Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Cell ; 72(1): 71-83.e7, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30220561

RESUMEN

Cancer cells entail metabolic adaptation and microenvironmental remodeling to survive and progress. Both calcium (Ca2+) flux and Ca2+-dependent signaling play a crucial role in this process, although the underlying mechanism has yet to be elucidated. Through RNA screening, we identified one long noncoding RNA (lncRNA) named CamK-A (lncRNA for calcium-dependent kinase activation) in tumorigenesis. CamK-A is highly expressed in multiple human cancers and involved in cancer microenvironment remodeling via activation of Ca2+-triggered signaling. Mechanistically, CamK-A activates Ca2+/calmodulin-dependent kinase PNCK, which in turn phosphorylates IκBα and triggers calcium-dependent nuclear factor κB (NF-κB) activation. This regulation results in the tumor microenvironment remodeling, including macrophage recruitment, angiogenesis, and tumor progression. Notably, our human-patient-derived xenograft (PDX) model studies demonstrate that targeting CamK-A robustly impaired cancer development. Clinically, CamK-A expression coordinates with the activation of CaMK-NF-κB axis, and its high expression indicates poor patient survival rate, suggesting its role as a potential biomarker and therapeutic target.


Asunto(s)
Carcinogénesis/genética , Neoplasias/genética , ARN Largo no Codificante/genética , Microambiente Tumoral/genética , Señalización del Calcio/genética , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Macrófagos/metabolismo , Macrófagos/patología , FN-kappa B/genética , Neoplasias/patología , Fosforilación , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
EMBO J ; 36(22): 3325-3335, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28963395

RESUMEN

The Hippo pathway plays essential roles in organ size control and cancer prevention via restricting its downstream effector, Yes-associated protein (YAP). Previous studies have revealed an oncogenic function of YAP in reprogramming glucose metabolism, while the underlying mechanism remains to be fully clarified. Accumulating evidence suggests long noncoding RNAs (lncRNAs) as attractive therapeutic targets, given their roles in modulating various cancer-related signaling pathways. In this study, we report that lncRNA breast cancer anti-estrogen resistance 4 (BCAR4) is required for YAP-dependent glycolysis. Mechanistically, YAP promotes the expression of BCAR4, which subsequently coordinates the Hedgehog signaling to enhance the transcription of glycolysis activators HK2 and PFKFB3. Therapeutic delivery of locked nucleic acids (LNAs) targeting BCAR4 attenuated YAP-dependent glycolysis and tumor growth. The expression levels of BCAR4 and YAP are positively correlated in tissue samples from breast cancer patients, where high expression of both BCAR4 and YAP is associated with poor patient survival outcome. Taken together, our study not only reveals the mechanism by which YAP reprograms glucose metabolism, but also highlights the therapeutic potential of targeting YAP-BCAR4-glycolysis axis for breast cancer treatment.


Asunto(s)
Glucosa/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal , Secuencia de Bases , Neoplasias de la Mama/genética , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Femenino , Glucólisis/genética , Células HEK293 , Hexoquinasa/genética , Hexoquinasa/metabolismo , Humanos , Modelos Biológicos , Fosfofructoquinasa-2/genética , Fosfofructoquinasa-2/metabolismo , Transcripción Genética , Resultado del Tratamiento , Regulación hacia Arriba/genética
4.
Hepatology ; 61(5): 1708-20, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25501710

RESUMEN

UNLABELLED: Liver fibrosis, a major cause of end-stage liver diseases, is closely regulated by multiple growth factors and cytokines. The correlation of fibroblast growth factor 2 (FGF2) with chronic liver injury has been reported, but the exact functions of different FGF2 isoforms in liver fibrogenesis remain unclear. Here, we report on the differential expression patterns and functions of low- and high-molecular-weight FGF2 (namely, FGF2(lmw) and FGF2(hmw) , respectively) in hepatic fibrogenesis using a CCl4 -induced mouse liver fibrosis model. FGF2(hmw) displayed a robust increase in CCl4 -induced hepatic fibrosis and promoted fibrogenesis. In contrast, endogenous FGF2(lmw) exhibited a slight increase in hepatic fibrosis and suppressed this pathological progression. Moreover, exogenous administration of recombinant FGF2(lmw) potently ameliorated CCl4 -induced liver fibrosis. Mechanistically, we showed that FGF2(lmw) treatment attenuated hepatic stellate cell activation and fibrosis by epigenetic down-regulation of Delta-like 1 expression through the p38 mitogen-activated protein kinase pathway. CONCLUSION: FGF2(lmw) and FGF2(hmw) have distinct roles in liver fibrogenesis. These findings demonstrate a potent antifibrotic effect of FGF2(lmw) administration, which may provide a novel approach to treat chronic liver diseases.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor 2 de Crecimiento de Fibroblastos/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/fisiología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/genética , Animales , Proteínas de Unión al Calcio , Factor 2 de Crecimiento de Fibroblastos/fisiología , Ratones , Ratones Endogámicos C57BL , Peso Molecular
5.
Front Biosci (Landmark Ed) ; 29(4): 139, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38682178

RESUMEN

BACKGROUND: Hypoxic-ischaemic encephalopathy (HIE) is a major cause of neonatal disability and mortality. Although hypothermia therapy offers some neuroprotection, the recovery of neurological function is limited. Therefore, new synergistic therapies are necessary to improve the prognosis. Mesenchymal stem cell-based therapy is emerging as a promising treatment option for HIE. In this study, we studied the therapeutic efficacy of human placenta-derived mesenchymal stem cells (PD-MSCs) in the HIE rat model and analyzed the underlying therapeutic mechanisms. METHODS: Rats were divided into 6 groups (n = 9 for each) as follows: control, HIE model, HIE + normal saline, and HIE + PD-MSC transplantation at days 7, 14 and 28 postpartum. Following PD-MSC transplantation, neurological behavior was evaluated using rotarod tests, traction tests, and the Morris water maze test. The degree of brain tissue damage was assessed by histological examination and Nissl staining. Expression levels of apoptosis-related proteins and inflammatory factors were quantified by Western blotting and enzyme-linked immunosorbent assays. Immunofluorescence was used to investigate the ability of PD-MSCs to repair the morphology and function of hippocampal neurons with hypoxic-ischaemic (HI) injury. RESULTS: PD-MSC transplantation enhanced motor coordination and muscle strength in HIE rats. This treatment also improved spatial memory ability by repairing pathological damage and preventing the loss of neurons in the cerebral cortex. The most effective treatment was observed in the HIE + PD-MSC transplantation at day 7 group. Expression levels of microtubule-associated protein-2 (MAP-2), B-cell lymphoma-2 (BCL-2), interleukin (IL)-10, and transforming growth factor (TGF -ß1) were significantly higher in the HIE + PD-MSC treatment groups compared to the HIE group, whereas the levels of BCL-2-associated X protein (BAX), BCL-2-associated agonist of cell death (BAD), IL-1ß and tumour necrosis factor α (TNF-α) were significantly lower. CONCLUSIONS: We demonstrated that intravenous injection of PD-MSC at 7, 14 and 28 days after intrauterine HI damage in a rat model could improve learning, memory, and motor function, possibly by inhibiting apoptosis and inflammatory damage. These findings indicate that autologous PD-MSC therapy could have potential application for the treatment of HIE.


Asunto(s)
Apoptosis , Hipoxia-Isquemia Encefálica , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Placenta , Ratas Sprague-Dawley , Animales , Femenino , Trasplante de Células Madre Mesenquimatosas/métodos , Embarazo , Hipoxia-Isquemia Encefálica/terapia , Humanos , Placenta/citología , Células Madre Mesenquimatosas/citología , Ratas , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Inflamación/terapia , Neuronas/metabolismo , Masculino
6.
J Biol Chem ; 286(14): 12340-8, 2011 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-21239501

RESUMEN

Chronic liver injury always progresses to fibrosis and eventually to cirrhosis, a massive health care burden worldwide. Delta-like 1 (Dlk1) is well known as an inhibitor of adipocyte differentiation. However, whether it is involved in liver fibrosis remains unclear. Here, we provide the first evidence that Dlk1 is a critical contributor to liver fibrosis through promoting activation of hepatic stellate cells (HSCs) during chronic liver injury. We found that upon liver injury, Dlk1 was dramatically induced and initially expressed in hepatocytes and then into the HSCs by a paracrine manner. It leads to the activation of HSCs, which is considered to be a pivotal event in liver fibrogenesis. Two forms (∼50 and ∼25 kDa) of the Dlk1 protein were detected by Western blot analysis. In vitro administration of Dlk1 significantly promoted HSC activation, whereas in vivo knockdown of Dlk1 dramatically inhibited HSC activation and the subsequent fibrosis. The large soluble form (∼50 kDa) of Dlk1 was shown to contribute to HSC activation. We were encouraged to find the Dlk1-promoted HSC activation and liver fibrosis can be depressed by transplantation of bone marrow-mesenchymal stem cells (BM-MSCs). Furthermore, we demonstrated that FGF2 was up-regulated in BM-MSCs under injury stimulation, and it probably participated in the inhibition of Dlk1 by BM-MSCs. Our findings provide a novel role of Dlk1 in liver fibrosis leading to a better understanding of the molecular basis in fibrosis and cirrhosis and also give insights into the cellular and molecular mechanisms of MSC biology in liver repair.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cirrosis Hepática/metabolismo , Trasplante de Células Madre Mesenquimatosas , Animales , Western Blotting , Proteínas de Unión al Calcio , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/genética , Cirrosis Hepática/genética , Masculino , Ratones , Ratones Endogámicos ICR , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa
7.
Cell Mol Life Sci ; 68(15): 2615-26, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21063894

RESUMEN

CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg cells) are critical for the maintenance of peripheral tolerance, and the suppression of autoimmune diseases and even tumors. Although Treg cells are well characterized in humans, little is known regarding their existence or occurrence in ancient vertebrates. In the present study, we report on the molecular and functional characterization of a Treg-like subset with the phenotype CD4-2(+)CD25-like(+)Foxp3-like(+) from a pufferfish (Tetraodon nigroviridis) model. Functional studies showed that depletion of this subset produced an enhanced mixed lymphocyte reaction (MLR) and nonspecific cytotoxic cell (NCC) activity in vitro, as well as inflammation of the intestine in vivo. The data presented here will not only enrich the knowledge of fish immunology but will also be beneficial for a better cross-species understanding of the evolutionary history of the Treg family and Treg-mediated regulatory networks in cellular immunity.


Asunto(s)
Evolución Biológica , Linfocitos T Reguladores/citología , Tetraodontiformes/inmunología , Vertebrados/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Antígenos CD4/genética , Antígenos CD4/metabolismo , Separación Celular , Células Cultivadas , Clonación Molecular , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Tetraodontiformes/genética , Tetraodontiformes/metabolismo , Vertebrados/genética , Vertebrados/metabolismo
8.
J Cell Mol Med ; 14(6B): 1494-508, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19780871

RESUMEN

Recent studies suggest that mesenchymal stem cells (MSCs) possess a greater differentiation potential than once thought and that they have the capacity to regenerate damaged tissues/organs. However, the evidence is insufficient, and the mechanism governing the recruitment and homing of MSCs to these injured sites is not well understood. We first examined the MSCs circulating in peripheral blood and then performed chemotaxis, wound healing and tubule-formation assays to investigate the migration capability of mouse bone marrow MSCs (mBM-MSCs) in response to liver-injury signals. In addition, BM-MSCs from donor enhanced green fluorescent protein transgenic male mice were transplanted into liver-injured co-isogenic female recipients, either by intra-bone marrow injection or through the caudal vein, to allow in vivo tracking analysis of the cell fate after transplantation. Donor-derived cells were analysed by in vivo imaging analysis, PCR, flow cytometry and frozen sections. Microarray and real-time PCR were used for chemokine/cytokine and receptor analyses. We successfully isolated circulating MSCs in peripheral blood of liver-injured mice and provided direct evidence that mBM-MSCs could be mobilized into the circulation and recruited into the liver after stimulation of liver injury. CCR9, CXCR4 and c-MET were essential for directing cellular migration towards the injured liver. The recruited mBM-MSCs may play different roles, including hepatic fate specification and down-regulation of the activity of hepatic stellate cells which inhibits over-accumulation of collagen and development of liver fibrosis. Our results provide new insights into liver repair involving endogenous BM-MSCs and add new information for consideration when developing clinical protocols involving the MSCs.


Asunto(s)
Células de la Médula Ósea/citología , Hígado/lesiones , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Animales , Anticuerpos/farmacología , Bioensayo , Movimiento Celular/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Cell Mol Med ; 13(8B): 2582-2592, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18705698

RESUMEN

Bone marrow stromal stem cells (BMSSCs) may have potential to differentiate in vitro and in vivo into hepatocytes. Here, we investigated the effects of valproic acid (VPA) involved in epigenetic modification, a direct inhibitor of histone deacetylase, on hepatic differentiation of mouse BMSSCs. Following the treatment of 2.5 mM VPA for 72 hrs, the in vitro expanded, highly purified and functionally active mouse BMSSCs from bone marrow were either exposed to some well-defined cytokines and growth factors in a sequential way (fibroblast growth factor-4 [FGF-4], followed by HGF, and HGF + OSM + ITS + dexamethasone, resembling the order of secretion during liver embryogenesis) or transplanted (caudal vein) in mice submitted to a protocol of chronic injury (chronic i.p. injection of CCl4). Additional exposure of the cells to VPA considerably improved the in vitro differentiation, as demonstrated by a more homogeneous cell population exhibited epithelial morphology, increasing expression of hepatic special genes and enhanced hepatic functions. Further more, in vivo results indicate that the pre-treatment of VPA significantly increased the homing efficiency of BMSSCs to the site of liver injury and, additionally, for supporting hepatic differentiation as well as in vitro. We have demonstrated the usefulness of VPA in the transdifferentiation of BMSSCs into hepatocytes both in vitro and in vivo, and regulation of fibroblast growth factor receptors (FGFRs) and c-Met gene expression through post-translational modification of core histones might be the primary initiating event for these effects. This mode could be helpful for liver engineering and clinical therapy.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Hígado/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Hígado/citología , Ratones , Ratones Endogámicos ICR , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Zhonghua Gan Zang Bing Za Zhi ; 15(8): 597-600, 2007 Aug.
Artículo en Zh | MEDLINE | ID: mdl-17711630

RESUMEN

OBJECTIVE: To establish a method through which murine bone marrow mesenchymal stem cells (MSCs) can be induced into hepatocytes in vitro. METHODS: A conditioned medium of injured hepatocytes (with CCl4 in vivo) was used to culture the isolated MSCs. The differentiated cells were identified by morphological observation, reverse transcription polymerase chain reaction (RT-PCR), immunofluorescence assay (for AFP, Albumin, and CK18) and periodic acid schiff reaction (PAS) for glycogen. RESULTS: The differentiated cells showed characteristics of hepatocytes. PT-PCR detected AFP mRNA on day 5 and it increased gradually until day 15, and then decreased; CK18 mRNA was detected on day 10; TAT was detected on day 20. Immunofluorescence assay for AFP, albumin and CK18 showed positive staining reactions on day 20. PAS positive glycogen granules appeared in the cytoplasm of the differentiated cells. CONCLUSION: MSCs of adult mice cultured in a conditioned medium of injured hepatocytes can differentiate into hepatocytes. This method can be used in further studying of the mechanism of transdifferentiation of MSCs into hepatocytes.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular , Hepatocitos/citología , Células Madre Mesenquimatosas/citología , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Medios de Cultivo Condicionados , Hígado/patología , Masculino , Ratones , Ratones Endogámicos ICR
11.
World J Gastroenterol ; 16(26): 3267-78, 2010 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-20614482

RESUMEN

AIM: To identify the key cytokines involved in hepatic differentiation of mouse bone marrow mesenchymal stem cells (mBM-MSCs) under liver-injury conditions. METHODS: Abdominal injection of CCl(4) was adopted to duplicate a mouse acute liver injury model. Global gene expression analysis was performed to evaluate the potential genes involved in hepatic commitment under liver-injury conditions. The cytokines involved in hepatic differentiation of mBM-MSCs was functionally examined by depletion experiment using specific antibodies, followed by rescue experiment and direct inducing assay. The hepatic differentiation was characterized by the expression of hepatic lineage genes and proteins, as well as functional features. RESULTS: Cytokines potentially participating in hepatic fate commitment under liver-injury conditions were initially measured by microarray. Among the up-regulated genes determined, 18 cytokines known to closely relate to liver growth, repair and development, were selected for further identification. The fibroblast growth factor-4 (FGF-4), hepatocyte growth factor (HGF) and oncostatin M (OSM) were finally found to be involved in hepatic differentiation of mBM-MSCs under liver-injury conditions. Hepatic differentiation could be dramatically decreased after removing FGF-4, HGF and OSM from the liver-injury conditioned medium, and could be rescued by supplementing these cytokines. The FGF-4, HGF and OSM play different roles in the hepatic differentiation of mBM-MSCs, in which FGF-4 and HGF are essential for the initiation of hepatic differentiation, while OSM is critical for the maturation of hepatocytes. CONCLUSION: FGF-4, HGF and OSM are the key cytokines involved in the liver-injury conditioned medium for the hepatic differentiation of mBM-MSCs.


Asunto(s)
Citocinas/fisiología , Hepatocitos/patología , Hepatocitos/fisiología , Hígado/lesiones , Células Madre Mesenquimatosas/patología , Enfermedad Aguda , Animales , Secuencia de Bases , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/genética , Citocinas/farmacología , Cartilla de ADN/genética , Factor 4 de Crecimiento de Fibroblastos/genética , Factor 4 de Crecimiento de Fibroblastos/farmacología , Factor 4 de Crecimiento de Fibroblastos/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/farmacología , Factor de Crecimiento de Hepatocito/fisiología , Hepatocitos/efectos de los fármacos , Hígado/patología , Hígado/fisiopatología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Oncostatina M/genética , Oncostatina M/farmacología , Oncostatina M/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Regulación hacia Arriba
12.
World J Gastroenterol ; 15(41): 5165-75, 2009 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-19891015

RESUMEN

AIM: To develop a protocol for direct hepatic lineage differentiation from early developmental progenitors to a population of mature hepatocytes. METHODS: Hepatic progenitor cells and then mature hepatocytes from mouse embryonic stem (ES) cells were obtained in a sequential manner, induced by valproic acid (VPA) and cytokines (hepatocyte growth factor, epidermal growth factor and insulin). Morphological changes of the differentiated cells were examined by phase-contrast microscopy and electron microscopy. Reverse transcription polymerase chain reaction and immunocytochemical analyses were used to evaluate the gene expression profiles of the VPA-induced hepatic progenitors and the hepatic progenitor-derived hepatocytes. Glycogen storage, cytochrome P450 activity, transplantation assay, differentiation of bile duct-like structures and tumorigenic analyses were performed for the functional identification of the differentiated cells. Furthermore, FACS and electron microscopy were used for the analyses of cell cycle profile and apoptosis in VPA-induced hepatic differentiated cells. RESULTS: Based on the combination of VPA and cytokines, mouse ES cells differentiated into a uniform and homogeneous cell population of hepatic progenitor cells and then matured into functional hepatocytes. The progenitor population shared several characteristics with ES cells and hepatic stem/progenitor cells, and represented a novel progenitor cell between ES and hepatic oval cells in embryonic development. The differentiated hepatocytes from progenitor cells shared typical characteristics with mature hepatocytes, including the patterns of gene expression, immunological markers, in vitro hepatocyte functions and in vivo capacity to restore acute-damaged liver function. In addition, the differentiation of hepatic progenitor cells from ES cells was accompanied by significant cell cycle arrest and selective survival of differentiating cells towards hepatic lineages. CONCLUSION: Hepatic cells of different developmental stages from early progenitors to matured hepatocytes can be acquired in the appropriate order based on sequential induction with VPA and cytokines.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Hepatocitos/citología , Insulina/farmacología , Ácido Valproico/farmacología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Linaje de la Célula , Células Cultivadas , Sistema Enzimático del Citocromo P-450/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/ultraestructura , Glucógeno/metabolismo , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Ratones , Modelos Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA