Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Immunol ; 207(6): 1599-1615, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34408010

RESUMEN

GFI1 is a DNA-binding transcription factor that regulates hematopoiesis by repressing target genes through its association with complexes containing histone demethylases such as KDM1A (LSD1) and histone deacetylases (HDACs). To study the consequences of the disruption of the complex between GFI1 and histone-modifying enzymes, we have used knock-in mice harboring a P2A mutation in GFI1 coding region that renders it unable to bind LSD1 and associated histone-modifying enzymes such as HDACs. GFI1P2A mice die prematurely and show increased numbers of memory effector and regulatory T cells in the spleen accompanied by a severe systemic inflammation with high serum levels of IL-6, TNF-α, and IL-1ß and overexpression of the gene encoding the cytokine oncostatin M (OSM). We identified lung alveolar macrophages, CD8 T cell from the spleen and thymic eosinophils, and monocytes as the sources of these cytokines in GFI1P2A mice. Chromatin immunoprecipitation showed that GFI1/LSD1 complexes occupy sites at the Osm promoter and an intragenic region of the Tnfα gene and that a GFI1P2A mutant still remains bound at these sites even without LSD1. Methylation and acetylation of histone H3 at these sites were enriched in cells from GFI1P2A mice, the H3K27 acetylation being the most significant. These data suggest that the histone modification facilitated by GFI1 is critical to control inflammatory pathways in different cell types, including monocytes and eosinophils, and that a disruption of GFI1-associated complexes can lead to systemic inflammation with fatal consequences.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Histona Demetilasas/metabolismo , Proteínas Mutantes/metabolismo , Transducción de Señal/genética , Síndrome de Respuesta Inflamatoria Sistémica/sangre , Factores de Transcripción/deficiencia , Animales , Trasplante de Médula Ósea/métodos , Linfocitos T CD8-positivos/inmunología , Citocinas/sangre , Citocinas/genética , Proteínas de Unión al ADN/genética , Femenino , Expresión Génica , Técnicas de Sustitución del Gen , Histonas/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Unión Proteica , Síndrome de Respuesta Inflamatoria Sistémica/genética , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Factores de Transcripción/genética
2.
Oncologist ; 26(4): e597-e602, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33274825

RESUMEN

BACKGROUND: Fluoropyrimidines are used in chemotherapy combinations for multiple cancers. Deficient dihydropyrimidine dehydrogenase activity can lead to severe life-threatening toxicities. DPYD*2A polymorphism is one of the most studied variants. The study objective was to document the impact of implementing this test in routine clinical practice. METHODS: We retrospectively performed chart reviews of all patients who tested positive for a heterozygous or homozygous DPYD*2A mutation in samples obtained from patients throughout the province of Quebec, Canada. RESULTS: During a period of 17 months, 2,617 patients were tested: 25 patients tested positive. All were White. Twenty-four of the 25 patients were heterozygous (0.92%), and one was homozygous (0.038%). Data were available for 20 patients: 15 were tested upfront, whereas five were identified after severe toxicities. Of the five patients confirmed after toxicities, all had grade 4 cytopenias, 80% grade ≥3 mucositis, 20% grade 3 rash, and 20% grade 3 diarrhea. Eight patients identified with DPYD*2A mutation prior to treatment received fluoropyrimidine-based chemotherapy at reduced initial doses. The average fluoropyrimidine dose intensity during chemotherapy was 50%. No grade ≥3 toxicities were observed. DPYD*2A test results were available in an average of 6 days, causing no significant delays in treatment initiation. CONCLUSION: Upfront genotyping before fluoropyrimidine-based treatment is feasible in clinical practice and can prevent severe toxicities and hospitalizations without delaying treatment initiation. The administration of chemotherapy at reduced doses appears to be safe in patients heterozygous for DPYD*2A. IMPLICATIONS FOR PRACTICE: Fluoropyrimidines are part of chemotherapy combinations for multiple cancers. Deficient dihydropyrimidine dehydrogenase activity can lead to severe life-threatening toxicities. This retrospective analysis demonstrates that upfront genotyping of DPYD before fluoropyrimidine-based treatment is feasible in clinical practice and can prevent severe toxicities and hospitalizations without delaying treatment initiation. This approach was reported previously, but insufficient data concerning its application in real practice are available. This is likely the first reported experience of systematic DPYD genotyping all over Canada and North America as well.


Asunto(s)
Dihidrouracilo Deshidrogenasa (NADP) , Fluorouracilo , Antimetabolitos Antineoplásicos , Canadá , Capecitabina/efectos adversos , Dihidrouracilo Deshidrogenasa (NADP)/genética , Genotipo , Humanos , Quebec/epidemiología , Estudios Retrospectivos
3.
Haematologica ; 102(3): 484-497, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28082345

RESUMEN

Mutations in GFI1B are associated with inherited bleeding disorders called GFI1B-related thrombocytopenias. We show here that mice with a megakaryocyte-specific Gfi1b deletion exhibit a macrothrombocytopenic phenotype along a megakaryocytic dysplasia reminiscent of GFI1B-related thrombocytopenia. GFI1B deficiency increases megakaryocyte proliferation and affects their ploidy, but also abrogates their responsiveness towards integrin signaling and their ability to spread and reorganize their cytoskeleton. Gfi1b-null megakaryocytes are also unable to form proplatelets, a process independent of integrin signaling. GFI1B-deficient megakaryocytes exhibit aberrant expression of several components of both the actin and microtubule cytoskeleton, with a dramatic reduction of α-tubulin. Inhibition of FAK or ROCK, both important for actin cytoskeleton organization and integrin signaling, only partially restored their response to integrin ligands, but the inhibition of PAK, a regulator of the actin cytoskeleton, completely rescued the responsiveness of Gfi1b-null megakaryocytes to ligands, but not their ability to form proplatelets. We conclude that Gfi1b controls major functions of megakaryocytes such as integrin-dependent cytoskeleton organization, spreading and migration through the regulation of PAK activity whereas the proplatelet formation defect in GFI1B-deficient megakaryocytes is due, at least partially, to an insufficient α-tubulin content.


Asunto(s)
Citoesqueleto/metabolismo , Integrinas/metabolismo , Megacariocitos/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Actinas/química , Actinas/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Megacariocitos/efectos de los fármacos , Megacariocitos/patología , Megacariocitos/ultraestructura , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Fenotipo , Recuento de Plaquetas , Inhibidores de Proteínas Quinasas/farmacología , Multimerización de Proteína , Trombocitopenia/sangre , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Trombocitopenia/metabolismo , Transcriptoma
4.
PLoS Biol ; 12(3): e1001807, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24618719

RESUMEN

The Cut homeobox 1 (CUX1) gene is a target of loss-of-heterozygosity in many cancers, yet elevated CUX1 expression is frequently observed and is associated with shorter disease-free survival. The dual role of CUX1 in cancer is illustrated by the fact that most cell lines with CUX1 LOH display amplification of the remaining allele, suggesting that decreased CUX1 expression facilitates tumor development while increased CUX1 expression is needed in tumorigenic cells. Indeed, CUX1 was found in a genome-wide RNAi screen to identify synthetic lethal interactions with oncogenic RAS. Here we show that CUX1 functions in base excision repair as an ancillary factor for the 8-oxoG-DNA glycosylase, OGG1. Single cell gel electrophoresis (comet assay) reveals that Cux1⁺/⁻ MEFs are haploinsufficient for the repair of oxidative DNA damage, whereas elevated CUX1 levels accelerate DNA repair. In vitro base excision repair assays with purified components demonstrate that CUX1 directly stimulates OGG1's enzymatic activity. Elevated reactive oxygen species (ROS) levels in cells with sustained RAS pathway activation can cause cellular senescence. We show that elevated expression of either CUX1 or OGG1 prevents RAS-induced senescence in primary cells, and that CUX1 knockdown is synthetic lethal with oncogenic RAS in human cancer cells. Elevated CUX1 expression in a transgenic mouse model enables the emergence of mammary tumors with spontaneous activating Kras mutations. We confirmed cooperation between Kras(G12V) and CUX1 in a lung tumor model. Cancer cells can overcome the antiproliferative effects of excessive DNA damage by inactivating a DNA damage response pathway such as ATM or p53 signaling. Our findings reveal an alternate mechanism to allow sustained proliferation in RAS-transformed cells through increased DNA base excision repair capability. The heightened dependency of RAS-transformed cells on base excision repair may provide a therapeutic window that could be exploited with drugs that specifically target this pathway.


Asunto(s)
Daño del ADN , Reparación del ADN/fisiología , Genes ras/fisiología , Proteínas de Homeodominio/fisiología , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células Cultivadas , Senescencia Celular/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Mamarias Experimentales/genética , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estrés Oxidativo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción
5.
Proc Natl Acad Sci U S A ; 111(50): E5411-9, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25468973

RESUMEN

To be effective, the adaptive immune response requires a large repertoire of antigen receptors, which are generated through V(D)J recombination in lymphoid precursors. These precursors must be protected from DNA damage-induced cell death, however, because V(D)J recombination generates double-strand breaks and may activate p53. Here we show that the BTB/POZ domain protein Miz-1 restricts p53-dependent induction of apoptosis in both pro-B and DN3a pre-T cells that actively rearrange antigen receptor genes. Miz-1 exerts this function by directly activating the gene for ribosomal protein L22 (Rpl22), which binds to p53 mRNA and negatively regulates its translation. This mechanism limits p53 expression levels and thus contains its apoptosis-inducing functions in lymphocytes, precisely at differentiation stages in which V(D)J recombination occurs.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Células Progenitoras Linfoides/fisiología , Proteínas Nucleares/metabolismo , Biosíntesis de Proteínas/fisiología , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Ribosómicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Recombinación V(D)J/genética , Análisis de Varianza , Animales , Muerte Celular/fisiología , Inmunoprecipitación de Cromatina , Citometría de Flujo , Regulación de la Expresión Génica/genética , Vectores Genéticos/genética , Immunoblotting , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Biosíntesis de Proteínas/genética , Proteínas Inhibidoras de STAT Activados/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Ubiquitina-Proteína Ligasas , Recombinación V(D)J/fisiología
6.
Nucleic Acids Res ; 40(10): 4483-95, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22319212

RESUMEN

The p110 Cut homeobox 1 (CUX1) transcription factor regulates genes involved in DNA replication and chromosome segregation. Using a genome-wide-approach, we now demonstrate that CUX1 also modulates the constitutive expression of DNA damage response genes, including ones encoding ATM and ATR, as well as proteins involved in DNA damage-induced activation of, and signaling through, these kinases. Consistently, RNAi knockdown or genetic inactivation of CUX1 reduced ATM/ATR expression and negatively impacted hallmark protective responses mediated by ATM and ATR following exposure to ionizing radiation (IR) and UV, respectively. Specifically, abrogation of CUX1 strongly reduced ATM autophosphorylation after IR, in turn causing substantial decreases in (i) levels of phospho-Chk2 and p53, (ii) γ-H2AX and Rad51 DNA damage foci and (iii) the efficiency of DNA strand break repair. Similarly remarkable reductions in ATR-dependent responses, including phosphorylation of Chk1 and H2AX, were observed post-UV. Finally, multiple cell cycle checkpoints and clonogenic survival were compromised in CUX1 knockdown cells. Our results indicate that CUX1 regulates a transcriptional program that is necessary to mount an efficient response to mutagenic insult. Thus, CUX1 ensures not only the proper duplication and segregation of the genetic material, but also the preservation of its integrity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/fisiología , Proteínas Nucleares/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Puntos de Control del Ciclo Celular , Supervivencia Celular , Células Cultivadas , Roturas del ADN , Reparación del ADN/genética , Regulación de la Expresión Génica , Inestabilidad Genómica , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Humanos , Ratones , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Interferencia de ARN , Recombinasa Rad51/análisis , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transducción de Señal , Transcripción Genética
7.
Proc Natl Acad Sci U S A ; 108(5): 1949-54, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245318

RESUMEN

Cell populations able to generate a large repertoire of genetic variants have increased potential to generate tumor cells that survive through the multiple selection steps involved in tumor progression. A mechanism for the generation of aneuploid cancer cells involves passage through a tetraploid stage. Supernumerary centrosomes, however, can lead to multipolar mitosis and cell death. Using tissue culture and transgenic mouse models of breast cancer, we report that Cut homeobox 1 (CUX1) causes chromosomal instability by activating a transcriptional program that prevents multipolar divisions and enables the survival of tetraploid cells that evolve to become genetically unstable and tumorigenic. Transcriptional targets of CUX1 involved in DNA replication and bipolar mitosis defined a gene expression signature that, across 12 breast cancer gene expression datasets, was associated with poor clinical outcome. The signature not only was higher in breast tumor subtypes of worse prognosis, like the basal-like and HER2(+) subtypes, but also identified poor outcome among estrogen receptor-positive/node-negative tumors, a subgroup considered to be at lower risk. The CUX1 signature therefore represents a unique criterion to stratify patients and provides insight into the molecular determinants of poor clinical outcome.


Asunto(s)
Ciclo Celular , Inestabilidad Cromosómica/fisiología , Proteínas de Homeodominio/fisiología , Mitosis/fisiología , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Línea Celular , Replicación del ADN , Femenino , Perfilación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Factores de Transcripción
8.
BMC Genomics ; 14: 258, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23590133

RESUMEN

BACKGROUND: Overexpression of the Cut homeobox 1 gene, CUX1, inversely correlates with patient survival in breast cancers. Cell-based assays and molecular studies have revealed that transcriptional regulation by CUX1 involves mostly the proteolytically processed p110 isoform. As there is no antibody specific to p110 CUX1 only, an alternate strategy must be employed to identify its targets. RESULTS: We expressed physiological levels of a tagged-p110 CUX1 protein and performed chromatin affinity purification followed by hybridization on ENCODE and promoter arrays. Targets were validated by chromatin immunoprecipitation and transcriptional regulation by CUX1 was analyzed in expression profiling and RT-qPCR assays following CUX1 knockdown or p110 CUX1 overexpression. Approximately 47% and 14% of CUX1 binding sites were respectively mapped less than 4 Kbp, or more than 40 Kbp, away from a transcription start site. More genes exhibited changes in expression following CUX1 knockdown than p110 CUX1 overexpression. CUX1 directly activated or repressed 7.4% and 8.4% of putative targets identified on the ENCODE and promoter arrays respectively. This proportion increased to 11.2% for targets with 2 binding sites or more. Transcriptional repression was observed in a slightly higher proportion of target genes. The CUX1 consensus binding motif, ATCRAT, was found at 47.2% of the CUX1 binding sites, yet only 8.3% of the CUX1 consensus motifs present on the array were bound in vivo. The presence of a consensus binding motif did not have an impact on whether a target gene was repressed or activated. Interestingly, the distance between a binding site and a transcription start site did not significantly reduced the ability of CUX1 to regulate a target gene. Moreover, CUX1 not only was able to regulate the next adjacent gene, but also regulated the gene located beyond this one as well as the gene located further away in the opposite direction. CONCLUSION: Our results demonstrate that p110 CUX1 can activate or repress transcription when bound at a distance and can regulate more than one gene on certain genomic loci.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Sitios de Unión/genética , Inmunoprecipitación de Cromatina , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Isoformas de Proteínas/metabolismo , Análisis de Matrices Tisulares , Factores de Transcripción , Sitio de Iniciación de la Transcripción , Activación Transcripcional/fisiología
9.
Curr Oncol ; 29(2): 497-509, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35200545

RESUMEN

Background: 5-FU-based chemoradiotherapy (CRT) could be associated with severe treatment-related toxicities in patients harboring at-risk DPYD polymorphisms. Methods: The studied population included consecutive patients with locoregionally advanced oropharyngeal carcinoma treated with carboplatin and 5-FU-based CRT one year before and after the implementation of upfront DPYD*2A genotyping. We aimed to determine the effect of DPYD genotyping on grade ≥3 toxicities. Results: 181 patients were analyzed (87 patients before and 94 patients following DPYD*2A screening). Of the patients, 91% (n = 86) were prospectively genotyped for the DPYD*2A allele. Of those screened, 2% (n = 2/87) demonstrated a heterozygous DPYD*2A mutation. Extended genotyping of DPYD*2A-negative patients later allowed for the retrospective identification of six additional patients with alternative DPYD variants (two c.2846A>T and four c.1236G>A mutations). Grade ≥3 toxicities occurred in 71% of the patients before DPYD*2A screening versus 62% following upfront genotyping (p = 0.18). When retrospectively analyzing additional non-DPYD*2A variants, the relative risks for mucositis (RR 2.36 [1.39-2.13], p = 0.0063), dysphagia (RR 2.89 [1.20-5.11], p = 0.019), and aspiration pneumonia (RR 13 [2.42-61.5)], p = 0.00065) were all significantly increased. Conclusion: The DPYD*2A, c.2846A>T, and c.1236G>A polymorphisms are associated with an increased risk of grade ≥3 toxicity to 5-FU. Upfront DPYD genotyping can identify patients in whom 5-FU-related toxicity should be avoided.


Asunto(s)
Carcinoma , Dihidrouracilo Deshidrogenasa (NADP) , Quimioradioterapia/efectos adversos , Dihidrouracilo Deshidrogenasa (NADP)/genética , Genotipo , Humanos , Estudios Retrospectivos
10.
J Biol Chem ; 284(40): 27701-11, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19635798

RESUMEN

In this study, we investigated the mechanism by which the CUX1 transcription factor can stimulate cell migration and invasion. The full-length p200 CUX1 had a weaker effect than the proteolytically processed p110 isoform; moreover, treatments that affect processing similarly impacted cell migration. We conclude that the stimulatory effect of p200 CUX1 is mediated in part, if not entirely, through the generation of p110 CUX1. We established a list of putative transcriptional targets with functions related to cell motility, and we then identified those targets whose expression was directly regulated by CUX1 in a cell line whose migratory potential was strongly stimulated by CUX1. We identified 18 genes whose expression was directly modulated by p110 CUX1, and its binding to all target promoters was validated in independent chromatin immunoprecipitation assays. These genes code for regulators of Rho-GTPases, cell-cell and cell-matrix adhesion proteins, cytoskeleton-associated proteins, and markers of epithelial-to-mesenchymal transition. Interestingly, p110 CUX1 activated the expression of genes that promote cell motility and at the same time repressed genes that inhibit this process. Therefore, the role of p110 CUX1 in cell motility involves its functions in both activation and repression of transcription. This was best exemplified in the regulation of the E-cadherin gene. Indeed, we uncovered a regulatory cascade whereby p110 CUX1 binds to the snail and slug gene promoters, activates their expression, and then cooperates with these transcription factors in the repression of the E-cadherin gene, thereby causing disorganization of cell-cell junctions.


Asunto(s)
Cadherinas/genética , Movimiento Celular , Regulación hacia Abajo , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Proteínas de la Membrana/genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Transducción de Señal , Animales , Bovinos , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Inmunoprecipitación de Cromatina , Perros , Regulación de la Expresión Génica , Genes Reporteros , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/genética , Humanos , Uniones Intercelulares/metabolismo , Ratones , Proteínas Nucleares/genética , Ocludina , Regiones Promotoras Genéticas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Represoras/genética , Caracoles/genética , Factores de Transcripción , Transcripción Genética , Activación Transcripcional
11.
Nucleic Acids Res ; 36(1): 189-202, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18003658

RESUMEN

Proteolytic processing of the CUX1 transcription factor generates an isoform, p110 that accelerates entry into S phase. To identify targets of p110 CUX1 that are involved in cell cycle progression, we performed genome-wide location analysis using a promoter microarray. Since there are no antibodies that specifically recognize p110, but not the full-length protein, we expressed physiological levels of a p110 isoform with two tags and purified chromatin by tandem affinity purification (ChAP). Conventional ChIP performed on synchronized populations of cells confirmed that p110 CUX1 is recruited to the promoter of cell cycle-related targets preferentially during S phase. Multiple approaches including silencing RNA (siRNA), transient infection with retroviral vectors, constitutive expression and reporter assays demonstrated that most cell cycle targets are activated whereas a few are repressed or not affected by p110 CUX1. Functional classes that were over-represented among targets included DNA replication initiation. Consistent with this finding, constitutive expression of p110 CUX1 led to a premature and more robust induction of replication genes during cell cycle progression, and stimulated the long-term replication of a plasmid bearing the oriP replicator of Epstein Barr virus (EBV).


Asunto(s)
Replicación del ADN/genética , Proteínas de Homeodominio/fisiología , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Activación Transcripcional , Animales , Línea Celular , Cromatina/aislamiento & purificación , Cromatografía de Afinidad , Genes cdc , Genómica , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Fase S/genética , Análisis de Secuencia de ADN , Factores de Transcripción
12.
Mol Cancer ; 8: 60, 2009 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-19656388

RESUMEN

BACKGROUND: Lats1 (large tumor suppressor 1) codes for a serine/threonine kinase that plays a role in the progression through mitosis. Genetic studies demonstrated that the loss of LATS1 in mouse, and of its ortholog wts (warts) in Drosophila, is associated with increased cancer incidence. There are conflicting reports, however, as to whether overexpression of Lats1 inhibits cell proliferation. CUX1 is a transcription factor that exists in different isoforms as a result of proteolytic processing or alternative transcription initiation. Expression of p110 and p75 CUX1 in transgenic mice increases the susceptibility to cancer in various organs and tissues. In tissue culture, p110 CUX1 was shown to accelerate entry into S phase and stimulate cell proliferation. RESULTS: Genome-wide location arrays in cell lines of various cell types revealed that Lats1 was a transcriptional target of CUX1. Scanning ChIP analysis confirmed that CUX1 binds to the immediate promoter of Lats1. Expression of Lats1 was reduced in cux1-/- MEFs, whereas it was increased in cells stably or transiently expressing p110 or p75 CUX1. Reporter assays confirmed that the immediate promoter of Lats1 was sufficient to confer transcriptional activation by CUX1. Lats1 was found to be overexpressed in tumors from the mammary gland, uterus and spleen that arise in p110 or p75 CUX1 transgenic mice. In tissue culture, such elevated LATS1 expression did not hinder cell cycle progression in cells overexpressing p110 CUX1. CONCLUSION: While inactivation of Lats1/wts in mouse and Drosophila can increase cancer incidence, results from the present study demonstrate that Lats1 is a transcriptional target of CUX1 that can be overexpressed in tumors of various tissue-types. Interestingly, two other studies documented the overexpression of LATS1 in human cervical cancers and basal-like breast cancers. We conclude that, similarly to other genes involved in mitotic checkpoint, cancer can be associated with either loss-of-function or overexpression of Lats1.


Asunto(s)
Genes Supresores de Tumor , Proteínas de Homeodominio/genética , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/genética , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Femenino , Proteínas de Homeodominio/biosíntesis , Humanos , Ratones , Ratones Transgénicos , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/biosíntesis , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Represoras/biosíntesis , Transcripción Genética , Activación Transcripcional
13.
Sci Rep ; 9(1): 6304, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-31004086

RESUMEN

Here we demonstrate a mode of reciprocal regulation between GFI1 and p53 that controls the induction of apoptosis in T cells. We show that GFI1 prevents induction of p53 dependent apoptosis by recruiting LSD1 to p53, which leads to the demethylation of its C-terminal domain. This is accompanied by a decrease of the acetylation of lysine 117 within the core domain of the murine p53 protein, which is required for transcriptional induction of apoptosis. Our results support a model in which the effect of GFI1's regulation of methylation at the c-terminus of p53 is ultimately mediated through control of acetylation at lysine 117 of p53. We propose that GFI1 acts prior to the occurrence of DNA damage to affect the post-translational modification state and limit the subsequent activation of p53. Once activated, p53 then transcriptionally activates GFI1, presumably in order to re-establish the homeostatic balance of p53 activity. These findings have implications for the activity level of p53 in various disease contexts where levels of GFI1 are either increased or decreased.


Asunto(s)
Apoptosis , Proteínas de Unión al ADN/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de Unión al ADN/genética , Ratones , Ratones Noqueados , Linfocitos T/citología , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética
14.
Leukemia ; 33(1): 110-121, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29925903

RESUMEN

Growth factor independent 1 (Gfi1) controls myeloid differentiation by regulating gene expression and limits the activation of p53 by facilitating its de-methylation at Lysine 372. In human myeloid leukemia, low GFI1 levels correlate with an inferior prognosis. Here, we show that knockdown (KD) of Gfi1 in mice causes a fatal myeloproliferative disease (MPN) that could progress to leukemia after additional mutations. Both KO and KD mice accumulate myeloid cells that show signs of metabolic stress and high levels of reactive oxygen species. However, only KO cells have elevated levels of Lysine 372 methylated p53. This suggests that in contrast to absence of GFI1, KD of GFI1 leads to the accumulation of myeloid cells because sufficient amount of GFI1 is present to impede p53-mediated cell death, leading to a fatal MPN. The combination of myeloid accumulation and the ability to counteract p53 activity under metabolic stress could explain the role of reduced GF1 expression in human myeloid leukemia.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/fisiología , Leucemia Mieloide/patología , Células Mieloides/patología , Trastornos Mieloproliferativos/patología , Factores de Transcripción/fisiología , Animales , Leucemia Mieloide/etiología , Leucemia Mieloide/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Trastornos Mieloproliferativos/etiología , Trastornos Mieloproliferativos/metabolismo , Estrés Oxidativo , Canales Catiónicos TRPC/fisiología
15.
Cancer Res ; 79(16): 4184-4195, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31273062

RESUMEN

Acute lymphoblastic leukemia (ALL) is an aggressive blood cancer that mainly affects children. Relapse rates are high and toxic chemotherapies that block DNA replication and induce DNA damage lead to health problems later in life, underlining the need for improved therapies. MYC is a transcription factor that is hyperactive in a large proportion of cancers including leukemia but is difficult to target in therapy. We show that ablation of the function of the BTB/POZ domain factor Zbtb17 (Miz-1), an important cofactor of c-Myc, significantly delayed T- and B-ALL/lymphoma in mice and interfered with the oncogenic transcriptional activity of c-Myc. Leukemic cells that still emerged in this system activated DNA replication pathways that could be targeted by current chemotherapeutic drugs such as cytarabine. Acute ablation of the Miz-1 POZ domain enhanced the effect of cytarabine treatment. The combined treatment was effective in both Eµ-Myc and Notch ICN-driven leukemia models and prolonged survival of tumor-bearing animals by accelerating apoptosis of leukemic cells. These observations suggest that targeting MIZ-1 could render current ALL chemotherapies more effective, with a better outcome for patients. SIGNIFICANCE: Ablation of the POZ domain of Miz-1 perturbs its interaction with c-MYC and delays the generation of T- and B-cell leukemias and lymphomas.


Asunto(s)
Citarabina/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Proteínas Inhibidoras de STAT Activados/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Antimetabolitos Antineoplásicos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Genes myc , Ratones Transgénicos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Dominios Proteicos , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
16.
Nat Commun ; 10(1): 1270, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894540

RESUMEN

Gfi1b is a transcriptional repressor expressed in hematopoietic stem cells (HSCs) and megakaryocytes (MKs). Gfi1b deficiency leads to expansion of both cell types and abrogates the ability of MKs to respond to integrin. Here we show that Gfi1b forms complexes with ß-catenin, its co-factors Pontin52, CHD8, TLE3 and CtBP1 and regulates Wnt/ß-catenin-dependent gene expression. In reporter assays, Gfi1b can activate TCF-dependent transcription and Wnt3a treatment enhances this activation. This requires interaction between Gfi1b and LSD1 and suggests that a tripartite ß-catenin/Gfi1b/LSD1 complex exists, which regulates Wnt/ß-catenin target genes. Consistently, numerous canonical Wnt/ß-catenin target genes, co-occupied by Gfi1b, ß-catenin and LSD1, have their expression deregulated in Gfi1b-deficient cells. When Gfi1b-deficient cells are treated with Wnt3a, their normal cellularity is restored and Gfi1b-deficient MKs regained their ability to spread on integrin substrates. This indicates that Gfi1b controls both the cellularity and functional integrity of HSCs and MKs by regulating Wnt/ß-catenin signaling pathway.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Megacariocitos/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Vía de Señalización Wnt , Proteína Wnt3A/genética , beta Catenina/genética , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Ontología de Genes , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células Madre Hematopoyéticas/citología , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Humanos , Células K562 , Megacariocitos/citología , Ratones , Ratones Noqueados , Anotación de Secuencia Molecular , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Represoras/deficiencia , Tamoxifeno , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
17.
Cell Stress ; 2(8): 213-215, 2018 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-31225488

RESUMEN

Despite recent advances in cancer treatment through personalized and precision medicine and new avenues such as immunotherapy and chimeric antibodies, the induction of DNA damage either through irradiation or specific compounds remains the primary approach to kill tumour cells. Improvements in our understanding of how tumour cells respond to DNA damage, and especially how this response differs from that of normal cells, are crucial to the development of better and more efficient therapies. We have recently shown that the activity of the oncogenic transcription factor GFI1, which is required for the development and maintenance of T and B cell leukemia, increases the ability of tumour cells to repair their DNA following damage (Vadnais et al. Nat Commun 9(1):1418). GFI1 accomplishes this by regulating the post-translational modifications (PTM) of key DNA repair proteins, including MRE11 and 53BP1, by the methyltransferase PRMT1. Here, GFI1 acts as an accessory protein required for the interaction between the enzyme and its substrates. This has implications for the treatment response of tumour cells overexpressing GFI1, which includes T cell leukemia, neuroendocrine lung carcinomas and aggressive subtypes of medulloblastoma, and suggests that targeting GFI1's activity and with this its capacity to aid DNA repair may open avenues for new therapeutic approaches.

18.
Nat Commun ; 9(1): 1418, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29651020

RESUMEN

GFI1 is a transcriptional regulator expressed in lymphoid cells, and an "oncorequisite" factor required for development and maintenance of T-lymphoid leukemia. GFI1 deletion causes hypersensitivity to ionizing radiation, for which the molecular mechanism remains unknown. Here, we demonstrate that GFI1 is required in T cells for the regulation of key DNA damage signaling and repair proteins. Specifically, GFI1 interacts with the arginine methyltransferase PRMT1 and its substrates MRE11 and 53BP1. We demonstrate that GFI1 enables PRMT1 to bind and methylate MRE11 and 53BP1, which is necessary for their function in the DNA damage response. Thus, our results provide evidence that GFI1 can adopt non-transcriptional roles, mediating the post-translational modification of proteins involved in DNA repair. These findings have direct implications for treatment responses in tumors overexpressing GFI1 and suggest that GFI1's activity may be a therapeutic target in these malignancies.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de la radiación , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Proteína Homóloga de MRE11/metabolismo , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Daño del ADN , Proteínas de Unión al ADN/genética , Rayos gamma , Humanos , Células Jurkat , Proteína Homóloga de MRE11/genética , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Represoras/genética , Transducción de Señal , Factores de Transcripción/genética , Transcripción Genética , Proteína 1 de Unión al Supresor Tumoral P53/genética
19.
PLoS One ; 11(7): e0160344, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27467586

RESUMEN

A regulatory circuit that controls myeloid versus B lymphoid cell fate in hematopoietic progenitors has been proposed, in which a network of the transcription factors Egr1/2, Nab, Gfi1 and PU.1 forms the core element. Here we show that a direct link between Gfi1, the transcription factor E2A and its inhibitor Id1 is a critical element of this regulatory circuit. Our data suggest that a certain threshold of Gfi1 is required to gauge E2A activity by adjusting levels of Id1 in multipotent progenitors, which are the first bipotential myeloid/lymphoid-restricted progeny of hematopoietic stem cells. If Gfi1 levels are high, Id1 is repressed enabling E2A to activate a specific set of B lineage genes by binding to regulatory elements for example the IL7 receptor gene. If Gfi1 levels fall below a threshold, Id1 expression increases and renders E2A unable to function, which prevents hematopoietic progenitors from engaging along the B lymphoid lineage.


Asunto(s)
Linfocitos B/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Factores de Transcripción/metabolismo , Animales , Linfocitos B/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos
20.
Sci Rep ; 6: 27379, 2016 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-27271479

RESUMEN

The proliferation and survival of hematopoietic stem cells (HSCs) has to be strictly coordinated to ensure the timely production of all blood cells. Here we report that the splice factor and RNA binding protein hnRNP L (heterogeneous nuclear ribonucleoprotein L) is required for hematopoiesis, since its genetic ablation in mice reduces almost all blood cell lineages and causes premature death of the animals. In agreement with this, we observed that hnRNP L deficient HSCs lack both the ability to self-renew and foster hematopoietic differentiation in transplanted hosts. They also display mitochondrial dysfunction, elevated levels of γH2AX, are Annexin V positive and incorporate propidium iodide indicating that they undergo cell death. Lin(-)c-Kit(+) fetal liver cells from hnRNP L deficient mice show high p53 protein levels and up-regulation of p53 target genes. In addition, cells lacking hnRNP L up-regulated the expression of the death receptors TrailR2 and CD95/Fas and show Caspase-3, Caspase-8 and Parp cleavage. Treatment with the pan-caspase inhibitor Z-VAD-fmk, but not the deletion of p53, restored cell survival in hnRNP L deficient cells. Our data suggest that hnRNP L is critical for the survival and functional integrity of HSCs by restricting the activation of caspase-dependent death receptor pathways.


Asunto(s)
Supervivencia Celular/fisiología , Células Madre Hematopoyéticas/citología , Ribonucleoproteína Heterogénea-Nuclear Grupo L/fisiología , Animales , Apoptosis/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo L/genética , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Estrés Fisiológico , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA