Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hepatology ; 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38691396

RESUMEN

The growing recognition of the role of the gut microbiome's impact on alcohol-related diseases, especially in alcohol-associated liver disease, emphasizes the need to understand molecular mechanisms involved in governing organ-organ communication to identify novel avenues to combat alcohol-related diseases. The gut-liver axis refers to the bidirectional communication and interaction between gut and liver. Intestinal microbiota play a pivotal role in maintaining homeostasis within the gut-liver axis and this axis plays a significant role in alcohol-associated liver disease. The intricate communication between intestine and liver involves communication between multiple cellular components in each organ that enable them to carry out their physiological functions. In this review, we focus on novel approaches to understanding how chronic alcohol exposure impacts the microbiome, and individual cells within the liver and intestine, as well as the impact of ethanol on the molecular machinery required for intra- and inter-organ communication.

2.
Cell Mol Gastroenterol Hepatol ; 17(1): 93-106, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37743012

RESUMEN

Liver fibrosis of different etiologies is a serious health problem worldwide. There is no effective therapy available for liver fibrosis except the removal of the underlying cause of injury or liver transplantation. Development of liver fibrosis is caused by fibrogenic myofibroblasts that are not present in the normal liver, but rather activate from liver resident mesenchymal cells in response to chronic toxic or cholestatic injury. Many studies indicate that liver fibrosis is reversible when the causative agent is removed. Regression of liver fibrosis is associated with the disappearance of activated myofibroblasts and resorption of the fibrous scar. In this review, we discuss the results of genetic tracing and cell fate mapping of hepatic stellate cells and portal fibroblasts, their specific characteristics, and potential phenotypes. We summarize research progress in the understanding of the molecular mechanisms underlying the development and reversibility of liver fibrosis, including activation, apoptosis, and inactivation of myofibroblasts.


Asunto(s)
Cirrosis Hepática , Miofibroblastos , Humanos , Miofibroblastos/patología , Cirrosis Hepática/patología , Fibroblastos/patología , Hepatocitos
3.
STAR Protoc ; 4(3): 102391, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37405925

RESUMEN

Here, we present a protocol for isolating human hepatocytes and neural progenitor cells from normal and nonalcoholic steatohepatitis livers. We describe steps for perfusion for scaled-up liver cell isolation and optimization of chemical digestion to achieve maximal yield and cell viability. We then detail a liver cell cryopreservation and potential applications, such as the use of human liver cells as a tool to link experimental and translational research.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Humanos , Células Cultivadas , Hepatocitos , Separación Celular/métodos
4.
Leukemia ; 37(2): 408-421, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36509891

RESUMEN

SOX11 overexpression has been associated with aggressive behavior of mantle cell lymphomas (MCL). SOX11 is overexpressed in embryonic and cancer stem cells (CSC) of some tumors. Although CSC have been isolated from primary MCL, their relationship to SOX11 expression and contribution to MCL pathogenesis and clinical evolution remain unknown. Here, we observed enrichment in leukemic and hematopoietic stem cells gene signatures in SOX11+ compared to SOX11- MCL primary cases. Musashi-2 (MSI2) emerged as one of the most significant upregulated stem cell-related genes in SOX11+ MCLs. SOX11 is directly bound to the MSI2 promoter upregulating its expression in vitro. MSI2 intronic enhancers were strongly activated in SOX11+ MCL cell lines and primary cases. MSI2 upregulation was significantly associated with poor overall survival independently of other high-risk features of MCL. MSI2 knockdown decreased the expression of genes related to apoptosis and stem cell features and significantly reduced clonogenic growth, tumor cell survival and chemoresistance in MCL cells. MSI2-knockdown cells had reduced tumorigenic engraftment into mice bone marrow and spleen compared to control cells in xenotransplanted mouse models. Our results suggest that MSI2 might play a key role in sustaining stemness and tumor cell survival, representing a possible novel target for therapeutic interventions in MCL.


Asunto(s)
Linfoma de Células del Manto , Proteínas de Unión al ARN , Animales , Ratones , Linfoma de Células del Manto/patología , Factores de Transcripción SOXC/genética , Proteínas de Unión al ARN/metabolismo
5.
Blood ; 130(4): 501-513, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28533307

RESUMEN

SOX11 overexpression in mantle cell lymphoma (MCL) has been associated with more aggressive behavior and worse outcome. However, SOX11 oncogenic pathways driving MCL tumor progression are poorly understood. Here, we demonstrate that SOX11 binds to regulatory regions of 2 important genes for microenvironment signals in cancer: (C-X-C motif) chemokine receptor 4 (CXCR4) and PTK2 (encoding for focal adhesion kinase [FAK]). Moreover, SOX11+ xenograft and human primary MCL tumors overexpress cell migration and stromal stimulation gene signatures compared with their SOX11- counterparts. We show that SOX11 directly upregulates CXCR4 and FAK expression, activating PI3K/AKT and ERK1/2 FAK-downstream pathways in MCL. Concordantly, SOX11+ MCL cells have higher cell migration, transmigration through endothelial cells, adhesion to stromal cells, and cell proliferation and display an increased resistance to conventional drug therapies compared with SOX11- MCL cells. Specific FAK inhibition blocks downstream PI3K/AKT- and ERK1/2-mediated phosphorylation. Additionally, specific FAK and PI3K inhibitors reduce SOX11-enhanced MCL cell migration and stromal interactions and revert cell adhesion-mediated drug resistance (CAM-DR) to the same levels as SOX11- MCL cells. In intravenous MCL xenograft models, SOX11+ MCL cells display higher cell migration, invasion, and growth compared with SOX11-knockdown cells, and specific FAK and CXCR4 inhibitors impair SOX11-enhanced MCL engraftment in bone marrow. Overall, our results suggest that SOX11 promotes MCL homing and invasion and increases CAM-DR through the direct regulation of CXCR4 and FAK expression and FAK/PI3K/AKT pathway activation, contributing to a more aggressive phenotype. Inhibition of this pathway may represent an efficient strategy to overcome stromal-mediated chemotherapy refractoriness in aggressive MCL.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Linfoma de Células del Manto/metabolismo , Receptores CCR4/metabolismo , Factores de Transcripción SOXC/metabolismo , Transducción de Señal , Microambiente Tumoral , Animales , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/genética , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/patología , Ratones , Ratones SCID , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores CCR4/genética , Factores de Transcripción SOXC/genética
6.
Blood ; 124(14): 2235-47, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25092176

RESUMEN

SOX11 is overexpressed in several solid tumors and in the vast majority of aggressive mantle cell lymphomas (MCLs). We have recently proven that SOX11 silencing reduces tumor growth in a MCL xenograft model, consistent with the indolent clinical course of the human SOX11-negative mantle cell lymphoma (MCL). However, the direct oncogenic mechanisms and downstream effector pathways implicated in SOX11-driven transformation remain poorly understood. Here, we observed that SOX11-positive xenograft and human primary MCL tumors overexpressed angiogenic gene signatures and had a higher microvascular density compared with their SOX11-negative counterparts. Conditioned media of SOX11-positive MCL cell lines induced in vitro endothelial cell proliferation, migration, tube formation, and activation of downstream angiogenic pathways. We identified PDGFA as a SOX11 direct target gene upregulated in MCL cells whose inhibition impaired SOX11-enhanced in vitro angiogenic effects on endothelial cells. In addition, platelet-derived growth factor A (PDGFA) was overexpressed in SOX11-positive but not in SOX11-negative MCL. In vivo, imatinib impaired tumor angiogenesis and lymphoma growth in SOX11-positive MCL xenograft tumors. Overall, our results demonstrate a prominent role for SOX11 as a driver of proangiogenic signals in MCL, and highlight the SOX11-PDGFA axis as a potential therapeutic target for the treatment of this aggressive disease.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Linfoma de Células del Manto/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Transcripción SOXC/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Medios de Cultivo Condicionados/química , Células Endoteliales/citología , Perfilación de la Expresión Génica , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Neovascularización Patológica , Proteómica , Transducción de Señal , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...