Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cells ; 11(10)2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35626685

RESUMEN

SMER28 (Small molecule enhancer of Rapamycin 28) is an autophagy-inducing compound functioning by a hitherto unknown mechanism. Here, we confirm its autophagy-inducing effect by assessing classical autophagy-related parameters. Interestingly, we also discovered several additional effects of SMER28, including growth retardation and reduced G1 to S phase progression. Most strikingly, SMER28 treatment led to a complete arrest of receptor tyrosine kinase signaling, and, consequently, growth factor-induced cell scattering and dorsal ruffle formation. This coincided with a dramatic reduction in phosphorylation patterns of PI3K downstream effectors. Consistently, SMER28 directly inhibited PI3Kδ and to a lesser extent p110γ. The biological relevance of our observations was underscored by SMER28 interfering with InlB-mediated host cell entry of Listeria monocytogenes, which requires signaling through the prominent receptor tyrosine kinase c-Met. This effect was signaling-specific, since entry of unrelated, gram-negative Salmonella Typhimurium was not inhibited. Lastly, in B cell lymphoma cells, which predominantly depend on tonic signaling through PI3Kδ, apoptosis upon SMER28 treatment is profound in comparison to non-hematopoietic cells. This indicates SMER28 as a possible drug candidate for the treatment of diseases that derive from aberrant PI3Kδ activity.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Serina-Treonina Quinasas TOR , Autofagia , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
2.
JHEP Rep ; 3(3): 100281, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34036256

RESUMEN

BACKGROUND & AIMS: The accumulation of neutral lipids within hepatocytes underlies non-alcoholic fatty liver disease (NAFLD), which affects a quarter of the world's population and is associated with hepatitis, cirrhosis, and hepatocellular carcinoma. Despite insights gained from both human and animal studies, our understanding of NAFLD pathogenesis remains limited. To better study the molecular changes driving the condition we aimed to generate a humanised NAFLD mouse model. METHODS: We generated TIRF (transgene-free Il2rg -/-/Rag2 -/-/Fah -/-) mice, populated their livers with human hepatocytes, and fed them a Western-type diet for 12 weeks. RESULTS: Within the same chimeric liver, human hepatocytes developed pronounced steatosis whereas murine hepatocytes remained normal. Unbiased metabolomics and lipidomics revealed signatures of clinical NAFLD. Transcriptomic analyses showed that molecular responses diverged sharply between murine and human hepatocytes, demonstrating stark species differences in liver function. Regulatory network analysis indicated close agreement between our model and clinical NAFLD with respect to transcriptional control of cholesterol biosynthesis. CONCLUSIONS: These NAFLD xenograft mice reveal an unexpected degree of evolutionary divergence in food metabolism and offer a physiologically relevant, experimentally tractable model for studying the pathogenic changes invoked by steatosis. LAY SUMMARY: Fatty liver disease is an emerging health problem, and as there are no good experimental animal models, our understanding of the condition is poor. We here describe a novel humanised mouse system and compare it with clinical data. The results reveal that the human cells in the mouse liver develop fatty liver disease upon a Western-style fatty diet, whereas the mouse cells appear normal. The molecular signature (expression profiles) of the human cells are distinct from the mouse cells and metabolic analysis of the humanised livers mimic the ones observed in humans with fatty liver. This novel humanised mouse system can be used to study human fatty liver disease.

3.
Nat Commun ; 9(1): 431, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29382845

RESUMEN

Up-Frameshift Suppressor 1 Homolog (UPF1) is a key factor for nonsense-mediated mRNA decay (NMD), a cellular process that can actively degrade mRNAs. Here, we study NMD inhibition during infection by human T-cell lymphotropic virus type I (HTLV-1) and characterise the influence of the retroviral Tax factor on UPF1 activity. Tax interacts with the central helicase core domain of UPF1 and might plug the RNA channel of UPF1, reducing its affinity for nucleic acids. Furthermore, using a single-molecule approach, we show that the sequential interaction of Tax with a RNA-bound UPF1 freezes UPF1: this latter is less sensitive to the presence of ATP and shows translocation defects, highlighting the importance of this feature for NMD. These mechanistic insights reveal how HTLV-1 hijacks the central component of NMD to ensure expression of its own genome.


