Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Pathol ; 246(2): 134-140, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29952003

RESUMEN

Geminin, a DNA replication licensing inhibitor, ensures faithful DNA replication in vertebrates. Several studies have shown that geminin depletion in vitro results in rereplication and DNA damage, whereas increased expression of geminin has been observed in human cancers. However, conditional inactivation of geminin during embryogenesis has not revealed any detectable DNA replication defects. In order to examine its role in vivo, we conditionally inactivated geminin in the murine colon and lung, and assessed chemically induced carcinogenesis. We show here that mice lacking geminin develop a significantly higher number of tumors and bear a larger tumor burden than sham-treated controls in urethane-induced lung and azoxymethane/dextran sodium sulfate-induced colon carcinogenesis. Survival is also significantly reduced in mice lacking geminin during lung carcinogenesis. A significant increase in the total number and grade of lesions (hyperplasias, adenomas, and carcinomas) was also confirmed by hematoxylin and eosin staining. Moreover, increased genomic aberrations, identified by increased ATR and γH2AX expression, was detected with immunohistochemistry analysis. In addition, we analyzed geminin expression in human colon cancer, and found increased expression, as well as a positive correlation with ATM/ATR levels and a non-monotonic association with γH2AX. Taken together, our data demonstrate that geminin acts as a tumor suppressor by safeguarding genome stability, whereas its overexpression is also associated with genomic instability. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Adenoma/genética , Carcinoma/genética , Neoplasias del Colon/genética , Geminina/genética , Genes Supresores de Tumor , Inestabilidad Genómica , Neoplasias Pulmonares/genética , Adenoma/inducido químicamente , Adenoma/metabolismo , Adenoma/patología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Azoximetano , Carcinoma/inducido químicamente , Carcinoma/metabolismo , Carcinoma/patología , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Geminina/deficiencia , Geminina/metabolismo , Predisposición Genética a la Enfermedad , Histonas/metabolismo , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Fosforilación , Uretano
2.
Development ; 142(1): 70-81, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25516969

RESUMEN

Balancing stem cell self-renewal and initiation of lineage specification programs is essential for the development and homeostasis of the hematopoietic system. We have specifically ablated geminin in the developing murine hematopoietic system and observed profound defects in the generation of mature blood cells, leading to embryonic lethality. Hematopoietic stem cells (HSCs) accumulated in the fetal liver following geminin ablation, while committed progenitors were reduced. Genome-wide transcriptome analysis identified key HSC transcription factors as being upregulated upon geminin deletion, revealing a gene network linked with geminin that controls fetal hematopoiesis. In order to obtain mechanistic insight into the ability of geminin to regulate transcription, we examined Hoxa9 as an example of a key gene in definitive hematopoiesis. We demonstrate that in human K562 cells geminin is associated with HOXA9 regulatory elements and its absence increases HOXA9 transcription similarly to that observed in vivo. Moreover, silencing geminin reduced recruitment of the PRC2 component SUZ12 to the HOXA9 locus and resulted in an increase in RNA polymerase II recruitment and H3K4 trimethylation (H3K4me3), whereas the repressive marks H3K9me3 and H3K27me3 were reduced. The chromatin landscape was also modified at the regulatory regions of HOXA10 and GATA1. K562 cells showed a reduced ability to differentiate to erythrocytes and megakaryocytes upon geminin silencing. Our data suggest that geminin is indispensable for fetal hematopoiesis and regulates the generation of a physiological pool of stem and progenitor cells in the fetal hematopoietic system.


Asunto(s)
Feto/citología , Geminina/deficiencia , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Factores de Transcripción/genética , Animales , Recuento de Células , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Epigénesis Genética , Geminina/metabolismo , Ontología de Genes , Sitios Genéticos , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Histonas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células K562 , Hígado/citología , Hígado/embriología , Ratones , Proteínas de Neoplasias , Complejo Represivo Polycomb 2/metabolismo , Procesamiento Proteico-Postraduccional , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética
3.
Stem Cells ; 35(2): 299-310, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27859962

RESUMEN

Molecular mechanisms governing maintenance, commitment, and differentiation of stem cells are largely unexploited. Molecules involved in the regulation of multiple cellular processes are of particular importance for stem cell physiology, as they integrate different signals and coordinate cellular decisions related with self-renewal and fate determination. Geminin has emerged as a critical factor in DNA replication and stem cell differentiation in different stem cell populations. Its inhibitory interaction with Cdt1, a member of the prereplicative complex, ensures the controlled timing of DNA replication and, consequently, genomic stability in actively proliferating cells. In embryonic as well as somatic stem cells, Geminin has been shown to interact with transcription factors and epigenetic regulators to drive gene expression programs and ultimately guide cell fate decisions. An ever-growing number of studies suggests that these interactions of Geminin and proteins regulating transcription are conserved among metazoans. Interactions between Geminin and proteins modifying the epigenome, such as members of the repressive Polycomb group and the SWI/SNF proteins of the permissive Trithorax, have long been established. The complexity of these interactions, however, is only just beginning to unravel, revealing key roles on maintaining stem cell self-renewal and fate specification. In this review, we summarize current knowledge and give new perspectives for the role of Geminin on transcriptional and epigenetic regulation, alongside with its regulatory activity in DNA replication and their implication in the regulation of stem and progenitor cell biology. Stem Cells 2017;35:299-310.


Asunto(s)
Replicación del ADN/genética , Epigénesis Genética , Geminina/metabolismo , Células Madre/metabolismo , Transcripción Genética , Animales , Inestabilidad Genómica , Humanos
4.
Dev Biol ; 409(2): 392-405, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26658318

RESUMEN

Neural crest cells comprise a multipotent, migratory cell population that generates a diverse array of cell and tissue types, during vertebrate development. Enteric Nervous System controls the function of the gastrointestinal tract and is mainly derived from the vagal and sacral neural crest cells. Deregulation on self-renewal and differentiation of the enteric neural crest cells is evident in enteric nervous system disorders, such as Hirschsprung disease, characterized by the absence of ganglia in a variable length of the distal bowel. Here we show that Geminin is essential for Enteric Nervous System generation as mice that lacked Geminin expression specifically in neural crest cells revealed decreased generation of vagal neural crest cells, and enteric neural crest cells (ENCCs). Geminin-deficient ENCCs showed increased apoptosis and decreased cell proliferation during the early stages of gut colonization. Furthermore, decreased number of committed ENCCs in vivo and the decreased self-renewal capacity of enteric progenitor cells in vitro, resulted in almost total aganglionosis resembling a severe case of Hirschsprung disease. Our results suggest that Geminin is an important regulator of self-renewal and survival of enteric nervous system progenitor cells.


Asunto(s)
Sistema Nervioso Entérico/patología , Geminina/metabolismo , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/patología , Cresta Neural/metabolismo , Células Madre/metabolismo , Animales , Recuento de Células , Muerte Celular , Diferenciación Celular , Proliferación Celular , Autorrenovación de las Células , Geminina/deficiencia , Eliminación de Gen , Intestinos/patología , Ratones , Cresta Neural/citología , Neuroglía/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo
5.
Cytotherapy ; 19(4): 521-530, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28162915

RESUMEN

BACKGROUND: Major barriers in using classical FOXP3+ regulatory T cells (Tregs) in clinical practice are their low numbers in the circulation, the lack of specific cell surface markers for efficient purification and the loss of expression of Treg signature molecules and suppressive function after in vitro expansion or in a pro-inflammatory microenviroment. A surface molecule with potent immunosuppressive function is the human leukocyte antigen-G (HLA-G), which is normally expressed in placenta protecting the "semi-allogeneic" fetus from maternal immune attack. Because HLA-G expression is strongly regulated by methylation, we asked whether hypomethylating agents (HA) may be used in vitro to induce HLA-G expression on conventional T cells and convert them to Tregs. METHODS: Human peripheral blood T cells were exposed to azacytidine/decitabine and analyzed for HLA-G expression and their in vitro suppressor properties. RESULTS: HA treatment induces de novo expression of HLA-G on T cells through hypomethylation of the HLA-G proximal promoter. The HA-induced CD4+HLA-Gpos T cells are FOXP3 negative and have potent in vitro suppression function, which is dependent to a large extent, but not exclusively, on the HLA-G molecule. Converted HLA-Gpos suppressors retain their suppressor function in the presence of tumor necrosis factor (TNF) and preserve hypomethylated the HLA-G promoter for at least 2 days after azacytidine exposure. Decitabine-treated T cells suppressed ex vivo the proliferation of T cells isolated from patients suffering from graft-versus-host disease (GVHD). DISCUSSION: We propose, in vitro generation of HLA-G-expressing T cells through pharmacological hypomethylation as a simple, Good Manufacturing Practice (GMP)-compatible and efficient strategy to produce a stable Treg subset of a defined phenotype that can be easily purified for adoptive immunotherapy.


Asunto(s)
Ingeniería Celular/métodos , Enfermedad Injerto contra Huésped/terapia , Antígenos HLA-G/metabolismo , Inmunoterapia Adoptiva/métodos , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Azacitidina/análogos & derivados , Azacitidina/farmacología , Técnicas de Cultivo de Célula , Células Cultivadas , Metilación de ADN/efectos de los fármacos , Decitabina , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedad Injerto contra Huésped/inmunología , Antígenos HLA-G/genética , Humanos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
6.
Respir Res ; 10: 82, 2009 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-19744320

RESUMEN

BACKGROUND: Cigarette smoke (CS) is the main cause in the development of chronic obstructive pulmonary disease (COPD), the pathogenesis of which is related to an extended inflammatory response. In this study, we investigated the effect of low and high doses of gas phase cigarette smoke (GPS) on cultured lymphocyte progenitor cells, using techniques to assess cell viability and to elucidate whether cells die of apoptosis or necrosis upon exposure to different doses of GPS. METHODS: In our approach we utilised a newly-established system of exposure of cells to GPS that is highly controlled, accurately reproducible and simulates CS dosage and kinetics that take place in the smokers' lung. This system was used to study the mode of cell death upon exposure to GPS in conjunction with a range of techniques widely used for cell death studies such as Annexin V staining, activation of caspase -3, cytoplasmic release of cytochrome C, loss of mitochondrial membrane potential and DNA fragmentation. RESULTS: Low doses of GPS induced specific apoptotic indexes in CCRF-CEM cells. Specifically, cytochrome C release and cleaved caspase-3 were detected by immunofluorescence, upon treatment with 1-3 puffs GPS. At 4 h post-exposure, caspase-3 activation was observed in western blot analysis, showing a decreasing pattern as GPS doses increased. Concomitant with this behaviour, a dose-dependent change in Delta psi m depolarization was monitored by flow cytometry 2 h post-exposure, while at 4 h Delta psi m collapse was observed at the higher doses, indicative of a shift to a necrotic demise. A reduction in DNA fragmentation events produced by 5 puffs GPS as compared to those provoked by 3 puffs GPS, also pointed towards a necrotic response at the higher dose of GPS. CONCLUSION: Collectively, our results support that at low doses gas phase cigarette smoke induces apoptosis in cultured T-lymphocytes, whereas at high doses GPS leads to necrotic death, by-passing the characteristic stage of caspase-3 activation and, thus, the apoptotic route.


Asunto(s)
Apoptosis/efectos de los fármacos , Nicotiana/efectos adversos , Humo/efectos adversos , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Línea Celular , Relación Dosis-Respuesta a Droga , Gases/efectos adversos , Humanos , Transición de Fase
7.
Data Brief ; 7: 889-93, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27077091

RESUMEN

We performed cDNA microarrays (Affymetrix Mouse Gene 1.0 ST Chip) to analyze the transcriptome of hematopoietic stem and progenitor cells (HSPCs) from E15.5dpc wild type and Geminin (Gmnn) knockout embryos. Lineage negative cells from embryonic livers were isolated using fluorescence activated cell sorting. RNA samples were used to examine the transcriptional programs regulated by Geminin during embryonic hematopoiesis. The data sets were analyzed using the GeneSpring v12.5 platform (Agilent). The list of differentially expressed genes was filtered in meta-analyses to investigate the molecular basis of the phenotype observed in the knockout embryos, which exhibited defective hematopoiesis and death. The data from this study are related to the research article "Geminin deletion increases the number of fetal hematopoietic stem cells by affecting the expression of key transcription factors" (Karamitros et al., 2015) [1]. The microarray dataset has been deposited at the Gene Expression Omnibus (GEO) under accession GEO: GSE53056.

8.
J Clin Invest ; 125(6): 2317-34, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25915587

RESUMEN

Mast cells (MCs) have been identified in various tumors; however, the role of these cells in tumorigenesis remains controversial. Here, we quantified MCs in human and murine malignant pleural effusions (MPEs) and evaluated the fate and function of these cells in MPE development. Evaluation of murine MPE-competent lung and colon adenocarcinomas revealed that these tumors actively attract and subsequently degranulate MCs in the pleural space by elaborating CCL2 and osteopontin. MCs were required for effusion development, as MPEs did not form in mice lacking MCs, and pleural infusion of MCs with MPE-incompetent cells promoted MPE formation. Once homed to the pleural space, MCs released tryptase AB1 and IL-1ß, which in turn induced pleural vasculature leakiness and triggered NF-κB activation in pleural tumor cells, thereby fostering pleural fluid accumulation and tumor growth. Evaluation of human effusions revealed that MCs are elevated in MPEs compared with benign effusions. Moreover, MC abundance correlated with MPE formation in a human cancer cell-induced effusion model. Treatment of mice with the c-KIT inhibitor imatinib mesylate limited effusion precipitation by mouse and human adenocarcinoma cells. Together, the results of this study indicate that MCs are required for MPE formation and suggest that MC-dependent effusion formation is therapeutically addressable.


Asunto(s)
Mastocitos/metabolismo , Derrame Pleural Maligno/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Humanos , Mesilato de Imatinib , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Neoplasias Pulmonares/dietoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Mastocitos/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Piperazinas/farmacología , Cavidad Pleural/metabolismo , Cavidad Pleural/patología , Derrame Pleural Maligno/tratamiento farmacológico , Derrame Pleural Maligno/genética , Derrame Pleural Maligno/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/farmacología , Triptasas/genética , Triptasas/metabolismo
9.
Virology ; 317(2): 308-20, 2003 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-14698669

RESUMEN

During infection of insect cells with Autographa californica nucleopolyhedrovirus (AcMNPV), the very late protein P10 forms large fibrillar structures in the cytoplasm and nuclei of infected cells. In this study we have used confocal microscopy in association with a novel P10 antiserum to localise and study P10 in virus-infected cells. P10 was shown to be a component of tubular-like structures that spiralled throughout the cytoplasm and nucleus of AcMNPV-infected cells. These structures were observed to colocalise partly with cortical microtubules. When microtubules were depolymerised with the drug nocodazole, P10 tubules failed to form and the protein appeared concentrated in cytoplasmic foci. For the first time, we provide direct evidence using both antibody pulldown and yeast two-hybrid experiments for the interaction of P10 with host-cell tubulin. It is suggested that this interaction may be a critical factor in AcMNPV-induced cell lysis.


Asunto(s)
Microtúbulos/metabolismo , Nucleopoliedrovirus/patogenicidad , Proteínas Virales/metabolismo , Actinas/metabolismo , Animales , Células Cultivadas , Efecto Citopatogénico Viral , Citoesqueleto/metabolismo , Microscopía Confocal , Proteínas Asociadas a Microtúbulos/metabolismo , Nocodazol/farmacología , Nucleopoliedrovirus/metabolismo , Pruebas de Precipitina , Spodoptera/virología , Técnicas del Sistema de Dos Híbridos
10.
J Gen Virol ; 85(Pt 4): 821-831, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15039525

RESUMEN

Infection of insect larvae with Autographa californica nucleopolyhedrovirus (AcMNPV) results in the liquefaction of the host, a process involving the action of virus-encoded chitinase and cathepsin gene products. Chitinase is localized in the endoplasmic reticulum (ER) during infection because of the presence of a C-terminal ER retrieval motif (KDEL). In this study, the KDEL coding region was removed from the chitinase gene so that expression of the modified chitinase remained under the control of its own gene promoter, at its native locus. The deletion of KDEL resulted in the redistribution of chitinase within the cell during virus infection. Chitinase lacking the KDEL motif was detectable at the plasma membrane and was also evident in the culture medium of virus-infected cells from as early as 12 h post-infection (p.i.). Secretion of chitinase from the cell continued up to 72 h p.i., until cytolysis. The biological activity of the recombinant virus in Trichoplusia ni larvae was enhanced, with a significant reduction in the lethal dose and lethal time associated with infection. Furthermore, a reduction in feeding damage caused by infected larvae was observed compared to AcMNPV-infected individuals.


Asunto(s)
Quitinasas/genética , Nucleopoliedrovirus/enzimología , Nucleopoliedrovirus/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Quitinasas/química , Quitinasas/metabolismo , ADN Viral/genética , Genoma Viral , Datos de Secuencia Molecular , Mariposas Nocturnas , Nucleopoliedrovirus/patogenicidad , Oligopéptidos , Señales de Clasificación de Proteína , Eliminación de Secuencia , Spodoptera , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA