hepatocytes. We then generated human liver tissue with these synthetic human iPSC-derived hepatocytes (iHeps) and other non-parenchymal cells demonstrating advanced programmable functions. Fabrication of synthetic human liver tissue with modifiable functional genetic programs may be a useful tool for drug discovery, investigating biology, and potentially creating bioengineered organs with specialized functions.

2.
Hepatol Commun ; 4(6): 859-875, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32490322

RESUMEN

Hepatocyte nuclear factor 4 alpha (HNF4α) is a transcription factor that plays a critical role in hepatocyte function, and HNF4α-based reprogramming corrects terminal liver failure in rats with chronic liver disease. In the livers of patients with advanced cirrhosis, HNF4α RNA expression levels decrease as hepatic function deteriorates, and protein expression is found in the cytoplasm. These findings could explain impaired hepatic function in patients with degenerative liver disease. In this study, we analyzed HNF4α localization and the pathways involved in post-translational modification of HNF4α in human hepatocytes from patients with decompensated liver function. RNA-sequencing analysis revealed that AKT-related pathways, specifically phospho-AKT, is down-regulated in cirrhotic hepatocytes from patients with terminal failure, in whom nuclear levels of HNF4α were significantly reduced, and cytoplasmic expression of HNF4α was increased. cMET was also significantly reduced in failing hepatocytes. Moreover, metabolic profiling showed a glycolytic phenotype in failing human hepatocytes. The contribution of cMET and phospho-AKT to nuclear localization of HNF4α was confirmed using Spearman's rank correlation test and pathway analysis, and further correlated with hepatic dysfunction by principal component analysis. HNF4α acetylation, a posttranslational modification important for nuclear retention, was also significantly reduced in failing human hepatocytes when compared with normal controls. Conclusion: These results suggest that the alterations in the cMET-AKT pathway directly correlate with HNF4α localization and level of hepatocyte dysfunction. This study suggests that manipulation of HNF4α and pathways involved in HNF4α posttranslational modification may restore hepatocyte function in patients with terminal liver failure.

3.
Cell Rep ; 31(9): 107711, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32492423

RESUMEN

The availability of an autologous transplantable auxiliary liver would dramatically affect the treatment of liver disease. Assembly and function in vivo of a bioengineered human liver derived from induced pluripotent stem cells (iPSCs) has not been previously described. By improving methods for liver decellularization, recellularization, and differentiation of different liver cellular lineages of human iPSCs in an organ-like environment, we generated functional engineered human mini livers and performed transplantation in a rat model. Whereas previous studies recellularized liver scaffolds largely with rodent hepatocytes, we repopulated not only the parenchyma with human iPSC-hepatocytes but also the vascular system with human iPS-endothelial cells, and the bile duct network with human iPSC-biliary epithelial cells. The regenerated human iPSC-derived mini liver containing multiple cell types was tested in vivo and remained functional for 4 days after auxiliary liver transplantation in immunocompromised, engineered (IL2rg-/-) rats.


Asunto(s)
Hepatocitos/trasplante , Ingeniería de Tejidos , Activinas/genética , Activinas/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Feto/citología , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Huésped Inmunocomprometido , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Andamios del Tejido/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Med Sci (Paris) ; 36(3): 261-263, 2020 Mar.
Artículo en Francés | MEDLINE | ID: mdl-32228845

RESUMEN

Organoids offer an elegant approach to model human diseases and test new drugs. Nonalcoholic fatty liver disease (NAFLD) whose incidence has dramatically increased in recent years with the rise of obesity, is defined by triglyceride accumulation in hepatocytes, inflammation, liver injury, and progression to fibrosis. There is currently no approved therapy but many pathways are being explored. Two American teams have created mini-steatotic livers using different approaches, both using induced pluripotent stem cells (iPS), thus offering new tools to test developing drugs.


TITLE: L'émergence des modèles miniatures de foie gras humain en 3D générés en laboratoire. ABSTRACT: Les organoïdes constituent une approche de choix pour modéliser a minima une maladie humaine et tester l'efficacité thérapeutique de certaines drogues. La stéatopathie métabolique ou maladie du foie gras, dont l'incidence a considérablement augmenté avec l'accroissement de l'obésité dans les pays développés, se caractérise par l'accumulation de triglycerides dans l'hépatocyte et une atteinte hépatique pouvant évoluer vers la fibrose. Il n'existe pas de traitement efficace, mais de nombreuses pistes sont actuellement explorées. Deux équipes américaines ont récemment utilisé les cellules souches pluripotentes induites (iPS) et la culture muticellulaire pour modéliser un mini-foie stéatosique par deux approches différentes, offrant ainsi de nouveaux outils pour tester les drogues en cours de développement.


Asunto(s)
Bioingeniería/tendencias , Ciencia del Laboratorio Clínico/tendencias , Enfermedad del Hígado Graso no Alcohólico/patología , Técnicas de Cultivo de Tejidos/tendencias , Animales , Bioingeniería/métodos , Células Cultivadas , Progresión de la Enfermedad , Hepatocitos/citología , Hepatocitos/patología , Hepatocitos/fisiología , Humanos , Hígado/patología , Hígado/fisiología , Cirrosis Hepática/patología , Ciencia del Laboratorio Clínico/métodos , Modelos Biológicos , Técnicas de Cultivo de Tejidos/métodos , Andamios del Tejido
5.
Cell Metab ; 30(2): 385-401.e9, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31390551

RESUMEN

The mechanisms by which steatosis of the liver progresses to non-alcoholic steatohepatitis and end-stage liver disease remain elusive. Metabolic derangements in hepatocytes controlled by SIRT1 play a role in the development of fatty liver in inbred animals. The ability to perform similar studies using human tissue has been limited by the genetic variability in man. We generated human induced pluripotent stem cells (iPSCs) with controllable expression of SIRT1. By differentiating edited iPSCs into hepatocytes and knocking down SIRT1, we found increased fatty acid biosynthesis that exacerbates fat accumulation. To model human fatty livers, we repopulated decellularized rat livers with human mesenchymal cells, fibroblasts, macrophages, and human SIRT1 knockdown iPSC-derived hepatocytes and found that the human iPSC-derived liver tissue developed macrosteatosis, acquired proinflammatory phenotype, and shared a similar lipid and metabolic profiling to human fatty livers. Biofabrication of genetically edited human liver tissue may become an important tool for investigating human liver biology and disease.


Asunto(s)
Ingeniería Celular , Hígado Graso/metabolismo , Células Madre Pluripotentes/metabolismo , Sirtuina 1/metabolismo , Adulto , Animales , Diferenciación Celular , Células Cultivadas , Ácidos Grasos/biosíntesis , Humanos , Masculino , Células Madre Pluripotentes/citología , Ratas , Ratas Sprague-Dawley , Sirtuina 1/deficiencia , Sirtuina 1/genética
6.
Stem Cells Dev ; 28(14): 907-919, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31122128

RESUMEN

Differentiation of stem cells to hepatocyte-like cells (HLCs) holds great promise for basic research, drug and toxicological investigations, and clinical applications. There are currently no protocols for the production of HLCs from stem cells, such as embryonic stem cells or induced pluripotent stem cells, that produce fully mature hepatocytes with a wide range of mature hepatic functions. This report describes a standard method to assess the maturation of stem cell-derived HLCs with a moderately high-throughput format, by analysing liver gene expression by quantitative RT-qPCR. This method also provides a robust data set of the expression of 62 genes expressed in normal liver, generated from 17 fetal and 25 mature human livers, so that investigators can quickly and easily compare the expression of these genes in their stem cell-derived HLCs with the values obtained in authentic fetal and mature human liver. The simple methods described in this study will provide a quick and accurate assessment of the efficacy of a differentiation protocol and will help guide the optimization of differentiation conditions.


Asunto(s)
Diferenciación Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Hígado/metabolismo , Hepatocitos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Hígado/citología
7.
Am J Pathol ; 189(3): 502-512, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30653953

RESUMEN

Endothelial cells are prevalent in our bodies and serve multiple functions. By lining the vasculature, they provide a barrier to tissues and facilitate the transport of molecules and cells. They also maintain hemostasis and modulate blood flow by reacting to chemokines and releasing signal molecules. Thus, endothelial dysfunction leads to a wide variety of diseases, including atherosclerosis and coronary artery disease. In today's era of stem cell research, induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) have emerged for research and engineering purposes. They are not only tools for studying disease states but are also a crucial part of efforts to engineer vessel and organ grafts. As the techniques in cell culture, microfluidics, and personalized medicine concomitantly improve, the potential for iPSC-ECs is enormous. We review functions of endothelium in our bodies, the development and uses of iPSC-ECs, and the possible avenues to explore in the future.


Asunto(s)
Aterosclerosis , Prótesis Vascular , Enfermedad de la Arteria Coronaria , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Ingeniería de Tejidos , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/terapia , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/terapia , Células Endoteliales/patología , Humanos , Células Madre Pluripotentes Inducidas/patología
8.
Hepatol Commun ; 2(5): 582-594, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29761173

RESUMEN

The mechanisms by which the liver fails in end-stage liver disease remain elusive. Disruption of the transcription factor network in hepatocytes has been suggested to mediate terminal liver failure in animals. However, this hypothesis remains unexplored in human subjects. To study the relevance of transcription factor expression in terminal stages of chronic liver failure in humans, we analyzed the expression of liver-enriched transcription factors (LETFs) hepatocyte nuclear factor (HNF)4α, HNF1α, forkhead box protein A2 (FOXA2), CCAAT/enhancer-binding protein (CEBP)α, and CEBPß. We then selected downstream genes responsible for some hepatic functions (ornithine transcarbamylase [OTC], cytochrome P450 3A4 [CYP3A4], coagulation factor VII [F7], cadherin 1 [CDH1], phospho-ezrin (Thr567)/radixin (Thr564)/moesin (Thr558) [p-ERM], phospho-myosin light chain [p-MLC], low-density lipoprotein receptor-related protein 1 [LRP1]) in liver tissue from patients at different stages of decompensated liver function based upon Child-Pugh classification, Model for End-Stage Liver Disease score, and degree of inflammatory activity/fibrosis. We first examined differential expression of LETF and determined whether a relationship exists between transcript and protein expression, and liver function. We found HNF4α expression was down-regulated and correlated well with the extent of liver dysfunction (P = 0.001), stage of fibrosis (P = 0.0005), and serum levels of total bilirubin (P = 0.009; r = 0.35), albumin (P < 0.001; r = 0.52), and prothrombin time activity (P = 0.002; r = 0.41). HNF4α expression also correlated with CYP3A4, OTC, and F7 as well as CDH1 RNA levels. The Rho/Rho-associated protein kinase pathways, which have been implicated in the regulation of HNF4α, were also differentially expressed, in concert with LRP1, a reported upstream regulator of RhoA function. Conclusion: HNF4α and other members of the LETFs appear to be important regulators of hepatocyte function in patients with chronic hepatic failure. (Hepatology Communications 2018;2:582-594).

9.
Am J Pathol ; 188(6): 1316-1327, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29673755

RESUMEN

Liver regeneration is a complex and unique process. When two-thirds of a mouse liver is removed, the remaining liver recovers its initial weight in approximately 10 days. The understanding of the mechanisms responsible for liver regeneration may help patients needing large liver resections or transplantation and may be applied to the field of regenerative medicine. All differentiated hepatocytes are capable of self-renewal, but different subpopulations of hepatocytes seem to have distinct proliferative abilities. In the setting of chronic liver diseases, a ductular reaction ensues in which liver progenitor cells (LPCs) proliferate in the periportal region. Although these LPCs have the capacity to differentiate into hepatocytes and biliary cells in vitro, their ability to participate in liver regeneration is far from clear. Their expansion has even been associated with increased fibrosis and poorer prognosis in chronic liver diseases. Controversies also remain on their origin: lineage studies in experimental mouse models of chronic injury have recently suggested that these LPCs originate from hepatocyte dedifferentiation, whereas in other situations, they seem to come from cholangiocytes. This review summarizes data published in the past 5 years in the liver regeneration field, discusses the mechanisms leading to regeneration disruption in chronic liver disorders, and addresses the potential use of novel approaches for regenerative medicine.


Asunto(s)
Hepatocitos/citología , Hepatopatías/terapia , Regeneración Hepática , Medicina Regenerativa , Animales , Humanos
10.
Organogenesis ; 13(3): 83-94, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28548903

RESUMEN

Organ-like microenviroment and 3-dimensional (3D) cell culture conformations have been suggested as promising approaches to mimic in a micro-scale a whole organ cellular functions and interactions present in vivo. We have used this approach to examine biologic features of hepatocellular carcinoma (HCC) cells. In this study, we demonstrate that hepatocellular carcinoma (HCC) cells, fibroblasts, endothelial cells and extracellular matrix can generate organoid-like spheroids that enhanced numerous features of human HCC observed in vivo. We show that the addition of non-parenchymal cells such as fibroblast and endothelial cells is required for spheroid formation as well as the maintenance of the tissue-like structure. Furthermore, HCC cells cultured as spheroids with non-parenchymal cells express more neo-angiogenesis-related markers (VEGFR2, VEGF, HIF-α), tumor-related inflammatory factors (CXCR4, CXCL12, TNF-α) and molecules-related to induced epithelial-mesenchymal transition (TGFß, Vimentin, MMP9) compared with organoids containing only HCC cells. These results demonstrate the importance of non-parenchymal cells in the cellular composition of HCC organoids. The novelty of the multicellular-based organotypic culture system strongly supports the integration of this approach in a high throughput approach to identified patient-specific HCC malignancy and accurate anti-tumor therapy screening after surgery.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Neovascularización Patológica/patología , Organoides/patología , Microambiente Tumoral , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/irrigación sanguínea , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Transición Epitelial-Mesenquimal , Humanos , Inflamación/patología , Neoplasias Hepáticas/irrigación sanguínea
11.
Organogenesis ; 13(1): 1-15, 2017 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-28055309

RESUMEN

Stem cell research has significantly evolved over the last few years, allowing the differentiation of pluripotent cells into almost any kind of lineage possible. Studies that focus on the liver have considerably taken a leap into this novel technology, and hepatocyte-like cells are being generated that are close to resembling actual hepatocytes both genotypically and phenotypically. The potential of this extends from disease models to bioengineering, and even also innovative therapies for end-stage liver disease. Nonetheless, too few attention has been given to the non-parenchymal cells which are also fundamental for normal liver function. This includes cholangiocytes, the cells of the biliary epithelium, without whose role in bile modification and metabolism would impair hepatocyte survival. Such can be observed in diseases that target them, so called cholangiopathies, for which there is much yet to study so as to improve therapeutical options. Protocols that describe the induction of human induced pluripotent stem cells into cholangiocytes are scarce, although progress is being achieved in this area as well. In order to give the current view on this emerging research field, and in hopes to motivate further advances, we present here a review on the known differentiation strategies with sight into future applications.


Asunto(s)
Conductos Biliares/citología , Técnicas de Cultivo de Célula/métodos , Técnicas de Cultivo de Célula/tendencias , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Investigación Biomédica , Hepatocitos/citología , Humanos
12.
Transplantation ; 101(1): 92-100, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28009756

RESUMEN

BACKGROUND: Auxiliary partial liver transplantation (APLT) in humans is a therapeutic modality used especially to treat liver failure in children or congenital metabolic disease. Animal models of APLT have helped to explore therapeutic options. Though many groups have suggested improvements, standardizing the surgical procedure has been challenging. Additionally, the question of whether graft livers are reconstituted by recipient-derived cells after transplantation has been controversial. The aim of this study was to improve experimental APLT in rats and to assess cell recruitment in the liver grafts. METHODS: To inhibit recipient liver regeneration and to promote graft regeneration, we treated recipients with retrorsine and added arterial anastomosis. Using green fluorescence protein transgenic rats as recipients, we examined liver resident cell recruitment within graft livers by immunofluorescence costaining. RESULTS: In the improved APLT model, we achieved well-regenerated grafts that could maintain regeneration for at least 4 weeks. Regarding the cell recruitment, there was no evidence of recipient-derived hepatocyte, cholangiocyte, or hepatic stellate cell recruitment into the graft. Macrophages/monocytes, however, were consistently recruited into the graft and increased over time, which might be related to inflammatory responses. Very few endothelial cells showed colocalization of markers. CONCLUSIONS: We have successfully established an improved rat APLT model with arterial anastomosis as a standard technique. Using this model, we have characterized cell recruitment into the regenerating grafts.


Asunto(s)
Movimiento Celular , Proliferación Celular , Regeneración Hepática , Trasplante de Hígado/métodos , Hígado/cirugía , Animales , Linaje de la Célula , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Hígado/fisiopatología , Regeneración Hepática/efectos de los fármacos , Masculino , Modelos Animales , Alcaloides de Pirrolicidina/farmacología , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Ratas Transgénicas , Factores de Tiempo
13.
PLoS One ; 11(2): e0149344, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26890260

RESUMEN

There are unprecedented epidemics of obesity, such as type II diabetes and non-alcoholic fatty liver diseases (NAFLD) in developed countries. A concerning percentage of American children are being affected by obesity and NAFLD. Studies have suggested that the maternal environment in utero might play a role in the development of these diseases later in life. In this study, we documented that inhibiting SIRT1 signaling in human fetal hepatocytes rapidly led to an increase in intracellular glucose and lipids levels. More importantly, both de novo lipogenesis and gluconeogenesis related genes were upregulated upon SIRT1 inhibition. The AKT/FOXO1 pathway, a major negative regulator of gluconeogenesis, was decreased in the human fetal hepatocytes inhibited for SIRT1, consistent with the higher level of gluconeogenesis. These results indicate that SIRT1 is an important regulator of lipid and carbohydrate metabolisms within human fetal hepatocytes, acting as an adaptive transcriptional response to environmental changes.


Asunto(s)
Glucosa/metabolismo , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Sirtuina 1/genética , Benzamidas/farmacología , Vías Biosintéticas/efectos de los fármacos , Células Cultivadas , Femenino , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Gluconeogénesis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Masculino , Naftoles/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirtuina 1/antagonistas & inhibidores
14.
Organogenesis ; 11(4): 173-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26588350

RESUMEN

In the recent decades, human genome engineering has been one of the major interesting research subjects, essentially because it raises new possibilities for personalized medicine and biotechnologies. With the development of engineered nucleases such as the Zinc Finger Nucleases (ZFNs), the Transcription activator-like effector nucleases (TALENs) and more recently the Clustered Regularly Interspaced short Palindromic Repeats (CRISPR), the field of human genome edition has evolved very rapidly. Every new genetic tool is broadening the scope of applications on human tissues, even before we can completely master each of these tools. In this review, we will present the recent advances regarding human genome edition tools, we will discuss the numerous implications they have in research and medicine, and we will mention the limits and concerns about such technologies.


Asunto(s)
Edición Génica , Ingeniería Genética/tendencias , Genoma Humano , Organogénesis/genética , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Modelos Animales de Enfermedad , Humanos
15.
Int J Hepatol ; 2012: 476910, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23050157

RESUMEN

Unraveling the molecular clues of liver proliferation has become conceivable thanks to the model of two-third hepatectomy. The synchronicity and the well-scheduled aspect of this process allow scientists to slowly decipher this mystery. During this phenomenon, quiescent hepatocytes of the remnant lobes are able to reenter into the cell cycle initiating the G1-S progression synchronously before completing the cell cycle. The major role played by this step of the cell cycle has been emphasized by loss-of-function studies showing a delay or a lack of coordination in the hepatocytes G1-S progression. Two growth factor receptors, c-Met and EGFR, tightly drive this transition. Due to the level of complexity surrounding EGFR signaling, involving numerous ligands, highly controlled regulations and multiple downstream pathways, we chose to focus on the EGFR pathway for this paper. We will first describe the EGFR pathway in its integrity and then address its essential role in the G1/S phase transition for hepatocyte proliferation. Recently, other levels of control have been discovered to monitor this pathway, which will lead us to discuss regulations of the EGFR pathway and highlight the potential effect of misregulations in pathologies.

16.
Rejuvenation Res ; 14(4): 353-63, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21548759

RESUMEN

It has been suggested that increasing age is correlated with an acceleration of the progression of liver fibrosis induced by various agents, such as hepatitis C virus or chronic alcohol consumption. However, the cellular and molecular changes underlying this predisposition are not entirely understood. In the context of an aging population, it becomes challenging to decipher the mechanisms responsible for this higher susceptibility of older individuals to this acquired liver disorder. To address this issue, we induced liver fibrosis by carbon tetrachloride (CCl(4)) chronic administration to 8-week- and 15-month-old mice. We confirmed that susceptibility to fibrosis development increased with age and showed that aging did not affect fibrosis resolution capacity. We then focused on the impairment of hepatocyte proliferation, oxidative stress, and inflammation as potential mechanisms accelerating the development of fibrosis in the elderly. We detected no inhibition of hepatocyte proliferation after CCl(4) injury in 15-month-old mice, whereas it was inhibited after a partial hepatectomy. Finally, we observed that, in a context in which liver oxidative stress was not differentially increased in both experimental groups, there was a higher recruitment of inflammatory cells, including mostly macrophages and lymphocytes, oriented toward a T helper 2 (T(H)2) response in older mice. Our data show that in conditions of equivalent levels of oxidative stress and maintained hepatocyte proliferative capacity, an increased inflammatory reaction mainly composed of CD4(+) lymphocytes and macrophages expressing T(H)2 cytokines is the main factor involved in the higher susceptibility to fibrosis with increasing age.


Asunto(s)
Envejecimiento/patología , Susceptibilidad a Enfermedades , Inflamación/complicaciones , Inflamación/patología , Cirrosis Hepática/complicaciones , Cirrosis Hepática/patología , Animales , Tetracloruro de Carbono , Proliferación Celular , Enfermedad Crónica , Hepatocitos/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Necrosis , Estrés Oxidativo
17.
Endocrinology ; 152(7): 2731-41, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21540290

RESUMEN

GH is a pleiotropic hormone that plays a major role in proliferation, differentiation, and metabolism via its specific receptor. It has been previously suggested that GH signaling pathways are required for normal liver regeneration but the molecular mechanisms involved have yet to be determined. The aim of this study was to identify the mechanisms by which GH controls liver regeneration. We performed two thirds partial hepatectomies in GH receptor (GHR)-deficient mice and wild-type littermates and showed a blunted progression in the G(1)/S transition phase of the mutant hepatocytes. This impaired liver regeneration was not corrected by reestablishing IGF-1 expression. Although the initial response to partial hepatectomy at the priming phase appeared to be similar between mutant and wild-type mice, cell cycle progression was significantly blunted in mutant mice. The main defect in GHR-deficient mice was the deficiency of the epidermal growth factor receptor activation during the process of liver regeneration. Finally, among the pathways activated downstream of GHR during G(1) phase progression, namely Erk1/2, Akt, and signal transducer and activator of transcription 3, we only found a reduced Erk1/2 phosphorylation in mutant mice. In conclusion, our results demonstrate that GH signaling plays a major role in liver regeneration and strongly suggest that it acts through the activation of both epidermal growth factor receptor and Erk1/2 pathways.


Asunto(s)
Receptores ErbB/metabolismo , Regeneración Hepática , Hígado/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores de Somatotropina/fisiología , Transducción de Señal , Animales , Cruzamientos Genéticos , Activación Enzimática , Inducción Enzimática , Receptores ErbB/genética , Fase G1 , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Hígado/citología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Receptores de Somatotropina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...