Asunto(s)
Productos del Gen tax/metabolismo , Interacciones Huésped-Patógeno/fisiología , Degradación de ARNm Mediada por Codón sin Sentido , ARN Helicasas/metabolismo , Transactivadores/metabolismo , Adenosina Trifosfato/metabolismo , Productos del Gen tax/genética , Células HeLa/virología , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Mutación , Dominios Proteicos , Transporte de Proteínas , ARN Helicasas/genética , Transactivadores/genética
4.
JCI Insight ; 3(2)2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29367462

RESUMEN

T cell receptor (TCR) affinity is a critical factor of Treg lineage commitment, but whether self-reactivity is a determining factor in peripheral Treg function remains unknown. Here, we report that a high degree of self-reactivity is crucial for tissue-specific Treg function in autoimmunity. Based on high expression of CD5, we identified a subset of self-reactive Tregs expressing elevated levels of T-bet, GITR, CTLA-4, and ICOS, which imparted significant protection from autoimmune diabetes. We observed that T-bet expression in Tregs, necessary for control of Th1 autoimmunity, could be induced in an IFNγ-independent fashion and, unlike in conventional T cells (Tconv), was strongly correlated with the strength of TCR signaling. The level of CD5 similarly identified human Tregs with an increased functional profile, suggesting that CD5hi Tregs may constitute an efficacious subpopulation appropriate for use in adoptive Treg therapies for treatment of inflammatory conditions. Overall, this work establishes an instrumental role of high TCR self-reactivity in driving Treg function.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/inmunología , Adulto , Animales , Antígenos CD5 , Células Cultivadas , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/terapia , Modelos Animales de Enfermedad , Femenino , Voluntarios Sanos , Humanos , Inmunoterapia Adoptiva/métodos , Islotes Pancreáticos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas de Dominio T Box/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Adulto Joven
5.
J Immunol ; 200(3): 909-914, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29282307

RESUMEN

Regulatory T cells (Tregs) use a distinct TCR repertoire and are more self-reactive compared with conventional T cells. However, the extent to which TCR affinity regulates the function of self-reactive Tregs is largely unknown. In this study, we used a two-TCR model to assess the role of TCR affinity in Treg function during autoimmunity. We observed that high- and low-affinity Tregs were recruited to the pancreas and contributed to protection from autoimmune diabetes. Interestingly, high-affinity cells preferentially upregulated the TCR-dependent Treg functional mediators IL-10, TIGIT, GITR, and CTLA4, whereas low-affinity cells displayed increased transcripts for Areg and Ebi3, suggesting distinct functional profiles. The results of this study suggest mechanistically distinct and potentially nonredundant roles for high- and low-affinity Tregs in controlling autoimmunity.


Asunto(s)
Autoinmunidad/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Anfirregulina/biosíntesis , Animales , Antígeno CTLA-4/biosíntesis , Adhesión Celular/inmunología , Proteína Relacionada con TNFR Inducida por Glucocorticoide/biosíntesis , Interleucina-10/biosíntesis , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Antígenos de Histocompatibilidad Menor/biosíntesis , Páncreas/citología , Páncreas/inmunología , Receptores de Citocinas/biosíntesis , Receptores Inmunológicos/biosíntesis
6.
Nat Commun ; 8(1): 39, 2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28659616

RESUMEN

Only one out of 10 drugs in development passes clinical trials. Many fail because experimental animal models poorly predict human xenobiotic metabolism. Human liver chimeric mice are a step forward in this regard, as the human hepatocytes in chimeric livers generate human metabolites, but the remaining murine hepatocytes contain an expanded set of P450 cytochromes that form the major class of drug-metabolizing enzymes. We therefore generated a conditional knock-out of the NADPH-P450 oxidoreductase (Por) gene combined with Il2rg - /- /Rag2 - /- /Fah - /- (PIRF) mice. Here we show that homozygous PIRF mouse livers are readily repopulated with human hepatocytes, and when the murine Por gene is deleted (<5%), they predominantly use human cytochrome metabolism. When given the anticancer drug gefitinib or the retroviral drug atazanavir, the Por-deleted humanized PIRF mice develop higher levels of the major human metabolites than current models. Humanized, murine Por-deficient PIRF mice can thus predict human drug metabolism and should be useful for preclinical drug development.Human liver chimeric mice are increasingly used for drug testing in preclinical development, but express residual murine p450 cytochromes. Here the authors generate mice lacking the Por gene in the liver, and show that human cytochrome metabolism is used following repopulation with human hepatocytes.


Asunto(s)
Sulfato de Atazanavir/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Quinazolinas/metabolismo , Animales , Antineoplásicos/metabolismo , Quimera , Sistema Enzimático del Citocromo P-450/genética , Citocromos/metabolismo , Femenino , Gefitinib , Genotipo , Inhibidores de la Proteasa del VIH/metabolismo , Humanos , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos
7.
Elife ; 62017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28467300

RESUMEN

The transcription factor TCF7L1 is an embryonic stem cell signature gene that is upregulated in multiple aggressive cancer types, but its role in skin tumorigenesis has not yet been defined. Here we document TCF7L1 upregulation in skin squamous cell carcinoma (SCC) and demonstrate that TCF7L1 overexpression increases tumor incidence, tumor multiplicity, and malignant progression in the chemically induced mouse model of skin SCC. Additionally, we show that downregulation of TCF7L1 and its paralogue TCF7L2 reduces tumor growth in a xenograft model of human skin SCC. Using separation-of-function mutants, we show that TCF7L1 promotes tumor growth, enhances cell migration, and overrides oncogenic RAS-induced senescence independently of its interaction with ß-catenin. Through transcriptome profiling and combined gain- and loss-of-function studies, we identified LCN2 as a major downstream effector of TCF7L1 that drives tumor growth. Our findings establish a tumor-promoting role for TCF7L1 in skin and elucidate the mechanisms underlying its tumorigenic capacity.


Asunto(s)
Carcinogénesis , Carcinoma de Células Escamosas/fisiopatología , Lipocalina 2/metabolismo , Neoplasias Cutáneas/fisiopatología , Proteína 1 Similar al Factor de Transcripción 7/metabolismo , beta Catenina/metabolismo , Animales , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Xenoinjertos , Humanos , Ratones
8.
Nat Commun ; 7: 12642, 2016 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-27572891

RESUMEN

Many metabolic liver disorders are refractory to drug therapy and require orthotopic liver transplantation. Here we demonstrate a new strategy, which we call metabolic pathway reprogramming, to treat hereditary tyrosinaemia type I in mice; rather than edit the disease-causing gene, we delete a gene in a disease-associated pathway to render the phenotype benign. Using CRISPR/Cas9 in vivo, we convert hepatocytes from tyrosinaemia type I into the benign tyrosinaemia type III by deleting Hpd (hydroxyphenylpyruvate dioxigenase). Edited hepatocytes (Fah(-/-)/Hpd(-/-)) display a growth advantage over non-edited hepatocytes (Fah(-/-)/Hpd(+/+)) and, in some mice, almost completely replace them within 8 weeks. Hpd excision successfully reroutes tyrosine catabolism, leaving treated mice healthy and asymptomatic. Metabolic pathway reprogramming sidesteps potential difficulties associated with editing a critical disease-causing gene and can be explored as an option for treating other diseases.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Terapia Genética/métodos , Redes y Vías Metabólicas/genética , Tirosinemias/genética , Animales , Línea Celular , Proliferación Celular/genética , Ciclohexanonas/uso terapéutico , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Exones/genética , Hepatocitos/metabolismo , Humanos , Hidrolasas/genética , Hígado/citología , Hígado/patología , Ratones , Ratones Endogámicos , Ratones Noqueados , Nitrobenzoatos/uso terapéutico , Oxidorreductasas/antagonistas & inhibidores , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Fenotipo , Tirosinemias/metabolismo , Tirosinemias/patología , Tirosinemias/terapia
9.
J Hepatol ; 65(2): 325-33, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27117591

RESUMEN

BACKGROUND & AIMS: Pediatric liver cancer is a rare but serious disease whose incidence is rising, and for which the therapeutic options are limited. Development of more targeted, less toxic therapies is hindered by the lack of an experimental animal model that captures the heterogeneity and metastatic capability of these tumors. METHODS: Here we established an orthotopic engraftment technique to model a series of patient-derived tumor xenograft (PDTX) from pediatric liver cancers of all major histologic subtypes: hepatoblastoma, hepatocellular cancer and hepatocellular malignant neoplasm. We utilized standard (immuno) staining methods for histological characterization, RNA sequencing for gene expression profiling and genome sequencing for identification of druggable targets. We also adapted stem cell culturing techniques to derive two new pediatric cancer cell lines from the xenografted mice. RESULTS: The patient-derived tumor xenografts recapitulated the histologic, genetic, and biological characteristics-including the metastatic behavior-of the corresponding primary tumors. Furthermore, the gene expression profiles of the two new liver cancer cell lines closely resemble those of the primary tumors. Targeted therapy of PDTX from an aggressive hepatocellular malignant neoplasm with the MEK1 inhibitor trametinib and pan-class I PI3 kinase inhibitor NVP-BKM120 resulted in significant growth inhibition, thus confirming this PDTX model as a valuable tool to study tumor biology and patient-specific therapeutic responses. CONCLUSIONS: The novel metastatic xenograft model and the isogenic xenograft-derived cell lines described in this study provide reliable tools for developing mutation- and patient-specific therapies for pediatric liver cancer. LAY SUMMARY: Pediatric liver cancer is a rare but serious disease and no experimental animal model currently captures the complexity and metastatic capability of these tumors. We have established a novel animal model using human tumor tissue that recapitulates the genetic and biological characteristics of this cancer. We demonstrate that our patient-derived animal model, as well as two new cell lines, are useful tools for experimental therapies.


Asunto(s)
Neoplasias Hepáticas , Animales , Carcinoma Hepatocelular , Línea Celular Tumoral , Niño , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Ratones , Trasplante de Neoplasias , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cell ; 162(2): 403-411, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26165941

RESUMEN

Small molecules that interfere with microtubule dynamics, such as Taxol and the Vinca alkaloids, are widely used in cell biology research and as clinical anticancer drugs. However, their activity cannot be restricted to specific target cells, which also causes severe side effects in chemotherapy. Here, we introduce the photostatins, inhibitors that can be switched on and off in vivo by visible light, to optically control microtubule dynamics. Photostatins modulate microtubule dynamics with a subsecond response time and control mitosis in living organisms with single-cell spatial precision. In longer-term applications in cell culture, photostatins are up to 250 times more cytotoxic when switched on with blue light than when kept in the dark. Therefore, photostatins are both valuable tools for cell biology, and are promising as a new class of precision chemotherapeutics whose toxicity may be spatiotemporally constrained using light.


Asunto(s)
Antimitóticos/química , Muerte Celular , Microtúbulos/efectos de los fármacos , Mitosis , Estilbenos/química , Animales , Antimitóticos/toxicidad , Línea Celular Tumoral , Citoesqueleto/química , Humanos , Luz , Ratones , Polimerizacion , Estilbenos/toxicidad
11.
Rev Diabet Stud ; 11(1): 19-34, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25148365

RESUMEN

Recent progress in understanding stem cell biology has been remarkable, especially in deciphering signals that support differentiation towards tissue-specific lineages. This achievement positions us firmly at the beginning of an era of patient-specific regenerative medicine and human disease modeling. It will be necessary to equip the progress in this era with a reliable source of self-renewing progenitor cells that differentiate into functional target cells. The generation of pancreatic progenitors that mature in vivo into functional beta-cells has raised the hope for new therapeutic options in diabetes, but key challenges still remain including the production of sufficient numbers of cells for research and transplantation. Recent approaches to this problem have shown that the presence of organ- and stage-specific mesenchyme improves the generation of progenitors, from endoderm to endocrine cells. Alternatively, utilization of three-dimensional culture may improve the efficiency and yield of directed differentiation. Here, we review the current knowledge of pancreatic directed differentiation and ex vivo expansion of pancreatic progenitors, including recent advances in differentiation strategies for the generation of pancreatic progenitors, and we discuss persistent challenges which will need to be overcome before personalized cell-based therapy becomes a practical strategy.


Asunto(s)
Células Madre Adultas/citología , Diferenciación Celular , Células Madre Embrionarias/citología , Modelos Biológicos , Páncreas/citología , Células Madre Pluripotentes/citología , Animales , Bioingeniería/métodos , Bioingeniería/tendencias , Proliferación Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Cultivo Primario de Células/tendencias
12.
J Virol ; 88(16): 8956-70, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24872589

RESUMEN

Human T-cell leukemia virus types 3 and 4 (HTLV-3 and HTLV-4) are recently isolated retroviruses. We have previously characterized HTLV-3- and HTLV-4-encoded antisense genes, termed APH-3 and APH-4, respectively, which, in contrast to HBZ, the HTLV-1 homologue, do not contain a typical bZIP domain (M. Larocque É Halin, S. Landry, S. J. Marriott, W. M. Switzer, and B. Barbeau, J. Virol. 85:12673-12685, 2011, doi:10.1128/JVI.05296-11). As HBZ differentially modulates the transactivation potential of various Jun family members, the effect of APH-3 and APH-4 on JunD-, c-Jun-, and JunB-mediated transcriptional activation was investigated. We first showed that APH-3 and APH-4 upregulated the transactivation potential of all tested Jun family members. Using an human telomerase catalytic subunit (hTERT) promoter construct, our results also highlighted that, unlike HBZ, which solely modulates hTERT expression via JunD, both APH-3 and APH-4 acted positively on the transactivation of the hTERT promoter mediated by tested Jun factors. Coimmunoprecipitation experiments demonstrated that these Jun proteins interacted with APH-3 and APH-4. Although no activation domain was identified for APH proteins, the activation domain of c-Jun was very important in the observed upregulation of its activation potential. We further showed that APH-3 and APH-4 required their putative bZIP-like domains and corresponding leucine residues for interaction and modulation of the transactivation potential of Jun factors. Our results demonstrate that HTLV-encoded antisense proteins behave differently, and that the bZIP-like domains of both APH-3 and APH-4 have retained their interaction potential for Jun members. These studies are important in assessing the differences between HBZ and other antisense proteins, which might further contribute to determining the role of HBZ in HTLV-1-associated diseases. IMPORTANCE HBZ, the antisense transcript-encoded protein from HTLV-1, is now well recognized as a potential factor for adult T-cell leukemia/lymphoma development. In order to better appreciate the mechanism of action of HBZ, comparison to antisense proteins from other HTLV viruses is important. Little is known in relation to the seemingly nonpathogenic HTLV-3 and HTLV-4 viruses, and studies of their antisense proteins are limited to our previously reported study (M. Larocque É Halin, S. Landry, S. J. Marriott, W. M. Switzer, and B. Barbeau, J. Virol. 85:12673-12685, 2011, doi:10.1128/JVI.05296-11). Here, we demonstrate that Jun transcription factors are differently affected by APH-3 and APH-4 compared to HBZ. These intriguing findings suggest that these proteins act differently on viral replication but also on cellular gene expression, and that highlighting their differences of action might lead to important information allowing us to understand the link between HTLV-1 HBZ and ATL in infected individuals.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , ADN sin Sentido/genética , Virus Linfotrópico T Tipo 3 Humano/genética , Virus Linfotrópico T Tipo 3 Humano/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Activación Transcripcional/genética , Animales , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Deltaretrovirus/genética , Deltaretrovirus/metabolismo , Células HEK293 , Células HeLa , Humanos , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-jun/genética , Telomerasa/genética , Telomerasa/metabolismo , Transcripción Genética/genética , Regulación hacia Arriba/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
13.
Carcinogenesis ; 34(11): 2664-72, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23784080

RESUMEN

Telomerase activity in cancer cells is dependent on the transcriptional regulation of the human telomerase reverse transcriptase (hTERT) gene, encoding the catalytic subunit of human telomerase. We have shown previously that HTLV-1 basic leucine zipper (HBZ), a viral regulatory protein encoded by the human retrovirus, human T-cell leukemia virus, type 1 (HTLV-1) cooperates with JunD to enhance hTERT transcription in adult T-cell leukemia (ATL) cells. Menin, the product of the tumor-suppressor MEN-1 gene, also interacts with JunD, represses its transcriptional activity and downregulates telomerase expression. The main objective of this study was to examine how menin and HBZ get involved in the regulation of hTERT transcription. In this study, we report that JunD and menin form a repressor complex of hTERT transcription in HBZ-negative cells. Conversely, in HBZ-positive cells, the formation of a JunD/HBZ/menin ternary complex and the recruitment of p300 histone acetyl transferase activity by HBZ lead to a decreased activity of the JunD-menin suppressor unit that correlates with the activation of hTERT transcription. Silencing HBZ or menin expression in ATL cells confirms that these proteins are differentially involved in telomerase regulation. These results propose that HBZ, by impeding the tumor-suppressor activity of menin, functions as a leukemogenic cofactor to upregulate gene transcription and promote JunD-mediated leukemogenesis.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Regulación Neoplásica de la Expresión Génica , Leucemia de Células T/patología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Telomerasa/genética , Proteínas Virales/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/antagonistas & inhibidores , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Western Blotting , Proliferación Celular , Inmunoprecipitación de Cromatina , Proteína p300 Asociada a E1A/genética , Células HeLa , Humanos , Inmunoprecipitación , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-jun/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas de los Retroviridae , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telomerasa/metabolismo , Transcripción Genética , Células Tumorales Cultivadas , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética
14.
Development ; 140(3): 675-86, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23293299

RESUMEN

Embryonic stem (ES) cells hold great promise with respect to their potential to be differentiated into desired cell types. Of interest are organs derived from the definitive endoderm, such as the pancreas and liver, and animal studies have revealed an essential role for Nodal in development of the definitive endoderm. Activin A is a related TGFß member that acts through many of the same downstream signaling effectors as Nodal and is thought to mimic Nodal activity. Detailed characterization of ES cell-derived endodermal cell types by gene expression analysis in vitro and functional analysis in vivo reveal that, despite their similarity in gene expression, Nodal and Activin-derived endodermal cells exhibit a distinct difference in functional competence following transplantation into the developing mouse embryo. Pdx1-expressing cells arising from the respective endoderm populations exhibit extended differences in their competence to mature into insulin/c-peptide-expressing cells in vivo. Our findings underscore the importance of functional cell-type evaluation during stepwise differentiation of stem cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Endodermo/citología , Proteína Nodal/metabolismo , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Cultivadas , Medios de Cultivo/metabolismo , Técnicas de Cultivo de Embriones , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Endodermo/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Humanos , Subunidades beta de Inhibinas/metabolismo , Subunidades beta de Inhibinas/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Proteína Nodal/farmacología , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo
15.
Nature ; 491(7426): 765-8, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23041930

RESUMEN

One goal of regenerative medicine, to use stem cells to replace cells lost by injury or disease, depends on producing an excess of the relevant cell for study or transplantation. To this end, the stepwise differentiation of stem cells into specialized derivatives has been successful for some cell types, but a major problem remains the inefficient conversion of cells from one stage of differentiation to the next. If specialized cells are to be produced in large numbers it will be necessary to expand progenitor cells, without differentiation, at some steps of the process. Using the pancreatic lineage as a model for embryonic-stem-cell differentiation, we demonstrate that this is a solvable problem. Co-culture with organ-matched mesenchyme permits proliferation and self-renewal of progenitors, without differentiation, and enables an expansion of more than a million-fold for human endodermal cells with full retention of their developmental potential. This effect is specific both to the mesenchymal cell and to the progenitor being amplified. Progenitors that have been serially expanded on mesenchyme give rise to glucose-sensing, insulin-secreting cells when transplanted in vivo. Theoretically, the identification of stage-specific renewal signals can be incorporated into any scheme for the efficient production of large numbers of differentiated cells from stem cells and may therefore have wide application in regenerative biology.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Madre Embrionarias/citología , Endodermo/citología , Mesodermo/citología , Páncreas/citología , Animales , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Glucosa/farmacología , Humanos , Insulina/metabolismo , Masculino , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo
16.
Rev Diabet Stud ; 7(2): 93-104, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21060968

RESUMEN

Diabetic patients suffer from the loss of insulin-secreting ß-cells, or from an improper working ß-cell mass. Due to the increasing prevalence of diabetes across the world, there is a compelling need for a renewable source of cells that could replace pancreatic ß-cells. In recent years, several promising approaches to the generation of new ß-cells have been developed. These include directed differentiation of pluripotent cells such as embryonic stem (ES) cells or induced pluripotent stem (iPS) cells, or reprogramming of mature tissue cells. High yield methods to differentiate cell populations into ß-cells, definitive endoderm, and pancreatic progenitors, have been established using growth factors and small molecules. However, the final step of directed differentiation to generate functional, mature ß-cells in sufficient quantities has yet to be achieved in vitro. Beside the needs of transplantation medicine, a renewable source of ß-cells would also be important in terms of a platform to study the pathogenesis of diabetes, and to seek alternative treatments. Finally, by generating new ß-cells, we could learn more details about pancreatic development and ß-cell specification. This review gives an overview of pancreas ontogenesis in the perspective of stem cell differentiation, and highlights the critical aspects of small molecules in the generation of a renewable ß-cell source. Also, it discusses longer term challenges and opportunities in moving towards a therapeutic goal for diabetes.


Asunto(s)
Diferenciación Celular , Células Secretoras de Insulina/citología , Páncreas/metabolismo , Células Madre/citología , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Secretoras de Insulina/metabolismo , Páncreas/citología , Páncreas/embriología , Páncreas/crecimiento & desarrollo , Hormonas Pancreáticas/genética , Hormonas Pancreáticas/metabolismo , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
J Virol ; 83(18): 9596-601, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19587051

RESUMEN

Ebolavirus (EBOV) is the etiological agent of a severe hemorrhagic fever with a high mortality rate. The spike glycoprotein (GP) is believed to be one of the major determinants of virus pathogenicity. In this study, we demonstrated the molecular mechanism responsible for the downregulation of surface markers caused by EBOV GP expression. We showed that expression of mature GP on the plasma membrane results in the masking of cellular surface proteins, including major histocompatibility complex class I. Overexpression of GP also results in the masking of certain antigenic epitopes on GP itself, causing an illusory effect of disappearance from the plasma membrane.


Asunto(s)
Ebolavirus/patogenicidad , Epítopos , Proteínas de la Membrana , Proteínas del Envoltorio Viral/fisiología , Línea Celular , Membrana Celular/virología , Regulación hacia Abajo , Ebolavirus/química , Antígenos de Histocompatibilidad Clase I , Humanos , Transfección , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
18.
Nat Chem Biol ; 5(4): 258-65, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19287398

RESUMEN

Stepwise differentiation from embryonic stem cells (ESCs) to functional insulin-secreting beta cells will identify key steps in beta-cell development and may yet prove useful for transplantation therapy for diabetics. An essential step in this schema is the generation of pancreatic progenitors--cells that express Pdx1 and produce all the cell types of the pancreas. High-content chemical screening identified a small molecule, (-)-indolactam V, that induces differentiation of a substantial number of Pdx1-expressing cells from human ESCs. The Pdx1-expressing cells express other pancreatic markers and contribute to endocrine, exocrine and duct cells, in vitro and in vivo. Further analyses showed that (-)-indolactam V works specifically at one stage of pancreatic development, inducing pancreatic progenitors from definitive endoderm. This study describes a chemical screening platform to investigate human ESC differentiation and demonstrates the generation of a cell population that is a key milepost on the path to making beta cells.


Asunto(s)
Carcinógenos/farmacología , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Indoles/farmacología , Células Secretoras de Insulina/citología , Lactamas/farmacología , Animales , Biomarcadores , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Madre Embrionarias/fisiología , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Transactivadores/metabolismo
19.
Gastroenterology ; 137(1): 297-308, 308.e1-4, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19208365

RESUMEN

BACKGROUND & AIMS: The hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (c-Met) system is an essential inducer of hepatocyte growth and proliferation. Although a fundamental role for the HGF receptor c-Met has been shown in acute liver regeneration, its cell-specific role in hepatocytes during chronic liver injury and fibrosis progression has not been determined. METHODS: Hepatocyte-specific c-Met knockout mice (c-Met(Delta hepa)) using the Cre-loxP system were studied in a bile duct ligation (BDL) model. Microarray analyses were performed to define HGF/c-Met-dependent gene expression. RESULTS: Two strategies for c-Met deletion in hepatocytes to generate hepatocyte-specific c-Met knockout mice were tested. Early deletion during embryonic development was lethal, whereas post-natal Cre expression was successful, leading to the generation of viable c-Met(Delta hepa) mice. BDL in these mice resulted in extensive necrosis and lower proliferation rates of hepatocytes. Gene array analysis of c-Met(Delta hepa) mice revealed a significant reduction of anti-apoptotic genes in c-Met-deleted hepatocytes. These findings could be tested functionally because c-Met(Delta hepa) mice showed a stronger apoptotic response after BDL and Jo-2 stimulation. The phenotype was associated with increased expression of proinflammatory cytokines (tumor necrosis factor-alpha and interleukin-6) and an enhanced recruitment of neutrophils. Activation of these mechanisms triggered a stronger profibrogenic response as evidenced by increased transforming growth factor-beta(1), alpha-smooth muscle actin, collagen-1alpha messenger RNA expression, and enhanced collagen-fiber staining in c-Met(Delta hepa) mice. CONCLUSIONS: Our results show that deletion of c-Met in hepatocytes leads to more liver cell damage and fibrosis in a chronic cholestatic liver injury model because c-Met triggers survival signals important for hepatocyte recovery.


Asunto(s)
Apoptosis , Colestasis Extrahepática/complicaciones , Cirrosis Hepática/prevención & control , Hígado/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Apoptosis/genética , Proliferación Celular , Colestasis Extrahepática/genética , Colestasis Extrahepática/metabolismo , Colestasis Extrahepática/patología , Enfermedad Crónica , Conducto Colédoco/cirugía , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica/métodos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hepatitis/metabolismo , Hepatitis/patología , Factor de Crecimiento de Hepatocito/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Mediadores de Inflamación/metabolismo , Ligadura , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Ratones , Ratones Noqueados , Necrosis , Infiltración Neutrófila , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-met/deficiencia , Proteínas Proto-Oncogénicas c-met/genética , Factores de Tiempo
20.
Int J Cancer ; 124(8): 1767-72, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19123478

RESUMEN

The receptor for the hepatocyte growth factor/scatter factor (HGF/SF), c-Met, plays a role in tumour promotion, progression and metastasis. In this study, we analysed chemically induced hepatocarcinogenesis in mice lacking a functional HGF receptor in their liver. Control and c-Met deficient mice were injected with a single dose of N-nitrosodiethylamine (DEN, 90 mICROg/g b.wt.) at 6 weeks of age and mice were subsequently kept on a phenobarbital (PB) containing diet (0.05%) for 35 weeks or on control diet. At the end of the experiment, the carcinogenic response in liver of the animals was monitored. Conditional c-met knockout (KO) mice showed a higher prevalence of macroscopically visible liver tumours and of glutamine synthetase positive and glucose-6-phosphatase deficient lesions in liver. Tumour promotion by PB led to significant increases in the number of preneoplastic and neoplastic lesions in liver of both wild-type and c-met knockout mice, with only minor differences in response. Our results indicate that a defect in c-Met-mediated signaling increases chemically induced tumour initiation in liver but does not significantly affect PB-mediated tumour promotion.


Asunto(s)
Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Dietilnitrosamina/farmacología , Glucosa-6-Fosfatasa/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas/inducido químicamente , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos , Fenobarbital/toxicidad , Fenotipo , Transducción de Señal , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA