Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Exp Cell Res ; 386(2): 111732, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31770531

RESUMEN

Acquisition of features of mesenchymal cells represents a key step of metastatic progression of cancer cells and searching for mechanisms underlying the acquisition will help design novel clinical strategies for suppressing the metastatic progression. The Deleted in Liver Cancer-1 (DLC-1) gene is a p122/RhoGAP tumor/metastatic suppressor gene. However, the mechanism underlying DLC-1's inhibition of metastasis still remains largely unknown. In this study, we revealed that the DLC-1-deficient, but not the DLC-1-competent, human non-small cell lung carcinoma cells (NSCLCs) could acquire the TGF-ß1-induced expression of CD105, a common surface marker of mesenchymal stem cells, with consequent increase in CD105-associated cell motility. Interestingly, the induced CD105 expression and cell motility were subjected to the inhibition by the DLC-1-RhoA-Rock1 signaling through inhibiting the serine phosphorylation at a linker region, but not at the C-terminus, of the Smad3 protein and Smad3 protein nuclear translocation down the canonical TGF-ß1 signaling. In addition, the evidence suggested that DLC-1 very likely exerted its inhibitory effects on the TGF-ß1 signaling and the associated CD105 acquisition in both the cytoplasm and the nucleus. Consistent to the in vitro findings, a reverse correlation between CD105 and DLC-1 in protein expression was identified in primary NSCLC tissues and their surrounding non-tumor tissues. In summary, this study revealed a novel anti-metastasis mechanism governed by the DLC-1 tumor/metastasis suppressor, thus helping design new diagnostic and therapeutic approaches for NSCLCs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Endoglina/genética , Proteínas Activadoras de GTPasa/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Factor de Crecimiento Transformador beta1/genética , Proteínas Supresoras de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular , Núcleo Celular/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Citosol/metabolismo , Endoglina/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metástasis Linfática , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fosforilación , Transporte de Proteínas , Transducción de Señal , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
2.
BMC Cancer ; 20(1): 1064, 2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33148199

RESUMEN

BACKGROUND: Immunomodulatory activities of human mesenchymal stromal /stem cells (hMSCs) has been widely recognized as the most critical function of hMSCs for exerting its therapeutic effects. However, the detailed mechanisms responsible for regulating the immunomodulation of hMSCs still remain largely unknown. Previous studies revealed that the Notch1 protein exerted a pro-immunomodulatory function probably through interacting with the protein(s) subjective to proteasome-mediated protein degradation. The DLC-1 protein represents a well characterized tumor suppressor subjective to proteasome-mediated degradation. However, the detailed signaling pathway of Notch1 and the involvement of DLC-1 in regulating the immunomodulation of hMSCs have not been studied before. METHODS: The transfection with cDNA or siRNA into hMSCs assisted by co-culture of hMSCs with peripheral blood mononuclear cells and small molecule inhibitors of signaling proteins, followed by immunoprecipitation, Western blotting, RT-PCR, and flowcytometry, were employed to characterize the Notch1 signaling, to identify DLC-1 as a candidate proteasome-targeted protein, and to characterize DLC-1 signaling pathway and its interaction with the Notch1 signaling, in the regulation of immunomodulation of hMSCs, specifically, the inhibition of pro-inflammatory CD4+-Th1 lymphocytes, and the release of immunomodulatory molecule IDO1. STATISTICAL ANALYSIS: One-way ANOVA was utilized as a statistical tool to analyze the data presented as means ± SEM of at least three separate experiments. RESULTS: The present study revealed that the Notch1-Hey1 axis, but not the Notch1-Hes1 axis, was likely responsible for mediating the pro-immunomodulatory function of the Notch1 signaling. The DLC-1 protein was found subjective to proteasome-mediated protein degradation mediated by the DDB1 and FBXW5 E3 ligases and served as an inhibitor of the immunomodulation of hMSCs through inhibiting Rock1, but not Rock2, downstream the DLC-1 signaling. The Notch1 signaling in the Notch1-Hey1 pathway and the DLC-1 signaling in the DLC-1-Rock1-FBXW5 pathway exhibited a mutual exclusion interaction in the regulation of immunomodulation of hMSCs. CONCLUSIONS: The present study uncovers a novel function of DLC-1 tumor suppressor in regulating the immunomodulation of hMSCs. It also proposes a novel mutual exclusion mechanism between the DLC-1 signaling and the Notch1 signaling that is possibly responsible for fine-tuning the immunomodulation of hMSCs with different clinical implications in hMSCs therapy.


Asunto(s)
Proteínas F-Box/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Inmunomodulación , Leucocitos Mononucleares/inmunología , Células Madre Mesenquimatosas/inmunología , Receptor Notch1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Quinasas Asociadas a rho/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Proteínas F-Box/genética , Proteínas Activadoras de GTPasa/genética , Genes Supresores de Tumor , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptor Notch1/genética , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Quinasas Asociadas a rho/genética
3.
Biologicals ; 56: 67-83, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30150108

RESUMEN

Sessions included an overview of past cell therapy (CT) conferences sponsored by the International Alliance for Biological Standardization (IABS). The sessions highlighted challenges in the field of human pluripotent stem cells (hPSCs) and also addressed specific points on manufacturing, bioanalytics and comparability, tumorigenicity testing, storage, and shipping. Panel discussions complemented the presentations. The conference concluded that a range of new standardization groups is emerging that could help the field, but ways must be found to ensure that these efforts are coordinated. In addition, there are opportunities for regulatory convergence starting with a gap analysis of existing guidelines to determine what might be missing and what issues might be creating divergence. More specific global regulatory guidance, preferably from WHO, would be welcome. IABS and the California Institute for Regenerative Medicine (CIRM) will explore with stakeholders the development of a practical and innovative road map to support early CT product (CTP) developers.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Células Madre Pluripotentes , Pruebas de Carcinogenicidad , Guías como Asunto , Humanos , Control de Calidad , Medicina Regenerativa
4.
BMC Cancer ; 13: 140, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23517112

RESUMEN

BACKGROUND: Malignant pleural mesothelioma (MPM) is an aggressive malignancy closely associated with asbestos exposure and extremely resistant to current treatments. It exhibits a steady increase in incidence, thus necessitating an urgent development of effective new treatments. METHODS: Proteasome inhibitors (PIs) and TNFα-Related Apoptosis Inducing Ligand (TRAIL), have emerged as promising new anti-MPM agents. To develop effective new treatments, the proapoptotic effects of PIs, MG132 or Bortezomib, and TRAIL were investigated in MPM cell lines NCI-H2052, NCI-H2452 and NCI-H28, which represent three major histological types of human MPM. RESULTS: Treatment with 0.5-1 µM MG132 alone or 30 ng/mL Bortezomib alone induced a limited apoptosis in MPM cells associated with the elevated Mcl-1 protein level and hyperactive PI3K/Akt signaling. However, whereas 10-20 ng/ml TRAIL alone induced a limited apoptosis as well, TRAIL and PI combination triggered a robust apoptosis in all three MPM cell lines. The robust proapoptotic activity was found to be the consequence of a positive feedback mechanism-governed amplification of caspase activation and cleavage of both Mcl-1 and Akt proteins, and exhibited a relative selectivity in MPM cells than in non-tumorigenic Met-5A mesothelial cells. CONCLUSION: The combinatorial treatment using TRAIL and PI may represent an effective new treatment for MPMs.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Pulmonares/patología , Mesotelioma/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Neoplasias Pleurales/patología , Inhibidores de Proteasoma/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Citometría de Flujo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Mesotelioma/tratamiento farmacológico , Mesotelioma/genética , Mesotelioma Maligno , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/antagonistas & inhibidores , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Neoplasias Pleurales/tratamiento farmacológico , Neoplasias Pleurales/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
5.
BMC Cancer ; 12: 418, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22994684

RESUMEN

BACKGROUND: The role of Th17 cells in colorectal tumorigenesis and development still remains unclear, despite the fact that it has been established in the pathogenesis of autoimmune diseases. METHODS: We first analyzed Th17 cells and Treg cells using flow cytometry in the circulation of colorectal adenoma (CRA) and colorectal carcinoma (CRC) patients and healthy controls, and the frequency of Th17 cells in peripheral blood mononuclear cells (PBMCs) stimulated by anti-CD3 plus anti-CD28 and treated by IL-1ß, IL-6, and TGF-ß in different concentrations. We then detected cytokines IL-1ß, IL-6, IL-17A, IL-21, IL-23 or TGF-ß by ELISA in sera and supernatants from both normal and tumor tissues cultured ex vivo. RESULTS: It was found that the percentage of Th17 and Treg cells increased in the circulation of both CRA and CRC patients; the increase of Th17 cells in the circulation occurred in early stages, whereas the increase of Treg cells in the circulation and the increase of Th17 cells in tumor tissues occurred in advanced stages. The subsequent cytokine profiling showed that, along CRC progression, IL-1ß, IL-17A and IL-23 underwent a similar change, while IL-6 in CRC exhibited an opposite change, with Th17 cells. In addition, high levels of TGF-ß and IL-17A were detected in tumor tissues rather than in normal mucosa. The in vitro experiment further demonstrated that IL-1ß, IL-6 or TGF-ß modulated Th17 cell expansion in PBMC. CONCLUSIONS: Our study reveals a unique change of Th17 cells, which is regulated possibly by IL-1ß, IL-6 and TGF-ß in the progression of CRC.


Asunto(s)
Adenoma/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Citocinas/metabolismo , Células Th17/inmunología , Adenoma/metabolismo , Adenoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma/inmunología , Carcinoma/metabolismo , Carcinoma/patología , Neoplasias Colorrectales/patología , Citocinas/sangre , Citocinas/farmacología , Progresión de la Enfermedad , Femenino , Humanos , Inmunofenotipificación , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th17/efectos de los fármacos , Células Th17/metabolismo
6.
PLoS One ; 15(1): e0227174, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31900469

RESUMEN

BEAS-2B was originally established as an immortalized but non-tumorigenic epithelial cell line from human bronchial epithelium. Because of general recognition for its bronchial epithelial origin, the BEAS-2B cell line has been widely used as an in vitro cell model in a large variety of studies associated with respiratory diseases including lung carcinogenesis. However, very few studies have discussed non-epithelial features of BEAS-2B cells, especially the features associated with mesenchymal stem cells (MSCs), which represent a group of fibroblast-like cells with limited self-renewal and differentiation potential to various cell lineages. In this study, we compared BEAS-2B with a human umbilical cord-derived MSCs (hMSCs) cell line, hMSC1, which served as a representative of hMSCs in terms of expressing common features of hMSCs. It was observed that both BEAS-2B and hMSC1 shared the same expression profile of surface markers of hMSCs and exhibited similar osteogenic and adipogenic differentiation potential. In addition, like hMSC1, the BEAS-2B cell line exhibited suppressive activities on proliferation of mitogen-activated total T lymphocytes as well as Th1 lymphocytes, and IFNγ-induced expression of IDO1, all thus demonstrating that BEAS-2B cells exhibited an almost identical characteristic profile with hMSCs, even though, there was a clear difference between BEAS-2B and hMSCs in the effects on type 2 macrophage polarization. Most importantly, the hMSCs features of BEAS-2B were unlikely a consequence of epithelial-mesenchymal transition. Therefore, this study provided a set of evidence to provoke reconsideration of epithelial origin of BEAS-2B.


Asunto(s)
Bronquios/citología , Células Epiteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Cordón Umbilical/citología , Células A549 , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Antígenos de Superficie/metabolismo , Carcinogénesis/metabolismo , Diferenciación Celular , Polaridad Celular/fisiología , Proliferación Celular , Técnicas de Cocultivo , Transición Epitelial-Mesenquimal , Femenino , Xenoinjertos , Humanos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células TH1/metabolismo
7.
PLoS One ; 12(8): e0182961, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28806404

RESUMEN

The stimulator of interferon genes (STING) protein has emerged as a critical signal transduction molecule in the innate immune response. Sustained activation of the STING signaling induced by cytosolic DNA has been considered to be the cause of a variety of autoimmune diseases characterized by uncontrolled inflammation. Therefore, it is important to understand the molecular basis of the regulation of STING signaling pathway. Here we demonstrate that the STING protein undergoes a proteasome-mediated degradation in human diploid cell (HDC) lines including MRC-5 following the transfection of double-stranded DNA (dsDNA). The degradation of STING is accompanied by the increased expression of both RIG-I and IL-6. Employing the RIG-I siRNA knockdown and an IL-6 neutralizing antibody greatly inhibits the degradation of STING induced by dsDNA. We further demonstrate that both IL-6 and RIG-I are downstream molecules of STING along the DNA sensor pathway. Therefore, STING degradation mediated by RIG-I and IL-6 may serve as a negative feedback mechanism to limit the uncontrolled innate immune response induced by dsDNA. We have further shown that RIG-I and IL-6 promote STING degradation by activating/dephosphorylating UNC-51-like kinase (ULK1). Interestingly, the STING protein is not significantly affected by dsDNA in non-HDC HEK293 cells. Our study thus has identified a novel signaling pathway for regulating STING in HDCs.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , ADN/metabolismo , Interleucina-6/metabolismo , Proteínas de la Membrana/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Retroalimentación Fisiológica , Células HEK293 , Humanos , Interferón beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Plásmidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Receptores Inmunológicos , Replicación Viral
8.
Sci Rep ; 7(1): 12484, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28970485

RESUMEN

MRC-5 represents the most frequent human diploid cells (HDCs)-type cell substrate in the production of human viral vaccines. However, early-passage MRC-5 is diminishing and, due to both technical and ethical issues, it is extremely difficult to derive novel HDCs from fetal lung tissues, which are the common sources of HDCs. Our previous studies suggested that human umbilical cord may represent an alternative but convenient source of new HDCs. Here, we established a three-tiered cell banking system of a hUC-MSC line, designated previously as Cell Collection and Research Center-1 (CCRC-1). The full characterization indicated that the banked CCRC-1 cells were free from adventitious agents and remained non-tumorigenic. The CCRC-1 cells sustained its rapid proliferation even at passage 30 and were susceptible to the infection of a wide spectrum of viruses. Interestingly, the CCRC-1 cells showed much higher production of EV71 or Rubella viruses than MRC-5 and Vero cells when growing in serum-free medium. More importantly, the EV71 vaccine produced from CCRC-1 cells induced immunogenicity while eliciting no detectable toxicities in the tested mice. Collectively, these studies further supported that CCRC-1, and likely other hUC-MSCs as well, may serve as novel, safe and high-yielding HDCs for the production of human viral vaccines.


Asunto(s)
Infecciones por Enterovirus/prevención & control , Células Madre Mesenquimatosas/virología , Rubéola (Sarampión Alemán)/prevención & control , Vacunación , Vacunas Virales/biosíntesis , Animales , Bancos de Muestras Biológicas , Línea Celular , Proliferación Celular , Chlorocebus aethiops , Medio de Cultivo Libre de Suero/química , Diploidia , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/virología , Femenino , Sangre Fetal/citología , Humanos , Inmunogenicidad Vacunal , Células Madre Mesenquimatosas/citología , Ratones , Ratones Desnudos , Rubéola (Sarampión Alemán)/inmunología , Rubéola (Sarampión Alemán)/virología , Virus de la Rubéola/efectos de los fármacos , Virus de la Rubéola/inmunología , Células Vero , Vacunas Virales/administración & dosificación
9.
Stem Cells Transl Med ; 6(11): 1956-1962, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29067781

RESUMEN

This article summarizes the recent activity of the International Stem Cell Banking Initiative (ISCBI) held at the California Institute for Regenerative Medicine (CIRM) in California (June 26, 2016) and the Korean National Institutes for Health in Korea (October 19-20, 2016). Through the workshops, ISCBI is endeavoring to support a new paradigm for human medicine using pluripotent stem cells (hPSC) for cell therapies. Priority considerations for ISCBI include ensuring the safety and efficacy of a final cell therapy product and quality assured source materials, such as stem cells and primary donor cells. To these ends, ISCBI aims to promote global harmonization on quality and safety control of stem cells for research and the development of starting materials for cell therapies, with regular workshops involving hPSC banking centers, biologists, and regulatory bodies. Here, we provide a brief overview of two such recent activities, with summaries of key issues raised. Stem Cells Translational Medicine 2017;6:1956-1962.


Asunto(s)
Bancos de Muestras Biológicas/normas , Células Madre Embrionarias Humanas/citología , Investigación con Células Madre , Bancos de Muestras Biológicas/organización & administración , Congresos como Asunto , Humanos , Cooperación Internacional
10.
Oncogene ; 24(31): 4873-82, 2005 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-15897898

RESUMEN

Environmental or occupational exposure to mineral dusts, mainly silica and asbestos, is associated with an increased incidence of lung inflammation, fibrosis, and/or cancer. To better understand the molecular events associated with these pulmonary diseases, we attempted to identify genes that are regulated by mineral dusts. Using a differential display reverse transcription polymerase chain reaction technique and mRNAs of alveolar macrophages from both normal individuals and coal miners, we identified a novel mineral dust-induced gene named mdig, which had not been fully characterized. The expression of mdig mRNA was detected in alveolar macrophages from coal miners but not from normal subjects. The inducible expression of mdig could be observed in A549 cells exposed to silica particles in a time-dependent manner. The full-length mdig mRNA was expressed in human lung cancer tissues but was barely detectable in the adjacent normal tissues. In addition, a number of lung cancer cell lines constitutively express mdig. Alternative spliced transcripts of mdig were detected in some lung cancer cell lines. Silencing mdig mRNA expression in A549 lung cancer cells by siRNA-mediated RNA interference inhibits cell proliferation and sensitizes the cells to silica-induced cytotoxicity. These results suggest that the mdig gene may be involved in the regulation of cell growth and possibly the development of cancer.


Asunto(s)
Minas de Carbón , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , ARN Mensajero/genética , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , División Celular , Línea Celular Tumoral , Dioxigenasas , Exones/genética , Histona Demetilasas , Humanos , Neoplasias Pulmonares/patología , Macrófagos Alveolares/fisiología , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , Valores de Referencia , Dióxido de Silicio/toxicidad , Transcripción Genética
11.
Oncogene ; 22(3): 445-50, 2003 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-12545165

RESUMEN

The human DLC-1 (deleted in liver cancer 1) gene was cloned from a primary human hepatocellular carcinoma (HCC) and mapped to the chromosome 8p21-22 region frequently deleted in common human cancers and suspected to harbor tumor suppressor genes. DLC-1 was found to be deleted or downregulated in a significant number of HCCs. We expanded our investigations to other cancers with recurrent deletions of 8p22, and in this study examined alterations of DLC-1 in primary human breast tumors, human breast, colon, and prostate tumor cell lines. Genomic deletion of DLC-1 was observed in 40% of primary breast tumors, whereas reduced or undetectable levels of DLC-1 mRNA were seen in 70% of breast, 70% of colon, and 50% of prostate tumor cell lines To see whether DLC-1 expression affects cell growth and tumorigenicity, two breast carcinoma cell lines lacking the expression of endogenous gene were transfected with the DLC-1 cDNA. In both cell lines, DLC-1 transfection caused significant growth inhibition and reduction of colony formation. Furthermore, introduction of the DLC-1 cDNA abolished the in vivo tumorigenicity in nude mice, suggesting that the DLC-1 gene plays a role in breast cancer by acting as a bona fide tumor suppressor gene.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas Supresoras de Tumor/genética , Adenocarcinoma/genética , Animales , Pruebas de Carcinogenicidad/métodos , División Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Femenino , Proteínas Activadoras de GTPasa , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Endogámicos BALB C , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Valores de Referencia , Transfección , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo
12.
Oncogene ; 23(7): 1405-11, 2004 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-14661059

RESUMEN

The deleted in liver cancer (DLC-1) gene at chromosome 8p21-22 is altered mainly by genomic deletion or aberrant promoter methylation in a large number of human cancers such as breast, liver, colon and prostate and is known to have an inhibitory effect on breast and liver tumor cell growth. Given the high frequency of deletion involving region 8p21-22 in human non-small cell lung carcinoma (NSCLC), we examined alterations of DLC-1 in a series of primary tumors and tumor cell lines and tested effects of DLC-1 on tumor cell growth. A significant decrease or absence of the DLC-1 mRNA expression was found in 95% of primary NSCLC (20/21) and 58% of NSCLC cell lines (11/19). Transcriptional silencing of DLC-1 was primarily associated with aberrant DNA methylation, rather than genomic deletion as 5-aza-2'-deoxycytidine induced reactivation of DLC-1 expression in 82% (9/11) NSCLC cell lines showing downregulated DLC-1. It was further evidenced by an aberrant DLC-1 promoter methylation pattern, which was detected by Southern blotting in 73% (8/11) of NSCLC cell lines with downregulation of the gene. The transfer of DLC-1 into three DLC-1 negative cell lines caused a significant inhibition in cell proliferation and/or a decrease in colony formation. Furthermore, stable transfer of DLC-1 abolished tumorigenicity in nude mice of two cell lines, suggesting that DLC-1 plays a role in NSCLC by acting as a bona fide new tumor suppressor gene.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares , Proteínas Supresoras de Tumor/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Metilación de ADN , Proteínas Activadoras de GTPasa , Humanos , Immunoblotting , Ratones , Ratones Desnudos , Regiones Promotoras Genéticas , Células Tumorales Cultivadas/trasplante , Proteínas Supresoras de Tumor/metabolismo
13.
FEBS Lett ; 579(5): 1191-6, 2005 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-15710412

RESUMEN

DLC-1 (deleted in liver cancer 1) is a Rho GTPase-activating protein that is able to inhibit cell growth and suppress tumorigenesis. We have used homologous recombination to inactivate the mouse DLC-1 gene (Arhgap7). Mice heterozygous for the targeted allele were phenotypically normal, but homozygous mutant embryos did not survive beyond 10.5 days post coitum. Histological analysis revealed that DLC-1-/- embryos had defects in the neural tube, brain, heart, and placenta. Cultured fibroblasts from DLC-1-deficient embryos displayed alterations in the organization of actin filaments and focal adhesions.


Asunto(s)
Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Citoesqueleto/genética , Citoesqueleto/metabolismo , Citoesqueleto/patología , Fibroblastos , Proteínas Activadoras de GTPasa/deficiencia , Proteínas Activadoras de GTPasa/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Ratones , Ratones Noqueados , Mutación/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
14.
Tissue Eng Part A ; 21(23-24): 2783-90, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25471126

RESUMEN

Stem cell-based medicinal products (SCMPs) are emerging as novel therapeutic products. The success of its development depends on the existence of an effective quality control system, which is constituted by quality control technologies, standards, reference materials, guidelines, and the associated management system in accordance with regulatory requirements along product lifespan. However, a worldwide, effective quality control system specific for SCMPs is still far from established partially due to the limited understanding of stem cell sciences and lack of quality control technologies for accurately assessing the safety and biological effectiveness of SCMPs before clinical use. Even though, based on the existing regulations and current stem cell sciences and technologies, initial actions toward the goal of establishing such a system have been taken as exemplified by recent development of new "interim guidelines" for governing quality control along development of SCMPs and new development of the associated quality control technologies in China. In this review, we first briefly introduced the major institutions involved in the regulation of cell substrates and therapeutic cell products in China and the existing regulatory documents and technical guidelines used as critical references for developing the new interim guidelines. With focus only on nonhematopoietic stem cells, we then discussed the principal quality attributes of SCMPs as well as our thinking of proper testing approaches to be established with relevant evaluation technologies to ensure all quality requirements of SCMPs along different manufacturing processes and development stages. At the end, some regulatory and technical challenges were also discussed with the conclusion that combined efforts should be taken to promote stem cell regulatory sciences to establish the effective quality control system for SCMPs.


Asunto(s)
Productos Biológicos/normas , Células Madre , Animales , Productos Biológicos/uso terapéutico , China , Humanos , Control de Calidad
15.
PLoS One ; 10(2): e0118168, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25692676

RESUMEN

Mesenchymal stem cells (MSCs) are a group of multipotent cells with key properties of multi-lineage differentiation, expressing a set of relatively specific surface markers and unique immunomodulatory functions. IDO1, a catabolic enzyme of tryptophan, represents a critical molecule mediating immunomodulatory functions of MSCs. However, the signaling pathways involved in regulating these key properties still remain elusive. To investigate the involvement of Notch signaling as well as other potential signaling pathway(s) in regulating these critical properties of MSCs, we treated human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) with γ-secreatase inhibitor I (GSI-I), which inhibits both Notch signaling and ubiquitin-proteasome activities. It was shown that the GSI-I treatment resulted in apoptosis, reduced expression of surface markers CD73, CD90 and CD105, reduced osteogenic differentiation, and reduction of the hUC-MSCs-mediated suppression of Th1 lymphocyte proliferation and the IFN-γ-induced IDO1 expression. Through distinguishing the effects of GSI-I between Notch inhibition and proteasome inhibition, it was further observed that, whereas both Notch inhibition and proteasome inhibition were attributable to the reduced CD105 expression and osteogenic differentiation, but not to the induced apoptosis. However, Notch inhibition, but not proteasome inhibition, only contributed to the significant effect of GSI-I on Th1 proliferation probably through reducing IDO1 promoter activity. In conclusion, the Notch signaling may represent a very important cell signaling capable of regulating multiple critical properties, especially the immunomodulatory functions of MSCs.


Asunto(s)
Antígenos CD/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Células Madre Mesenquimatosas/fisiología , Oligopéptidos/farmacología , Receptores de Superficie Celular/genética , Transducción de Señal/efectos de los fármacos , Cordón Umbilical/citología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Endoglina , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunomodulación/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Receptores Notch/metabolismo , Células TH1/citología
16.
Neoplasia ; 6(4): 412-9, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15256063

RESUMEN

The identification of genes undergoing genetic or epigenetic alterations and contributing to the development of cancer is critical to our understanding of the molecular mechanisms of carcinogenesis. A new approach in identifying alterations of genes that might be relevant to the process of tumor development was used in this study by examining the gene expression profile in human lung cancer cells exposed to 5-aza-2'-deoxycytidine (5-aza-dC). A cDNA array analysis was carried out on 5-aza-dC-treated and untreated non small cell lung cancer (NSCLC) cell line NCI-H522. Sixteen and 14 genes were upregulated and downregulated, respectively, by 5-aza-dC treatment. Among them, downregulation of tyrosine protein kinase ABL2 (ABL2) gene and upregulation of hint/protein kinase C inhibitor 1 (Hint/PKCI-1), DVL1, TIMP-1, and TRP-1 genes were found in expanded observations in two or three of five 5-aza-dC-treated NSCLC cell lines. Among these genes, we found that cDNA transfer of Hint/PKCI-1 resulted in a significant in vitro growth inhibition in two cell lines exhibiting 5-aza-dC-induced upregulation of Hint/PKCI-1 and significantly reduced in vivo tumorigenicity of one NSCLC cell line. Hint/PKCI-1, which is the only other characterized human histidine triad (HIT) nucleotide-binding protein in addition to tumor-suppressor gene FHIT, might be involved in lung carcinogenesis.


Asunto(s)
Azacitidina/análogos & derivados , Azacitidina/toxicidad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Antimetabolitos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas , División Celular/efectos de los fármacos , Línea Celular Tumoral , Metilación de ADN , Decitabina , Humanos , Cinética , Neoplasias Pulmonares , Análisis de Secuencia por Matrices de Oligonucleótidos , Transfección
17.
Mol Cancer ; 3: 14, 2004 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-15125777

RESUMEN

BACKGROUND: CHK2 kinase is a tumor suppressor that plays important role in DNA damage signaling, cell cycle regulation and DNA damage induced apoptosis. CHK2 kinase expression was known to be ubiquitous in mammalian cells. CHK2-/- cells were remarkably resistant to DNA damage induced apoptosis, mimicking the clinical behavior of non-small cell lung cancer to conventional chemo and radiation therapy. RESULT: We reported that the CHK2 expression is diminished or absent in both non-small cell lung cancer (NSCLC) cell lines and clinical lung cancer tumor specimens. The absent CHK2 expression in NSCLC was due to hypermethylation of the CHK2 gene promoter, preventing from binding of a transcriptional factor, leading to silence of the CHK2 gene transcription. CONCLUSION: Since the CHK2 null mice showed a remarkable radioresistance, which bear significant similarity to clinical behavior of NSCLC, down-regulation of CHK2 kinase expression by CHK2 gene silencing and methylation in non-small cell lung cancer suggest a critical role of CHK2 kinase in DNA damage induced apoptosis and a novel mechanism of the resistance of NSCLC to DNA damage based therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Metilación de ADN , Regulación Enzimológica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Neoplasias Pulmonares/enzimología , Regiones Promotoras Genéticas/fisiología , Proteínas Serina-Treonina Quinasas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Quinasa de Punto de Control 2 , Cisplatino/administración & dosificación , Cisplatino/metabolismo , Daño del ADN/genética , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Femenino , Silenciador del Gen , Humanos , Inmunohistoquímica/métodos , Concentración 50 Inhibidora , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Proteínas Serina-Treonina Quinasas/inmunología , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Factores de Transcripción/metabolismo
18.
Gene ; 288(1-2): 119-27, 2002 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-12034501

RESUMEN

DLC-1 (deleted in liver cancer 1) is a candidate tumor suppressor gene for hepatocellular carcinoma and other cancers. It is the human homologue of rat p122, which has been shown to function as a GTPase activating protein for RhoA, and it may be involved in signal transduction pathways regulating cell proliferation and adhesion. To establish an animal model for studying the regulation and function of DLC-1, we have undertaken the characterization of the mouse DLC-1 gene. Northern blot analysis shows that the mouse DLC-1 mRNA is widely expressed, with the highest levels in heart, liver, and lung. Mouse genomic clones that contain the entire DLC-1 gene of 47 kb were isolated. The mouse gene consists of 14 exons, and the structural organization is highly similar to that of the human gene. The promoter region of the mouse gene was GC-rich and contained potential binding sites for transcription factors SP1, GCF, and AP-2. A polymorphic microsatellite marker in intron 8 was used for mapping the gene (Arhgap7) to 20 cM on mouse chromosome 8 and for allelotyping of mouse liver tumor DNAs.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Proteínas Supresoras de Tumor/genética , Animales , Secuencia de Bases , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Mapeo Cromosómico , ADN/química , ADN/genética , Exones , Femenino , Eliminación de Gen , Expresión Génica , Genes/genética , Humanos , Intrones , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos , Ratones Transgénicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN
19.
J Mol Diagn ; 5(1): 48-53, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12552080

RESUMEN

DNA amplification in cancer cells frequently involves oncogenes whose increased expression confers a selective advantage on tumor cell growth. In an attempt to identify novel oncogenes involved in hepatocarcinogenesis, representational difference analysis (RDA) was performed using DNA from a primary human hepatocellular carcinoma (HCC) that showed high-level DNA amplifications on chromosomes 1p32 and 11q13 by comparative genomic hybridization. Ten amplification fragments were isolated by RDA, and when used to probe Southern blots of tumor DNA, there was a 5- to 50-fold increase in hybridization intensity relative to normal DNA. The sequence of one amplification product matched that of the EMS1 oncogene, which is located on chromosome 11q13 and is amplified in other cancers. We detected EMS1 amplification in 3 of 17 primary HCC. Overexpression of EMS1 mRNA was observed in 12 of 14 HCC cell lines in the absence of gene amplification or an increased copy-number of the gene. The EMS1 gene encodes cortactin, a cortical actin-associated protein that is a substrate for Src kinase and is involved in cytoskeleton organization. Alterations of the EMS1 gene that lead to overexpression of cortactin may be associated with tumor development in HCC. EMS1 amplification and overexpresion is indicative of unfavorable prognosis in several cancers and may have similar prognostic implications in liver cancer.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Proteínas de Microfilamentos/genética , Oncogenes , Secuencia de Bases , Biomarcadores de Tumor/genética , Cromosomas Humanos Par 11/genética , Cortactina , ADN de Neoplasias/genética , Amplificación de Genes , Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Técnicas de Diagnóstico Molecular , Reacción en Cadena de la Polimerasa , Pronóstico , Células Tumorales Cultivadas
20.
Cancer Genet Cytogenet ; 140(2): 113-7, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12645648

RESUMEN

Aberrant methylation of CpG islands within the promoter regions of tumor suppressor or cancer-related genes is a common mechanism leading to the silencing of gene expression. To determine whether aberrant methylation is a contributing factor to transcriptional inactivation of DLC-1 (deleted in liver cancer-1), a candidate tumor suppressor gene, we examined its methylation status in twelve hepatocellular carcinoma. breast, colon, and prostate tumor cell lines with low or undetectable expression of DLC-1. By Southern blot analysis of DNA digested with the methylation sensitive enzyme HpaII, we found a different degree of promoter hypermethylation in all cell lines with aberrant DLC-1 expression. The hypermethylation status was reversed by the addition of 5-aza-2'-deoxycytidine, a demethylating agent, in one human hepatocellular carcinoma line. These observations suggest that hypermethylation is responsible for abrogating the function of the DLC-1 gene in a subset of liver, breast, colon, and prostate cancers.


Asunto(s)
Azacitidina/análogos & derivados , Islas de CpG , Metilación de ADN , ADN de Neoplasias/química , Genes Supresores de Tumor , Proteínas de Neoplasias/genética , Neoplasias/genética , Regiones Promotoras Genéticas/genética , Proteínas Supresoras de Tumor/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Azacitidina/farmacología , Secuencia de Bases , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Metilación de ADN/efectos de los fármacos , ADN de Neoplasias/genética , Decitabina , Femenino , Proteínas Activadoras de GTPasa , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Datos de Secuencia Molecular , Proteínas de Neoplasias/deficiencia , Regiones Promotoras Genéticas/efectos de los fármacos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Células Tumorales Cultivadas/química , Células Tumorales Cultivadas/patología , Proteínas Supresoras de Tumor/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA