Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Biochem ; 120(10): 18332-18345, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31257681

RESUMEN

MYC and RUNX oncogenes each trigger p53-mediated failsafe responses when overexpressed in vitro and collaborate with p53 deficiency in vivo. However, together they drive rapid onset lymphoma without mutational loss of p53. This phenomenon was investigated further by transcriptomic analysis of premalignant thymus from RUNX2/MYC transgenic mice. The distinctive contributions of MYC and RUNX to transcriptional control were illustrated by differential enrichment of canonical binding sites and gene ontology analyses. Pathway analysis revealed signatures of MYC, CD3, and CD28 regulation indicative of activation and proliferation, but also strong inhibition of cell death pathways. In silico analysis of discordantly expressed genes revealed Tnfsrf8/CD30, Cish, and Il13 among relevant targets for sustained proliferation and survival. Although TP53 mRNA and protein levels were upregulated, its downstream targets in growth suppression and apoptosis were largely unperturbed. Analysis of genes encoding p53 posttranslational modifiers showed significant upregulation of three genes, Smyd2, Set, and Prmt5. Overexpression of SMYD2 was validated in vivo but the functional analysis was constrained by in vitro loss of p53 in RUNX2/MYC lymphoma cell lines. However, an early role is suggested by the ability of SMYD2 to block senescence-like growth arrest induced by RUNX overexpression in primary fibroblasts.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Linfoma/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Senescencia Celular/genética , Senescencia Celular/fisiología , Biología Computacional , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Linfoma/genética , Ratones , Ratones Transgénicos , Análisis de Componente Principal , Proteínas Proto-Oncogénicas c-myc/genética , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Timo/metabolismo , Proteína p53 Supresora de Tumor
2.
J Cell Biochem ; 119(3): 2750-2762, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29052866

RESUMEN

RUNX gene over-expression inhibits growth of primary cells but transforms cells with tumor suppressor defects, consistent with reported associations with tumor progression. In contrast, chromosomal translocations involving RUNX1 are detectable in utero, suggesting an initiating role in leukemias. How do cells expressing RUNX1 fusion oncoproteins evade RUNX-mediated growth suppression? Previous studies showed that the TEL-RUNX1 fusion from t(12;21) B-ALLs is unable to induce senescence-like growth arrest (SLGA) in primary fibroblasts while potent activity is displayed by the RUNX1-ETO fusion found in t(8;21) AMLs. We now show that SLGA potential is suppressed in TEL-RUNX1 but reactivated by deletion of the TEL HLH domain or mutation of a key residue (K99R). Attenuation of SLGA activity is also a feature of RUNX1-ETO9a, a minor product of t(8;21) translocations with increased leukemogenicity. Finally, while RUNX1-ETO induces SLGA it also drives a potent senescence-associated secretory phenotype (SASP), and promotes the immortalization of rare cells that escape SLGA. Moreover, the RUNX1-ETO SASP is not strictly linked to growth arrest as it is largely suppressed by RUNX1 and partially activated by RUNX1-ETO9a. These findings underline the heterogeneous nature of premature senescence and the multiple mechanisms by which this failsafe process is subverted in cells expressing RUNX1 oncoproteins.


Asunto(s)
Puntos de Control del Ciclo Celular , Senescencia Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Proteínas de Unión al ADN/genética , Humanos , Ratones , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas/genética , Proteína 1 Compañera de Translocación de RUNX1/genética , Factores de Transcripción/genética
3.
Oncotarget ; 8(38): 62818-62819, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28968950
4.
Adv Exp Med Biol ; 962: 247-264, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28299662

RESUMEN

The observation that the Runx genes act as targets for transcriptional activation by retroviral insertion identified a new family of dominant oncogenes. However, it is now clear that Runx genes are 'conditional' oncogenes whose over-expression is growth inhibitory unless accompanied by another event such as concomitant over-expression of MYC or loss of p53 function. Remarkably, while the oncogenic activities of either MYC or RUNX over-expression are suppressed while p53 is intact, the combination of both neutralises p53 tumour suppression in vivo by as yet unknown mechanisms. Moreover, there is emerging evidence that endogenous, basal RUNX activity is important to maintain the viability and proliferation of MYC-driven lymphoma cells. There is also growing evidence that the human RUNX genes play a similar conditional oncogenic role and are selected for over-expression in end-stage cancers of multiple types. Paradoxically, reduced RUNX activity can also predispose to cell immortalisation and transformation, particularly by mutant Ras. These apparently conflicting observations may be reconciled in a stage-specific model of RUNX involvement in cancer. A question that has yet to be fully addressed is the extent to which the three Runx genes are functionally redundant in cancer promotion and suppression.


Asunto(s)
Subunidades alfa del Factor de Unión al Sitio Principal/genética , Neoplasias/genética , Oncogenes/genética , Retroviridae/crecimiento & desarrollo , Animales , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Proteína p53 Supresora de Tumor/genética
5.
J Virol ; 91(5)2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031367

RESUMEN

The human genome displays a rich fossil record of past gammaretrovirus infections, yet no current epidemic is evident, despite environmental exposure to viruses that infect human cells in vitro Feline leukemia viruses (FeLVs) rank high on this list, but neither domestic nor workplace exposure has been associated with detectable serological responses. Nonspecific inactivation of gammaretroviruses by serum factors appears insufficient to explain these observations. To investigate further, we explored the susceptibilities of primary and established human cell lines to FeLV-B, the most likely zoonotic variant. Fully permissive infection was common in cancer-derived cell lines but was also a feature of nontransformed keratinocytes and lung fibroblasts. Cells of hematopoietic origin were generally less permissive and formed discrete groups on the basis of high or low intracellular protein expression and virion release. Potent repression was observed in primary human blood mononuclear cells and a subset of leukemia cell lines. However, the early steps of reverse transcription and integration appear to be unimpaired in nonpermissive cells. FeLV-B was subject to G→A hypermutation with a predominant APOBEC3G signature in partially permissive cells but was not mutated in permissive cells or in nonpermissive cells that block secondary viral spread. Distinct cellular barriers that protect primary human blood cells are likely to be important in protection against zoonotic infection with FeLV.IMPORTANCE Domestic exposure to gammaretroviruses such as feline leukemia viruses (FeLVs) occurs worldwide, but the basis of human resistance to infection remains incompletely understood. The potential threat is evident from the human genome sequence, which reveals many past epidemics of gammaretrovirus infection, and from recent cross-species jumps of gammaretroviruses from rodents to primates and marsupials. This study examined resistance to infection at the cellular level with the most prevalent human cell-tropic FeLV variant, FeLV-B. We found that blood cells are uniquely resistant to infection with FeLV-B due to the activity of cellular enzymes that mutate the viral genome. A second block, which appears to suppress viral gene expression after the viral genome has integrated into the host cell genome, was identified. Since cells derived from other normal human cell types are fully supportive of FeLV replication, innate resistance of blood cells could be critical in protecting against cross-species infection.


Asunto(s)
Virus de la Leucemia Felina/fisiología , Infecciones por Retroviridae/virología , Desaminasa APOBEC-3G/genética , Desaminasa APOBEC-3G/metabolismo , Animales , Gatos , Línea Celular Tumoral , Susceptibilidad a Enfermedades , Expresión Génica , Genoma Viral , Células HEK293 , Humanos , Mutación , Especificidad de la Especie , Tropismo Viral , Integración Viral , Replicación Viral , Zoonosis/virología
7.
PLoS One ; 11(4): e0154070, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27097319

RESUMEN

Retroviruses have been foundational in cancer research since early studies identified proto-oncogenes as targets for insertional mutagenesis. Integration of murine gamma-retroviruses into the host genome favours promoters and enhancers and entails interaction of viral integrase with host BET/bromodomain factors. We report that this integration pattern is conserved in feline leukaemia virus (FeLV), a gamma-retrovirus that infects many human cell types. Analysis of FeLV insertion sites in the MCF-7 mammary carcinoma cell line revealed strong bias towards active chromatin marks with no evidence of significant post-integration growth selection. The most prominent FeLV integration targets had little overlap with the most abundantly expressed transcripts, but were strongly enriched for annotated cancer genes. A meta-analysis based on several gamma-retrovirus integration profiling (GRIP) studies in human cells (CD34+, K562, HepG2) revealed a similar cancer gene bias but also remarkable cell-type specificity, with prominent exceptions including a universal integration hotspot at the long non-coding RNA MALAT1. Comparison of GRIP targets with databases of super-enhancers from the same cell lines showed that these have only limited overlap and that GRIP provides unique insights into the upstream drivers of cell growth. These observations elucidate the oncogenic potency of the gamma-retroviruses and support the wider application of GRIP to identify the genes and growth regulatory circuits that drive distinct cancer types.


Asunto(s)
Genes Relacionados con las Neoplasias , Virus de la Leucemia Felina/genética , Neoplasias/genética , Neoplasias/virología , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología , Integración Viral , Animales , Gatos , Línea Celular Tumoral , Cromatina/genética , Cromatina/virología , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Infecciones por Retroviridae/complicaciones , Infecciones por Retroviridae/genética , Sitio de Iniciación de la Transcripción , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/genética
8.
Oncotarget ; 7(17): 22973-87, 2016 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-27056890

RESUMEN

The Runx genes function as dominant oncogenes that collaborate potently with Myc or loss of p53 to induce lymphoma when over-expressed. Here we examined the requirement for basal Runx1 activity for tumor maintenance in the Eµ-Myc model of Burkitt's lymphoma. While normal Runx1fl/fl lymphoid cells permit mono-allelic deletion, primary Eµ-Myc lymphomas showed selection for retention of both alleles and attempts to enforce deletion in vivo led to compensatory expansion of p53null blasts retaining Runx1. Surprisingly, Runx1 could be excised completely from established Eµ-Myc lymphoma cell lines in vitro without obvious effects on cell phenotype. Established lines lacked functional p53, and were sensitive to death induced by introduction of a temperature-sensitive p53 (Val135) allele. Transcriptome analysis of Runx1-deleted cells revealed a gene signature associated with lymphoid proliferation, survival and differentiation, and included strong de-repression of recombination-activating (Rag) genes, an observation that was mirrored in a panel of human acute leukemias where RUNX1 and RAG1,2 mRNA expression were negatively correlated. Notably, despite their continued growth and tumorigenic potential, Runx1null lymphoma cells displayed impaired proliferation and markedly increased sensitivity to DNA damage and dexamethasone-induced apoptosis, validating Runx1 function as a potential therapeutic target in Myc-driven lymphomas regardless of their p53 status.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Linfoma/patología , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Apoptosis , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Femenino , Humanos , Linfoma/genética , Linfoma/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células Tumorales Cultivadas
9.
Sci Rep ; 5: 15658, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26489514

RESUMEN

Although best known for its role in bone development and associated structures the transcription factor RUNX2 is expressed in a wide range of lineages, including those of the mammary gland. Previous studies have indicated that Runx2 can regulate aspects of mammary cell function and influence the properties of cancer cells. In this study we investigate the role of Runx2 in the mammary stem/progenitor population and its relationship with WNT signalling. Results show that RUNX2 protein is differentially expressed throughout embryonic and adult development of the murine mammary gland with high levels of expression in mammary stem-cell enriched cultures. Importantly, functional analysis reveals a role for Runx2 in mammary stem/progenitor cell function in in vitro and in vivo regenerative assays. Furthermore, RUNX2 appears to be associated with WNT signalling in the mammary epithelium and is specifically upregulated in mouse models of WNT-driven breast cancer. Overall our studies reveal a novel function for Runx2 in regulating mammary epithelial cell regenerative potential, possibly acting as a downstream target of WNT signalling.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Epitelio/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular/genética , Epitelio/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Vía de Señalización Wnt/genética
11.
Viruses ; 7(4): 2014-29, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25912714

RESUMEN

Infection of human cancer xenografts in mice with murine leukemia viruses (MLVs) is a long-standing observation, but the likelihood of infection in vivo and its biological consequences are poorly understood. We therefore conducted a prospective study in commonly used xenograft recipient strains. From BALB/c nude mice engrafted with MCF7 human mammary carcinoma cells, we isolated a virus that was virtually identical to Bxv1, a locus encoding replication-competent xenotropic MLV (XMLV). XMLV was detected in 9/17 (53%) independently isolated explants. XMLV was not found in primary leukemias or in THP1 leukemia cells grown in Bxv1-negative NSG (NOD/SCID/γCnull) mice, although MCF7 explants harbored replication-defective MLV proviruses. To assess the significance of infection for xenograft behavior in vivo, we examined changes in growth and global transcription in MCF7 and the highly susceptible Raji Burkitt lymphoma cell line chronically infected with XMLV. Raji cells showed a stronger transcriptional response that included up-regulation of chemokines and effectors of innate antiviral immunity. In conclusion, the risk of de novo XMLV infection of xenografts is high in Bxv1 positive mice, while infection can have positive or negative effects on xenograft growth potential with significant consequences for interpretation of many xenograft studies.


Asunto(s)
Retrovirus Endógenos/aislamiento & purificación , Xenoinjertos/virología , Neoplasias/virología , Animales , Humanos , Ratones , Ratones Endogámicos BALB C , Estudios Prospectivos
12.
Dis Model Mech ; 7(5): 525-34, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24626992

RESUMEN

RUNX2, a master regulator of osteogenesis, is oncogenic in the lymphoid lineage; however, little is known about its role in epithelial cancers. Upregulation of RUNX2 in cell lines correlates with increased invasiveness and the capacity to form osteolytic disease in models of breast and prostate cancer. However, most studies have analysed the effects of this gene in a limited number of cell lines and its role in primary breast cancer has not been resolved. Using a human tumour tissue microarray, we show that high RUNX2 expression is significantly associated with oestrogen receptor (ER)/progesterone receptor (PR)/HER2-negative breast cancers and that patients with high RUNX2 expression have a poorer survival rate than those with negative or low expression. We confirm RUNX2 as a gene that has a potentially important functional role in triple-negative breast cancer. To investigate the role of this gene in breast cancer, we made a transgenic model in which Runx2 is specifically expressed in murine mammary epithelium under the control of the mouse mammary tumour virus (MMTV) promoter. We show that ectopic Runx2 perturbs normal development in pubertal and lactating animals, delaying ductal elongation and inhibiting lobular alveolar differentiation. We also show that the Runx2 transgene elicits age-related, pre-neoplastic changes in the mammary epithelium of older transgenic animals, suggesting that elevated RUNX2 expression renders such tissue more susceptible to oncogenic changes and providing further evidence that this gene might have an important, context-dependent role in breast cancer.


Asunto(s)
Neoplasias de la Mama/clasificación , Neoplasias de la Mama/metabolismo , Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Glándulas Mamarias Animales/patología , Análisis de Matrices Tisulares , Animales , Neoplasias de la Mama/patología , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Hiperplasia , Lactancia , Glándulas Mamarias Animales/metabolismo , Virus del Tumor Mamario del Ratón/fisiología , Ratones Transgénicos , Persona de Mediana Edad , Paridad , Lesiones Precancerosas/patología , Embarazo , Factor de Transcripción STAT5/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo
13.
PLoS Genet ; 10(2): e1004167, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24586197

RESUMEN

Retroviral insertional mutagenesis (RIM) is a powerful tool for cancer genomics that was combined in this study with deep sequencing (RIM/DS) to facilitate a comprehensive analysis of lymphoma progression. Transgenic mice expressing two potent collaborating oncogenes in the germ line (CD2-MYC, -Runx2) develop rapid onset tumours that can be accelerated and rendered polyclonal by neonatal Moloney murine leukaemia virus (MoMLV) infection. RIM/DS analysis of 28 polyclonal lymphomas identified 771 common insertion sites (CISs) defining a 'progression network' that encompassed a remarkably large fraction of known MoMLV target genes, with further strong indications of oncogenic selection above the background of MoMLV integration preference. Progression driven by RIM was characterised as a Darwinian process of clonal competition engaging proliferation control networks downstream of cytokine and T-cell receptor signalling. Enhancer mode activation accounted for the most efficiently selected CIS target genes, including Ccr7 as the most prominent of a set of chemokine receptors driving paracrine growth stimulation and lymphoma dissemination. Another large target gene subset including candidate tumour suppressors was disrupted by intragenic insertions. A second RIM/DS screen comparing lymphomas of wild-type and parental transgenics showed that CD2-MYC tumours are virtually dependent on activation of Runx family genes in strong preference to other potent Myc collaborating genes (Gfi1, Notch1). Ikzf1 was identified as a novel collaborating gene for Runx2 and illustrated the interface between integration preference and oncogenic selection. Lymphoma target genes for MoMLV can be classified into (a) a small set of master regulators that confer self-renewal; overcoming p53 and other failsafe pathways and (b) a large group of progression genes that control autonomous proliferation in transformed cells. These findings provide insights into retroviral biology, human cancer genetics and the safety of vector-mediated gene therapy.


Asunto(s)
Genes myb/genética , Linfoma/genética , Virus de la Leucemia Murina de Moloney/genética , Mutagénesis Insercional/genética , Proteína p53 Supresora de Tumor/genética , Animales , Carcinogénesis , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Células Germinativas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factor de Transcripción Ikaros/genética , Linfoma/patología , Linfoma/virología , Ratones
14.
J Endocrinol ; 209(2): 245-54, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307122

RESUMEN

In late puberty, estrogen decelerates bone growth by stimulating growth plate maturation. In this study, we analyzed the mechanism of estrogen action using two pubertal growth plate specimens of one girl at Tanner stage B2 and Tanner stage B3. Histological analysis showed that progression of puberty coincided with characteristic morphological changes: a decrease in total growth plate height (P=0.002), height of the individual zones (P<0.001), and an increase in intercolumnar space (P<0.001). Microarray analysis of the specimens identified 394 genes (72% upregulated and 28% downregulated) that changed with the progression of puberty. Overall changes in gene expression were small (average 1.38-fold upregulated and 1.36-fold downregulated genes). The 394 genes mapped to 13 significantly changing pathways (P<0.05) associated with growth plate maturation (e.g. extracellular matrix, cell cycle, and cell death). We next scanned the upstream promoter regions of the 394 genes for the presence of evolutionarily conserved binding sites for transcription factors implicated in growth plate maturation such as estrogen receptor (ER), androgen receptor, ELK1, STAT5B, cyclic AMP response element (CREB), and RUNX2. High-quality motif sites for RUNX2 (87 genes), ELK1 (43 genes), and STAT5B (31 genes), but not ER, were evolutionarily conserved, indicating their functional relevance across primates. Moreover, we show that some of these sites are direct target genes of these transcription factors as shown by ChIP assays.


Asunto(s)
Desarrollo del Adolescente , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Placa de Crecimiento/metabolismo , Hormonas/metabolismo , Adolescente , Niño , Femenino , Perfilación de la Expresión Génica , Genoma Humano , Placa de Crecimiento/anatomía & histología , Placa de Crecimiento/crecimiento & desarrollo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
15.
Cancer Res ; 70(14): 5860-9, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20587518

RESUMEN

The Runx genes (Runx1, 2, and 3) regulate cell fate in development and can operate as either oncogenes or tumor suppressors in cancer. The oncogenic potential of ectopic Runx expression has been shown in transgenic mice that develop lymphoma in potent synergy with overexpressed Myc, and in established fibroblasts that display altered morphology and increased tumorigenicity. Candidate oncogenic functions of overexpressed Runx genes include resistance to apoptosis in response to intrinsic and extrinsic stresses. In a search for gene targets responsible for this aspect of Runx phenotype, we have identified three key enzymes in sphingolipid metabolism (Sgpp1, Ugcg, and St3gal5/Siat9) as direct targets for Runx transcriptional regulation in a manner consistent with survival and apoptosis resistance. Consistent with these changes in gene expression, mass spectrometric analysis showed that ectopic Runx reduces intracellular long-chain ceramides in NIH3T3 fibroblasts and elevated extracellular sphingosine 1 phosphate. Runx expression also opposed the activation of c-Jun-NH(2)-kinase and p38(MAPK), key mediators of ceramide-induced death, and suppressed the onset of apoptosis in response to exogenous tumor necrosis factor alpha. The survival advantage conferred by ectopic Runx could be partially recapitulated by exogenous sphingosine 1 phosphate and was accompanied by reduced phosphorylation of p38(MAPK). These results reveal a novel link between transcription factor oncogenes and lipid signaling pathways involved in cancer cell survival and chemoresistance.


Asunto(s)
Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Esfingolípidos/metabolismo , Animales , Sitios de Unión , Subunidades alfa del Factor de Unión al Sitio Principal/biosíntesis , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Mol Ther ; 17(6): 1031-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19337236

RESUMEN

The emergence of leukemia following gene transfer to restore common cytokine receptor gamma chain (gammaC) function in X-linked severe combined immunodeficiency (SCID-X1) has raised important questions with respect to gene therapy safety. To explore the risk factors involved, we tested the oncogenic potential of human gammaC in new strains of transgenic mice expressing the gene under the control of the CD2 promoter and locus control region (LCR). These mice demonstrated mildly perturbed T-cell development, with an increased proportion of thymic CD8 cells, but showed no predisposition to tumor development even on highly tumor prone backgrounds or after gamma-retrovirus infection. The human CD2-gammaC transgene rescued T and B-cell development in gammaC(-/-) mice but with an age-related delay, mimicking postnatal reconstitution in SCID-X1 gene therapy subjects. However, we noted that gammaC(-/-) mice are acutely susceptible to murine leukemia virus (MLV) leukemogenesis, and that this trait was not corrected by the gammaC transgene. We conclude that the SCID-X1 phenotype can be corrected safely by stable ectopic expression of gammaC and that the transgene is not significantly oncogenic when expressed in this context. However, an underlying predisposition conferred by the SCID-X1 background appears to collaborate with insertional mutagenesis to increase the risk of tumor development.


Asunto(s)
Terapia Genética/efectos adversos , Subunidad gamma Común de Receptores de Interleucina/fisiología , Linfoma/etiología , Linfoma/genética , Retroviridae/fisiología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Linfocitos B/metabolismo , Western Blotting , Antígenos CD2/genética , Citometría de Flujo , Genotipo , Humanos , Inmunofenotipificación , Técnicas In Vitro , Subunidad gamma Común de Receptores de Interleucina/genética , Linfoma/inmunología , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas/genética , Retroviridae/genética , Linfocitos T/metabolismo , Timo/metabolismo
17.
Blood Cells Mol Dis ; 43(1): 12-9, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19269865

RESUMEN

Runx1 is essential for the homeostatic control of normal hematopoiesis and is required for lymphoid development. Translocations or point mutations that result in RUNX1 loss or disrupted function predispose to leukemia but data derived from model systems suggests that Runx genes can also be pro-oncogenic. Here we investigate the effects of enforced Runx1 expression in lymphoid lineages both in vivo and in vitro and show that transgene expression enhanced cell survival in the thymus and bone marrow but strongly inhibited the expansion of hematopoietic and B cell progenitors in vitro. Despite this, modestly enhanced levels of Runx1 accelerated Myc-induced lymphomagenesis in both the B cell and T cell lineages. Together these data provide formal proof that wild type Runx1 can promote oncogenesis in lymphoid tissues and that, in addition to loss of function, gain of function may have an aetiological role in leukemia.


Asunto(s)
Linfocitos B/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Linfoma/genética , Linfocitos T/patología , Animales , Linfocitos B/citología , Proliferación Celular , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Genes myc , Tejido Linfoide/citología , Tejido Linfoide/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T/citología
19.
Cancer Res ; 67(23): 11263-71, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18056452

RESUMEN

The Runx genes play paradoxical roles in cancer where they can function either as dominant oncogenes or tumor suppressors according to context. We now show that the ability to induce premature senescence in primary murine embryonic fibroblasts (MEF) is a common feature of all three Runx genes. However, ectopic Runx-induced senescence contrasts with Ras oncogene-induced senescence, as it occurs directly and lacks the hallmarks of proliferative stress. Moreover, a fundamental role for Runx function in the senescence program is indicated by the effects of Runx2 disruption, which renders MEFs prone to spontaneous immortalization and confers an early growth advantage that is resistant to stress-induced growth arrest. Runx2(-/-) cells are refractory to H-Ras(V12)-induced premature senescence, despite the activation of a cascade of growth inhibitors and senescence markers, and are permissive for oncogenic transformation. The aberrant behavior of Runx2(-/-) cells is associated with signaling defects and elevated expression of S-G(2)-M cyclins and their associated cyclin dependent kinase activities that may override the effects of growth inhibitory signals. Coupling of stress responses to the cell cycle represents a novel facet of Runx tumor suppressor function and provides a rationale for the lineage-specific effects of loss of Runx function in cancer.


Asunto(s)
Transformación Celular Neoplásica , Senescencia Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Fibroblastos/fisiología , Genes ras/fisiología , Células 3T3 , Animales , Western Blotting , Ciclo Celular , Ensayo de Unidades Formadoras de Colonias , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Quinasas Ciclina-Dependientes/metabolismo , Inmunoprecipitación , Ratones , Ratones Desnudos
20.
Cancer Res ; 67(11): 5126-33, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17545590

RESUMEN

In this study, we have exploited the power of insertional mutagenesis to elucidate tumor progression pathways in mice carrying two oncogenes (MYC/Runx2) that collaborate to drive early lymphoma development. Neonatal infection of these mice with Moloney murine leukemia virus resulted in accelerated tumor onset with associated increases in clonal complexity and lymphoid dissemination. Large-scale analysis of retroviral integration sites in these tumors revealed a profound bias towards a narrow range of target genes, including Jdp2 (Jundm2), D cyclin, and Pim family genes. Remarkably, direct PCR analysis of integration hotspots revealed that every progressing tumor consisted of multiple clones harboring hits at these loci, giving access to large numbers of independent insertion events and uncovering the contrasting mutagenic mechanisms operating at each target gene. Direct PCR analysis showed that high-frequency targeting occurs only in the tumor environment in vivo and is specific for the progression gene set. These results indicate that early lymphomas in MYC/Runx2 mice remain dependent on exogenous growth signals, and that progression can be achieved by constitutive activation of pathways converging on a cell cycle checkpoint that acts as the major rate-limiting step for lymphoma outgrowth.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Genes myc , Linfoma/genética , Secuencia de Aminoácidos , Animales , Transformación Celular Viral , Progresión de la Enfermedad , Marcación de Gen , Linfoma/patología , Linfoma/virología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Datos de Secuencia Molecular , Virus de la Leucemia Murina de Moloney/genética , Mutagénesis Insercional , Proteínas Proto-Oncogénicas c-pim-1/genética , ARN Mensajero/genética , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA