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













Base de datos
Intervalo de año de publicación
1.
Acta Neuropathol ; 144(5): 1027-1048, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36070144

RESUMEN

Histone H3 mutations at amino acids 27 (H3K27M) and 34 (H3G34R) are recurrent drivers of pediatric-type high-grade glioma (pHGG). H3K27M mutations lead to global disruption of H3K27me3 through dominant negative PRC2 inhibition, while H3G34R mutations lead to local losses of H3K36me3 through inhibition of SETD2. However, their broader oncogenic mechanisms remain unclear. We characterized the H3.1K27M, H3.3K27M and H3.3G34R interactomes, finding that H3K27M is associated with epigenetic and transcription factor changes; in contrast H3G34R removes a break on cryptic transcription, limits DNA methyltransferase access, and alters mitochondrial metabolism. All 3 mutants had altered interactions with DNA repair proteins and H3K9 methyltransferases. H3K9me3 was reduced in H3K27M-containing nucleosomes, and cis-H3K9 methylation was required for H3K27M to exert its effect on global H3K27me3. H3K9 methyltransferase inhibition was lethal to H3.1K27M, H3.3K27M and H3.3G34R pHGG cells, underscoring the importance of H3K9 methylation for oncohistone-mutant gliomas and suggesting it as an attractive therapeutic target.


Asunto(s)
Glioma , Histonas , Aminoácidos/genética , Niño , ADN , Glioma/genética , Glioma/metabolismo , Histonas/genética , Humanos , Mutación/genética , Nucleosomas , Factores de Transcripción/genética
2.
Mol Cell Proteomics ; 21(10): 100411, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36089195

RESUMEN

Chromatin structure, transcription, DNA replication, and repair are regulated via locus-specific incorporation of histone variants and posttranslational modifications that guide effector chromatin-binding proteins. Here we report unbiased, quantitative interactomes for the replication-coupled (H3.1) and replication-independent (H3.3) histone H3 variants based on BioID proximity labeling, which allows interactions in intact, living cells to be detected. Along with a significant proportion of previously reported interactions detected by affinity purification followed by mass spectrometry, three quarters of the 608 histone-associated proteins that we identified are new, uncharacterized histone associations. The data reveal important biological nuances not captured by traditional biochemical means. For example, we found that the chromatin assembly factor-1 histone chaperone not only deposits the replication-coupled H3.1 histone variant during S-phase but also associates with H3.3 throughout the cell cycle in vivo. We also identified other variant-specific associations, such as with transcription factors, chromatin regulators, and with the mitotic machinery. Our proximity-based analysis is thus a rich resource that extends the H3 interactome and reveals new sets of variant-specific associations.


Asunto(s)
Chaperonas de Histonas , Histonas , Histonas/metabolismo , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Cromatina , Factor 1 de Ensamblaje de la Cromatina/genética , Factor 1 de Ensamblaje de la Cromatina/metabolismo , Factores de Transcripción/metabolismo , Nucleosomas
3.
Nat Commun ; 13(1): 588, 2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35102191

RESUMEN

High-grade diffuse glioma (HGG) is the leading cause of brain tumour death. While the genetic drivers of HGG have been well described, targeting these has thus far had little impact on survival suggesting other mechanisms are at play. Here we interrogate the alternative splicing landscape of pediatric and adult HGG through multi-omic analyses, uncovering an increased splicing burden compared with normal brain. The rate of recurrent alternative splicing in cancer drivers exceeds their mutation rate, a pattern that is recapitulated in pan-cancer analyses, and is associated with worse prognosis in HGG. We investigate potential oncogenicity by interrogating cancer pathways affected by alternative splicing in HGG; spliced cancer drivers include members of the RAS/MAPK pathway. RAS suppressor neurofibromin 1 is differentially spliced to a less active isoform in >80% of HGG downstream from REST upregulation, activating the RAS/MAPK pathway and reducing glioblastoma patient survival. Overall, our results identify non-mutagenic mechanisms by which cancers activate oncogenic pathways which need to accounted for in personalized medicine approaches.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Oncogenes/genética , Empalme del ARN/genética , Adulto , Empalme Alternativo/genética , Animales , Secuencia de Bases , Sitios de Unión , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Niño , Cromatina/metabolismo , Exones/genética , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Glioma/patología , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Mutación/genética , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Represoras/metabolismo , Empalmosomas/genética , Factores de Transcripción/metabolismo , Proteínas ras/metabolismo
4.
Nat Commun ; 11(1): 6216, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33277484

RESUMEN

Histone H3 lysine 27 (H3K27M) mutations represent the canonical oncohistone, occurring frequently in midline gliomas but also identified in haematopoietic malignancies and carcinomas. H3K27M functions, at least in part, through widespread changes in H3K27 trimethylation but its role in tumour initiation remains obscure. To address this, we created a transgenic mouse expressing H3.3K27M in diverse progenitor cell populations. H3.3K27M expression drives tumorigenesis in multiple tissues, which is further enhanced by Trp53 deletion. We find that H3.3K27M epigenetically activates a transcriptome, enriched for PRC2 and SOX10 targets, that overrides developmental and tissue specificity and is conserved between H3.3K27M-mutant mouse and human tumours. A key feature of the H3K27M transcriptome is activation of a RAS/MYC axis, which we find can be targeted therapeutically in isogenic and primary DIPG cell lines with H3.3K27M mutations, providing an explanation for the common co-occurrence of alterations in these pathways in human H3.3K27M-driven cancer. Taken together, these results show how H3.3K27M-driven transcriptome remodelling promotes tumorigenesis and will be critical for targeting cancers with these mutations.


Asunto(s)
Neoplasias Encefálicas/genética , Epigénesis Genética , Glioma/genética , Histonas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas ras/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Modelos Animales de Enfermedad , Epigenómica , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Histonas/metabolismo , Humanos , Lisina/genética , Lisina/metabolismo , Metilación , Ratones Noqueados , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/metabolismo
5.
Cancer Cell ; 37(4): 569-583.e5, 2020 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-32289278

RESUMEN

Pediatric low-grade gliomas (pLGG) are frequently driven by genetic alterations in the RAS-mitogen-activated protein kinase (RAS/MAPK) pathway yet show unexplained variability in their clinical outcome. To address this, we characterized a cohort of >1,000 clinically annotated pLGG. Eighty-four percent of cases harbored a driver alteration, while those without an identified alteration also often exhibited upregulation of the RAS/MAPK pathway. pLGG could be broadly classified based on their alteration type. Rearrangement-driven tumors were diagnosed at a younger age, enriched for WHO grade I histology, infrequently progressed, and rarely resulted in death as compared with SNV-driven tumors. Further sub-classification of clinical-molecular correlates stratified pLGG into risk categories. These data highlight the biological and clinical differences between pLGG subtypes and opens avenues for future treatment refinement.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Variaciones en el Número de Copia de ADN , Regulación Neoplásica de la Expresión Génica , Reordenamiento Génico , Glioma/genética , Mutación , Adolescente , Neoplasias Encefálicas/clasificación , Neoplasias Encefálicas/patología , Niño , Preescolar , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica , Glioma/clasificación , Glioma/patología , Humanos , Lactante , Recién Nacido , Masculino , Proteínas Quinasas Activadas por Mitógenos/genética , Neurofibromina 1/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas ras/genética
6.
Acta Neuropathol Commun ; 4(1): 93, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27577993

RESUMEN

Paediatric brain tumours arising in the thalamus present significant diagnostic and therapeutic challenges to physicians due to their sensitive midline location. As such, genetic analysis for biomarkers to aid in the diagnosis, prognosis and treatment of these tumours is needed. Here, we identified 64 thalamic gliomas with clinical follow-up and characterized targeted genomic alterations using newly optimized droplet digital and NanoString-based assays. The median age at diagnosis was 9.25 years (range, 0.63-17.55) and median survival was 6.43 (range, 0.01-27.63) years. Our cohort contained 42 and 22 tumours reviewed as low and high grade gliomas, respectively. Five (12 %) low grade and 11 (50 %) high grade gliomas were positive for the H3F3A/HIST1H3B K27M (H3K27M) mutation. Kaplan-Meier survival analysis revealed significantly worse overall survival for patients harbouring the H3K27M mutation versus H3F3A/HIST1H3B wild type (H3WT) samples (log-rank p < 0.0001) with a median survival of 1.02 vs. 9.12 years. Mitogen-activated protein kinase (MAPK) pathway activation via BRAF or FGFR1 hotspot mutations or fusion events were detected in 44 % of patients, and was associated with long-term survival in the absence of H3K27M (log-rank p < 0.0001). Multivariate analysis demonstrated H3K27M status and high grade histology to be the most significant independent predictors of poor overall survival with hazard ratios of 6.945 and 7.721 (p < 0.0001), respectively. In contrast, MAPK pathway activation is a predictor of favourable patient outcome, although not independent of other clinical factors. Importantly, we show that low grade malignancies may harbour H3K27M mutations and that these tumours show a dismal survival compared to low grade H3WT cases. Our data strongly supports the inclusion of targeted genetic testing in childhood thalamic tumours to most accurately stratify patients into appropriate risk groups.


Asunto(s)
Neoplasias Encefálicas/genética , Glioma/genética , Sistema de Señalización de MAP Quinasas/genética , Tálamo , Adolescente , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Niño , Preescolar , Femenino , Estudios de Seguimiento , Glioma/patología , Glioma/cirugía , Humanos , Lactante , Estimación de Kaplan-Meier , Masculino , Análisis Multivariante , Mutación , Clasificación del Tumor , Pronóstico , Modelos de Riesgos Proporcionales , Tálamo/patología , Tálamo/cirugía
7.
Acta Neuropathol ; 128(6): 863-77, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25120190

RESUMEN

Pediatric ependymomas are highly recurrent tumors resistant to conventional chemotherapy. Telomerase, a ribonucleoprotein critical in permitting limitless replication, has been found to be critically important for the maintenance of tumor-initiating cells (TICs). These TICs are chemoresistant, repopulate the tumor from which they are identified, and are drivers of recurrence in numerous cancers. In this study, telomerase enzymatic activity was directly measured and inhibited to assess the therapeutic potential of targeting telomerase. Telomerase repeat amplification protocol (TRAP) (n = 36) and C-circle assay/telomere FISH/ATRX staining (n = 76) were performed on primary ependymomas to determine the prevalence and prognostic potential of telomerase activity or alternative lengthening of telomeres (ALT) as telomere maintenance mechanisms, respectively. Imetelstat, a phase 2 telomerase inhibitor, was used to elucidate the effect of telomerase inhibition on proliferation and tumorigenicity in established cell lines (BXD-1425EPN, R254), a primary TIC line (E520) and xenograft models of pediatric ependymoma. Over 60 % of pediatric ependymomas were found to rely on telomerase activity to maintain telomeres, while no ependymomas showed evidence of ALT. Children with telomerase-active tumors had reduced 5-year progression-free survival (29 ± 11 vs 64 ± 18 %; p = 0.03) and overall survival (58 ± 12 vs 83 ± 15 %; p = 0.05) rates compared to those with tumors lacking telomerase activity. Imetelstat inhibited proliferation and self-renewal by shortening telomeres and inducing senescence in vitro. In vivo, Imetelstat significantly reduced subcutaneous xenograft growth by 40 % (p = 0.03) and completely abolished the tumorigenicity of pediatric ependymoma TICs in an orthotopic xenograft model. Telomerase inhibition represents a promising therapeutic approach for telomerase-active pediatric ependymomas found to characterize high-risk ependymomas.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Ependimoma/tratamiento farmacológico , Indoles/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Niacinamida/análogos & derivados , Telomerasa/antagonistas & inhibidores , Animales , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/enzimología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Preescolar , Estudios de Cohortes , Supervivencia sin Enfermedad , Ependimoma/diagnóstico , Ependimoma/enzimología , Femenino , Humanos , Ratones , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/enzimología , Trasplante de Neoplasias , Células Madre Neoplásicas/enzimología , Niacinamida/farmacología , Oligonucleótidos , Telomerasa/metabolismo , Telómero/efectos de los fármacos , Telómero/metabolismo
8.
Cancer Discov ; 4(10): 1198-213, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100205

RESUMEN

UNLABELLED: Alkylating agents are a first-line therapy for the treatment of several aggressive cancers, including pediatric glioblastoma, a lethal tumor in children. Unfortunately, many tumors are resistant to this therapy. We sought to identify ways of sensitizing tumor cells to alkylating agents while leaving normal cells unharmed, increasing therapeutic response while minimizing toxicity. Using an siRNA screen targeting over 240 DNA damage response genes, we identified novel sensitizers to alkylating agents. In particular, the base excision repair (BER) pathway, including 3-methylpurine-DNA glycosylase (MPG), as well as ataxia telangiectasia mutated (ATM), were identified in our screen. Interestingly, we identified MPG as a direct novel substrate of ATM. ATM-mediated phosphorylation of MPG was required for enhanced MPG function. Importantly, combined inhibition or loss of MPG and ATM resulted in increased alkylating agent-induced cytotoxicity in vitro and prolonged survival in vivo. The discovery of the ATM-MPG axis will lead to improved treatment of alkylating agent-resistant tumors. SIGNIFICANCE: Inhibition of ATM and MPG-mediated BER cooperate to sensitize tumor cells to alkylating agents, impairing tumor growth in vitro and in vivo with no toxicity to normal cells, providing an ideal therapeutic window.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , ADN Glicosilasas/metabolismo , Resistencia a Antineoplásicos , Factores de Edad , Animales , Línea Celular Tumoral , Análisis por Conglomerados , Variaciones en el Número de Copia de ADN , Reparación del ADN , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Activación Enzimática , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Modelos Biológicos , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/genética , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Nat Genet ; 46(5): 451-6, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24705254

RESUMEN

Diffuse intrinsic pontine glioma (DIPG) is a fatal brain cancer that arises in the brainstem of children, with no effective treatment and near 100% fatality. The failure of most therapies can be attributed to the delicate location of these tumors and to the selection of therapies on the basis of assumptions that DIPGs are molecularly similar to adult disease. Recent studies have unraveled the unique genetic makeup of this brain cancer, with nearly 80% found to harbor a p.Lys27Met histone H3.3 or p.Lys27Met histone H3.1 alteration. However, DIPGs are still thought of as one disease, with limited understanding of the genetic drivers of these tumors. To understand what drives DIPGs, we integrated whole-genome sequencing with methylation, expression and copy number profiling, discovering that DIPGs comprise three molecularly distinct subgroups (H3-K27M, silent and MYCN) and uncovering a new recurrent activating mutation affecting the activin receptor gene ACVR1 in 20% of DIPGs. Mutations in ACVR1 were constitutively activating, leading to SMAD phosphorylation and increased expression of the downstream activin signaling targets ID1 and ID2. Our results highlight distinct molecular subgroups and novel therapeutic targets for this incurable pediatric cancer.


Asunto(s)
Receptores de Activinas Tipo I/genética , Neoplasias del Tronco Encefálico/genética , Regulación Neoplásica de la Expresión Génica/genética , Genoma Humano/genética , Glioma/genética , Animales , Neoplasias del Tronco Encefálico/clasificación , Niño , Variaciones en el Número de Copia de ADN , Metilación de ADN , Perfilación de la Expresión Génica , Glioma/clasificación , Humanos , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Fosforilación , Análisis de Secuencia de ADN , Proteínas Smad/metabolismo , Pez Cebra
10.
PLoS One ; 9(3): e91540, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24626475

RESUMEN

KIF14 (kinesin family member 14) is a mitotic kinesin and an important oncogene in several cancers. Tumor KIF14 expression levels are independently predictive of poor outcome, and in cancer cells KIF14 can modulate metastatic behavior by maintaining appropriate levels of cell adhesion and migration proteins at the cell membrane. Thus KIF14 is an exciting potential therapeutic target. Understanding KIF14's regulation in cancer cells is crucial to the development of effective and selective therapies to block its tumorigenic function(s). We previously determined that close to 30% of serous ovarian cancers (OvCa tumors) exhibit low-level genomic gain, indicating one mechanism of KIF14 overexpression in tumors. We now report on transcriptional and epigenetic regulation of KIF14. Through promoter deletion analyses, we identified one cis-regulatory region containing binding sites for Sp1, HSF1 and YY1. siRNA-mediated knockdown of these transcription factors demonstrated endogenous regulation of KIF14 overexpression by Sp1 and YY1, but not HSF1. ChIP experiments confirmed an enrichment of both Sp1 and YY1 binding to the endogenous KIF14 promoter in OvCa cell lines with high KIF14 expression. A strong correlation was seen in primary serous OvCa tumors between Sp1, YY1 and KIF14 expression, further evidence that these transcription factors are important players in KIF14 overexpression. Hypomethylation patterns were observed in primary serous OvCa tumors, suggesting a minor role for promoter methylation in the control of KIF14 gene expression. miRNA expression analysis determined that miR-93, miR-144 and miR-382 had significantly lower levels of expression in primary serous OvCa tumors than normal tissues; treatment of an OvCa cell line with miRNA mimics and inhibitors specifically modulated KIF14 mRNA levels, pointing to potential novel mechanisms of KIF14 overexpression in primary tumors. Our findings reveal multiple mechanisms of KIF14 upregulation in cancer cells, offering new targets for therapeutic interventions to reduce KIF14 in tumors, aiming at improved prognosis.


Asunto(s)
Epigénesis Genética , Regulación de la Expresión Génica , Cinesinas/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias Ováricas/metabolismo , Transcripción Genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Metilación de ADN , Femenino , Eliminación de Gen , Células HeLa , Humanos , Cinesinas/genética , Mitosis , Metástasis de la Neoplasia , Proteínas Oncogénicas/genética , Neoplasias Ováricas/genética , Pronóstico , Regiones Promotoras Genéticas , ARN Interferente Pequeño/metabolismo , Factor de Transcripción Sp1/metabolismo , Regulación hacia Arriba , Factor de Transcripción YY1/metabolismo
11.
Lancet Oncol ; 14(4): 327-34, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23498719

RESUMEN

BACKGROUND: Retinoblastoma is the childhood retinal cancer that defined tumour-suppressor genes. Previous work shows that mutation of both alleles of the RB1 retinoblastoma suppressor gene initiates disease. We aimed to characterise non-familial retinoblastoma tumours with no detectable RB1 mutations. METHODS: Of 1068 unilateral non-familial retinoblastoma tumours, we compared those with no evidence of RB1 mutations (RB1(+/+)) with tumours carrying a mutation in both alleles (RB1(-/-)). We analysed genomic copy number, RB1 gene expression and protein function, retinal gene expression, histological features, and clinical data. FINDINGS: No RB1 mutations (RB1(+/+)) were reported in 29 (2·7%) of 1068 unilateral retinoblastoma tumours. 15 of the 29 RB1(+/+) tumours had high-level MYCN oncogene amplification (28-121 copies; RB1(+/+)MYCN(A)), whereas none of 93 RB1(-/-) primary tumours tested showed MYCN amplification (p<0·0001). RB1(+/+)MYCN(A) tumours expressed functional RB1 protein, had fewer overall genomic copy-number changes in genes characteristic of retinoblastoma than did RB1(-/-) tumours, and showed distinct aggressive histological features. MYCN amplification was the sole copy-number change in one RB1(+/+)MYCN(A) retinoblastoma. One additional MYCN(A) tumour was discovered after the initial frequencies were determined, and this is included in further analyses. Median age at diagnosis of the 17 children with RB1(+/+)MYCN(A) tumours was 4·5 months (IQR 3·5-10), compared with 24 months (15-37) for 79 children with non-familial unilateral RB1(-/-) retinoblastoma. INTERPRETATION: Amplification of the MYCN oncogene might initiate retinoblastoma in the presence of non-mutated RB1 genes. These unilateral RB1(+/+)MYCN(A) retinoblastomas are characterised by distinct histological features, only a few of the genomic copy-number changes that are characteristic of retinoblastoma, and very early age of diagnosis. FUNDING: National Cancer Institute-National Institutes of Health, Canadian Institutes of Health Research, German Research Foundation, Canadian Retinoblastoma Society, Hyland Foundation, Toronto Netralaya and Doctors Lions Clubs, Ontario Ministry of Health and Long Term Care, UK-Essen, and Foundations Avanti-STR and KiKa.


Asunto(s)
Dosificación de Gen , Proteínas Nucleares , Proteínas Oncogénicas , Proteína de Retinoblastoma , Retinoblastoma , Alelos , Línea Celular Tumoral , Niño , Preescolar , Femenino , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Humanos , Lactante , Mutación , Proteína Proto-Oncogénica N-Myc , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Polimorfismo de Nucleótido Simple , Retinoblastoma/genética , Retinoblastoma/metabolismo , Retinoblastoma/patología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo
12.
BMC Cancer ; 12: 69, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22336108

RESUMEN

BACKGROUND: Most human cancers show inactivation of both pRB- and p53-pathways. While retinoblastomas are initiated by loss of the RB1 tumor suppressor gene, TP53 mutations have not been found. High expression of the p53-antagonist MDM2 in human retinoblastomas may compromise p53 tumor surveillance so that TP53 mutations are not selected for in retinoblastoma tumorigenesis. We previously showed that p14ARF protein, which activates p53 by inhibiting MDM2, is low in retinoblastomas despite high mRNA expression. METHODS: In human fetal retinas, adult retinas, and retinoblastoma cells, we determined endogenous p14ARF mRNA, ARF protein, and miR-24 expression, while integrity of p53 signalling in WERI-Rb1 cells was tested using an adenovirus vector expressing p14ARF. To study p14ARF biogenesis, retinoblastoma cells were treated with the proteasome inhibitor, MG132, and siRNA against miR-24. RESULTS: In human retinoblastoma cell lines, p14ARF mRNA was disproportionally high relative to the level of p14ARF protein expression, suggesting a perturbation of p14ARF regulation. When p14ARF was over-expressed by an adenovirus vector, expression of p53 and downstream targets increased and cell growth was inhibited indicating an intact p14ARF-p53 axis. To investigate the discrepancy between p14ARF mRNA and protein in retinoblastoma, we examined p14ARF biogenesis. The proteasome inhibitor, MG132, did not cause p14ARF accumulation, although p14ARF normally is degraded by proteasomes. miR-24, a microRNA that represses p14ARF expression, is expressed in retinoblastoma cell lines and correlates with lower protein expression when compared to other cell lines with high p14ARF mRNA. Transient over-expression of siRNA against miR-24 led to elevated p14ARF protein in retinoblastoma cells. CONCLUSIONS: In retinoblastoma cells where high levels of p14ARF mRNA are not accompanied by high p14ARF protein, we found a correlation between miR-24 expression and low p14ARF protein. p14ARF protein levels were restored without change in mRNA abundance upon miR-24 inhibition suggesting that miR-24 could functionally repress expression, effectively blocking p53 tumor surveillance. During retinal tumorigenesis, miR-24 may intrinsically compromise the p53 response to RB1 loss.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/fisiología , Neoplasias de la Retina/metabolismo , Retinoblastoma/genética , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Línea Celular Tumoral , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Leupeptinas/farmacología , ARN/metabolismo , ARN Mitocondrial , ARN Interferente Pequeño/metabolismo , Retina/embriología , Retina/metabolismo , Neoplasias de la Retina/genética , Proteína p14ARF Supresora de Tumor/efectos de los fármacos , Proteína p14ARF Supresora de Tumor/genética
14.
Int J Cancer ; 130(8): 1844-54, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21618518

RESUMEN

The novel oncogene KIF14 (kinesin family member 14) shows genomic gain and overexpression in many cancers including OvCa (ovarian cancer). We discovered that expression of the mitotic kinesin KIF14 is predictive of poor outcome in breast and lung cancers. We now determine the prognostic significance of KIF14 expression in primary OvCa tumors, and evaluate KIF14 action on OvCa cell tumorigenicity in vitro. Gene-specific multiplex PCR and real-time QPCR were used to measure KIF14 genomic (109 samples) and mRNA levels (122 samples) in OvCa tumors. Association of KIF14 with clinical variables was studied using Kaplan-Meier survival and Cox regression analyses. Cellular effects of KIF14 overexpression (stable transfection) and inhibition (stable shRNA knockdown) were studied by proliferation (cell counts), survival (Annexin V immunocytochemistry) and colony formation (soft-agar growth). KIF14 genomic gain (>2.6 copies) was present in 30% of serous OvCas, and KIF14 mRNA was elevated in 91% of tumors versus normal epithelium. High KIF14 in tumors independently predicted for worse outcome (p = 0.03) with loss of correlation with proliferation marker expression and increased rates of recurrence. Overexpression of KIF14 in OvCa cell lines increased proliferation and colony formation (p < 0.01), whereas KIF14 knockdown induced apoptosis and dramatically reduced colony formation (p < 0.05). Our findings indicate that KIF14 mRNA is an independent prognostic marker in serous OvCa. Dependence of OvCa cells on KIF14 for maintenance of in vitro colony formation suggests a role of KIF14 in promoting a tumorigenic phenotype, beyond its known role in proliferation.


Asunto(s)
Biomarcadores de Tumor/genética , Cinesinas/genética , Proteínas Oncogénicas/genética , Neoplasias Ováricas/genética , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/patología , Adenocarcinoma de Células Claras/terapia , Biomarcadores de Tumor/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Cistadenocarcinoma Seroso/terapia , Variaciones en el Número de Copia de ADN , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Cinesinas/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Pronóstico , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Modelos de Riesgos Proporcionales , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/estadística & datos numéricos
15.
Invest Ophthalmol Vis Sci ; 52(10): 7618-24, 2011 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-21862643

RESUMEN

PURPOSE: Human retinoblastoma arises from an undefined developing retinal cell after inactivation of RB1. This is emulated in a murine retinoblastoma model by inactivation of pRB by retinal-specific expression of simian virus 40 large T-antigen (TAg-RB). Some mutational events after RB1 loss in humans are recapitulated at the expression level in TAg-RB, supporting preclinical evidence that this model is useful for comparative studies between mouse and human. Here, the characteristics of the TAg-RB cell of origin are defined. METHODS: TAg-RB mice were killed at ages from embryonic day (E)18 to postnatal day (P)35. Tumors were analyzed by immunostaining, DNA copy number PCR, or real-time quantitative RT-PCR for TAg protein, retinal cell type markers, and retinoblastoma-relevant genes. RESULTS: TAg expression began at P8 in a row of inner nuclear layer cells that increased in number through P21 to P28, when clusters reminiscent of small tumors emerged from cells that escaped a wave of apoptosis. Early TAg-expressing cells coexpressed the developmental marker Chx10 and glial markers CRALBP, clusterin, and carbonic anhydrase II (Car2), but not TuJ1, an early neuronal marker. Emerging tumors retained expression of only Chx10 and carbonic anhydrase II. As with human retinoblastoma, TAg-RB tumors showed decreased Cdh11 DNA copy number and gain of Kif14 and Mycn. It was confirmed that TAg-RB tumors lose expression of tumor suppressor cadherin-11 and overexpress oncogenes Kif14, Dek, and E2f3. CONCLUSIONS: TAg-RB tumors displayed molecular similarity to human retinoblastoma and origin in a cell with features of differentiated Müller glia with progenitor properties.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Regulación de la Expresión Génica/fisiología , Células Madre Neoplásicas/patología , Neuroglía/patología , Neoplasias de la Retina/patología , Retinoblastoma/patología , Animales , Apoptosis , Biomarcadores de Tumor , Caspasa 3/metabolismo , Diferenciación Celular/genética , Linaje de la Célula , Proliferación Celular , ADN de Neoplasias/genética , Técnica del Anticuerpo Fluorescente Indirecta , Silenciador del Gen/fisiología , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias de la Retina/genética , Neoplasias de la Retina/metabolismo , Retinoblastoma/genética , Retinoblastoma/metabolismo , Proteína de Retinoblastoma/genética , Tubulina (Proteína)/metabolismo
16.
PLoS Genet ; 6(4): e1000923, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20421947

RESUMEN

CDH11 gene copy number and expression are frequently lost in human retinoblastomas and in retinoblastomas arising in TAg-RB mice. To determine the effect of Cdh11 loss in tumorigenesis, we crossed Cdh11 null mice with TAg-RB mice. Loss of Cdh11 had no gross morphological effect on the developing retina of Cdh11 knockout mice, but led to larger retinal volumes in mice crossed with TAg-RB mice (p = 0.01). Mice null for Cdh11 presented with fewer TAg-positive cells at postnatal day 8 (PND8) (p = 0.01) and had fewer multifocal tumors at PND28 (p = 0.016), compared to mice with normal Cdh11 alleles. However, tumor growth was faster in Cdh11-null mice between PND8 and PND84 (p = 0.003). In tumors of Cdh11-null mice, cell death was decreased 5- to 10-fold (p<0.03 for all markers), while proliferation in vivo remained unaffected (p = 0.121). Activated caspase-3 was significantly decreased and beta-catenin expression increased in Cdh11 knockdown experiments in vitro. These data suggest that Cdh11 displays tumor suppressor properties in vivo and in vitro in murine retinoblastoma through promotion of cell death.


Asunto(s)
Apoptosis , Cadherinas/genética , Genes Supresores de Tumor , Retinoblastoma/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Mutación , Retinoblastoma/genética
18.
Biochem Biophys Res Commun ; 375(1): 1-5, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18644346

RESUMEN

It is still not clear whether the p53 pathway is altered in retinoblastoma development. We assessed the expression of the p53 pathway genes p14(ARF), mouse double minute 2 (MDM2), and mouse double minute 4 (MDM4) in human retinoblastoma compared to normal retina. Primary human retinoblastomas, retinoblastoma cell lines and normal retinas were assessed for p14(ARF) and MDM4 mRNA by quantitative RT-PCR. p14(ARF), MDM2, and MDM4 protein were measured by immunoblot and immunohistochemistry. Compared to retina, p14(ARF) mRNA expression was notably increased in retinoblastoma but p14(ARF) protein was undetectable. MDM2 and MDM4 proteins were expressed in 22/22 retinoblastomas. MDM2 was expressed in 3/10 retinas tested, and MDM4 in 10/10 retinas. The expression level of MDM2 protein in retinoblastomas and retina was comparable, while MDM4 protein was overexpressed in one retinoblastoma cell line Y79 and two primary retinoblastomas. We observe that overexpression of MDM2 and MDM4 is not a necessary step in retinoblastoma development. However, loss of detectable p14(ARF) protein and resultant lack of functional inactivation of these p53 inhibitors may contribute to retinoblastoma development by constitutive inhibition of p53.


Asunto(s)
Proteínas Proto-Oncogénicas c-mdm2/biosíntesis , Neoplasias de la Retina/metabolismo , Retinoblastoma/metabolismo , Proteína p14ARF Supresora de Tumor/biosíntesis , Adolescente , Adulto , Preescolar , Femenino , Expresión Génica , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-mdm2/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Retina/metabolismo , Neoplasias de la Retina/genética , Retinoblastoma/genética , Proteína p14ARF Supresora de Tumor/genética
19.
Gene Expr Patterns ; 5(5): 687-94, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15939381

RESUMEN

Although RB1 function is disrupted in the majority of human cancers, an undefined cell of developing human retina is uniquely sensitive to cancer induction when the RB1 tumor suppressor gene is lost. Murine retinoblastoma is initiated only when two of the RB family of genes, RB1 and p107 or p130, are inactivated. Although whole embryonic retina shows RB family gene expression by several techniques, when E14 developing retina was depleted of the earliest differentiating cells, ganglion cells, the remaining proliferating murine embryonic retinal progenitor cells clearly did not express RB1 or p130, while the longer splice form of p107 was expressed. Each retinal cell type expressed some member of the RB family at some stage of differentiation. Rod photoreceptors stained for the RB1 protein product, pRB, and p107 in only a brief window of postnatal murine development, with no detectable staining for any of the RB family proteins in adult human and mouse rod photoreceptors. Adult mouse and human Muller glia, ganglion and rare horizontal cells, and adult human, but not adult mouse, cone photoreceptors stained for pRB. The RB gene family is dynamically and variably expressed through retinal development in specific retinal cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas Nucleares/biosíntesis , Proteínas/metabolismo , Retina/embriología , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/embriología , Proteína de Retinoblastoma/biosíntesis , Proteína de Retinoblastoma/genética , Animales , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos , ARN Mensajero/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma , Proteína p130 Similar a la del Retinoblastoma , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Factores de Tiempo
20.
Cancer Res ; 62(4): 967-71, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11861365

RESUMEN

All retinoblastomas (RBs) show genomic changes in addition to loss of both RB1 alleles. Quantitative-multiplex PCR analysis identified in 41 of 70 (59%) RBs a 0.6-Mb minimal region of chromosome 6p22 gain from which we cloned the human kinesin gene, RBKIN/KIF13A, by rapid amplification of retinal cDNA. RBKIN is expressed ubiquitously in adult tissues, at low levels in fetal tissues, and at high levels in RB. Antisense RBKIN oligonucleotide reduced the growth rate of RB cell lines. RBKIN and/or another closely linked gene are candidate oncogenes contributing to malignant progression of RB.


Asunto(s)
Cromosomas Humanos Par 6/genética , Cinesinas/genética , Oncogenes/genética , Retinoblastoma/genética , División Celular/efectos de los fármacos , División Celular/genética , Clonación Molecular , ADN Complementario/genética , ADN de Neoplasias/genética , Femenino , Amplificación de Genes , Expresión Génica , Humanos , Cinesinas/biosíntesis , Masculino , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacología , Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA