Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Cancer ; 153(1): 183-196, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36912284

RESUMEN

Fanconi anemia (FA) is a heritable malformation, bone marrow failure and cancer predisposition syndrome that confers an exceptionally high risk of squamous carcinomas. These carcinomas originate in epithelia lining the mouth, proximal esophagus, vulva and anus: their origins are not understood, and no effective ways have been identified to prevent or delay their appearance. Many FA-associated carcinomas are also therapeutically challenging: they may be multi-focal and stage-advanced at diagnosis, and most individuals with FA cannot tolerate standard-of-care systemic therapies such as DNA cross-linking drugs or ionizing radiation due to constitutional DNA damage hypersensitivity. We developed the Fanconi Anemia Cancer Cell Line Resource (FA-CCLR) to foster new work on the origins, treatment and prevention of FA-associated carcinomas. The FA-CCLR consists of Fanconi-isogenic head and neck squamous cell carcinoma (HNSCC) cell line pairs generated from five individuals with FA-associated HNSCC, and five individuals with sporadic HNSCC. Sporadic, isogenic HNSCC cell line pairs were generated in parallel with FA patient-derived isogenic cell line pairs to provide comparable experimental material to use to identify cell and molecular phenotypes driven by germline or somatic loss of Fanconi pathway function, and the subset of these FA-dependent phenotypes that can be modified, complemented or suppressed. All 10 FANC-isogenic cell line pairs are available to academic, non-profit and industry investigators via the "Fanconi Anemia Research Materials" Resource and Repository at Oregon Health & Sciences University, Portland OR.


Asunto(s)
Carcinoma de Células Escamosas , Anemia de Fanconi , Neoplasias de Cabeza y Cuello , Femenino , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Anemia de Fanconi/genética , Anemia de Fanconi/complicaciones , Anemia de Fanconi/patología , Ciencia Traslacional Biomédica , Neoplasias de Cabeza y Cuello/genética , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral
2.
Nature ; 470(7332): 105-9, 2011 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-21151107

RESUMEN

Studies in embryonic development have guided successful efforts to direct the differentiation of human embryonic and induced pluripotent stem cells (PSCs) into specific organ cell types in vitro. For example, human PSCs have been differentiated into monolayer cultures of liver hepatocytes and pancreatic endocrine cells that have therapeutic efficacy in animal models of liver disease and diabetes, respectively. However, the generation of complex three-dimensional organ tissues in vitro remains a major challenge for translational studies. Here we establish a robust and efficient process to direct the differentiation of human PSCs into intestinal tissue in vitro using a temporal series of growth factor manipulations to mimic embryonic intestinal development. This involved activin-induced definitive endoderm formation, FGF/Wnt-induced posterior endoderm pattering, hindgut specification and morphogenesis, and a pro-intestinal culture system to promote intestinal growth, morphogenesis and cytodifferentiation. The resulting three-dimensional intestinal 'organoids' consisted of a polarized, columnar epithelium that was patterned into villus-like structures and crypt-like proliferative zones that expressed intestinal stem cell markers. The epithelium contained functional enterocytes, as well as goblet, Paneth and enteroendocrine cells. Using this culture system as a model to study human intestinal development, we identified that the combined activity of WNT3A and FGF4 is required for hindgut specification whereas FGF4 alone is sufficient to promote hindgut morphogenesis. Our data indicate that human intestinal stem cells form de novo during development. We also determined that NEUROG3, a pro-endocrine transcription factor that is mutated in enteric anendocrinosis, is both necessary and sufficient for human enteroendocrine cell development in vitro. PSC-derived human intestinal tissue should allow for unprecedented studies of human intestinal development and disease.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Intestinos/citología , Activinas/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Tipificación del Cuerpo/efectos de los fármacos , Técnicas de Cultivo de Célula , Células Cultivadas , Medios de Cultivo/química , Medios de Cultivo/farmacología , Células Madre Embrionarias/efectos de los fármacos , Endodermo/citología , Endodermo/efectos de los fármacos , Endodermo/embriología , Factor 4 de Crecimiento de Fibroblastos/farmacología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Intestinos/anatomía & histología , Intestinos/efectos de los fármacos , Intestinos/embriología , Microvellosidades/efectos de los fármacos , Morfogénesis/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Organogénesis/efectos de los fármacos , Factores de Tiempo , Proteínas Wnt/farmacología , Proteína Wnt3 , Proteína Wnt3A
3.
J Virol ; 88(19): 11315-26, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25031356

RESUMEN

UNLABELLED: DNA repair plays a crucial role in embryonic and somatic stem cell biology and cell reprogramming. The Fanconi anemia (FA) pathway, which promotes error-free repair of DNA double-strand breaks, is required for somatic cell reprogramming to induced pluripotent stem cells (iPSC). Thus, cells from Fanconi anemia patients, which lack this critical pathway, fail to be reprogrammed to iPSC under standard conditions unless the defective FA gene is complemented. In this study, we utilized the oncogenes of high-risk human papillomavirus 16 (HPV16) to overcome the resistance of FA patient cells to reprogramming. We found that E6, but not E7, recovers FA iPSC colony formation and, furthermore, that p53 inhibition is necessary and sufficient for this activity. The iPSC colonies resulting from each of these approaches stained positive for alkaline phosphatase, NANOG, and Tra-1-60, indicating that they were fully reprogrammed into pluripotent cells. However, FA iPSC were incapable of outgrowth into stable iPSC lines regardless of p53 suppression, whereas their FA-complemented counterparts grew efficiently. Thus, we conclude that the FA pathway is required for the growth of iPSC beyond reprogramming and that p53-independent mechanisms are involved. IMPORTANCE: A novel approach is described whereby HPV oncogenes are used as tools to uncover DNA repair-related molecular mechanisms affecting somatic cell reprogramming. The findings indicate that p53-dependent mechanisms block FA cells from reprogramming but also uncover a previously unrecognized defect in FA iPSC proliferation independent of p53.


Asunto(s)
Reprogramación Celular/genética , Anemia de Fanconi/genética , Células Madre Pluripotentes Inducidas/virología , Queratinocitos/virología , Proteínas Oncogénicas Virales/genética , Proteínas Represoras/genética , Proteína p53 Supresora de Tumor/genética , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Roturas del ADN de Doble Cadena , Reparación del ADN , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Queratinocitos/metabolismo , Queratinocitos/patología , Proteína Homeótica Nanog , Proteínas Oncogénicas Virales/metabolismo , Cultivo Primario de Células , Proteoglicanos/genética , Proteoglicanos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo , Transducción Genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo
4.
Mol Ther Methods Clin Dev ; 32(2): 101213, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38596536

RESUMEN

Pulmonary macrophage transplantation (PMT) is a gene and cell transplantation approach in development as therapy for hereditary pulmonary alveolar proteinosis (hPAP), a surfactant accumulation disorder caused by mutations in CSF2RA/B (and murine homologs). We conducted a toxicology study of PMT of Csf2ra gene-corrected macrophages (mGM-Rα+Mϕs) or saline-control intervention in Csf2raKO or wild-type (WT) mice including single ascending dose and repeat ascending dose studies evaluating safety, tolerability, pharmacokinetics, and pharmacodynamics. Lentiviral-mediated Csf2ra cDNA transfer restored GM-CSF signaling in mGM-Rα+Mϕs. Following PMT, mGM-Rα+Mϕs engrafted, remained within the lungs, and did not undergo uncontrolled proliferation or result in bronchospasm, pulmonary function abnormalities, pulmonary or systemic inflammation, anti-transgene product antibodies, or pulmonary fibrosis. Aggressive male fighting caused a similarly low rate of serious adverse events in saline- and PMT-treated mice. Transient, minor pulmonary neutrophilia and exacerbation of pre-existing hPAP-related lymphocytosis were observed 14 days after PMT of the safety margin dose but not the target dose (5,000,000 or 500,000 mGM-Rα+Mϕs, respectively) and only in Csf2raKO mice but not in WT mice. PMT reduced lung disease severity in Csf2raKO mice. Results indicate PMT of mGM-Rα+Mϕs was safe, well tolerated, and therapeutically efficacious in Csf2raKO mice, and established a no adverse effect level and 10-fold safety margin.

5.
J Virol ; 86(15): 8131-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623785

RESUMEN

High-risk human papillomaviruses (HPVs) deregulate epidermal differentiation and cause anogenital and head and neck squamous cell carcinomas (SCCs). The E7 gene is considered the predominant viral oncogene and drives proliferation and genome instability. While the implementation of routine screens has greatly reduced the incidence of cervical cancers which are almost exclusively HPV positive, the proportion of HPV-positive head and neck SCCs is on the rise. High levels of HPV oncogene expression and genome load are linked to disease progression, but genetic risk factors that regulate oncogene abundance and/or genome amplification remain poorly understood. Fanconi anemia (FA) is a genome instability syndrome characterized at least in part by extreme susceptibility to SCCs. FA results from mutations in one of 15 genes in the FA pathway, whose protein products assemble in the nucleus and play important roles in DNA damage repair. We report here that loss of FA pathway components FANCA and FANCD2 stimulates E7 protein accumulation in human keratinocytes and causes increased epithelial proliferation and basal cell layer expansion in the HPV-positive epidermis. Additionally, FANCD2 loss stimulates HPV genome amplification in differentiating cells, demonstrating that the intact FA pathway functions to restrict the HPV life cycle. These findings raise the possibility that FA genes suppress HPV infection and disease and suggest possible mechanism(s) for reported associations of HPV with an FA cohort in Brazil and for allelic variation of FA genes with HPV persistence in the general population.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Genoma Viral/fisiología , Papillomavirus Humano 16/fisiología , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/metabolismo , Replicación Viral/fisiología , Brasil/epidemiología , Carcinoma de Células Escamosas/epidemiología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/virología , Línea Celular Transformada , Anemia de Fanconi/epidemiología , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Anemia de Fanconi/virología , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Femenino , Neoplasias de Cabeza y Cuello/epidemiología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/virología , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Queratinocitos/virología , Masculino , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/epidemiología , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/patología
6.
Sci Rep ; 12(1): 45, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997070

RESUMEN

Head-and-neck squamous cell carcinomas (HNSCCs) are relatively common in patients with Fanconi anemia (FA), a hereditary chromosomal instability disorder. Standard chemo-radiation therapy is not tolerated in FA due to an overall somatic hypersensitivity to such treatment. The question is how to find a suitable alternative treatment. We used whole-exome and whole genome mRNA sequencing to identify major genomic and transcriptomic events associated with FA-HNSCC. CRISPR-engineered FA-knockout models were used to validate a number of top hits that were likely to be druggable. We identified deletion of 18q21.2 and amplification of 11q22.2 as prevailing copy-number alterations in FA HNSCCs, the latter of which was associated with strong overexpression of the cancer-related genes YAP1, BIRC2, BIRC3 (at 11q22.1-2). We then found the drug AZD5582, a known small molecule inhibitor of BIRC2-3, to selectively kill FA tumor cells that overexpressed BIRC2-3. This occurred at drug concentrations that did not affect the viability of untransformed FA cells. Our data indicate that 11q22.2 amplifications are relatively common oncogenic events in FA-HNSCCs, as holds for non FA-HNSCC. Therefore, chemotherapeutic inhibition of overexpressed BIRC2-3 may provide the basis for an approach to develop a clinically realistic treatment of FA-HNSCCs that carry 11q22.2 amplifications.


Asunto(s)
Proteína 3 que Contiene Repeticiones IAP de Baculovirus/genética , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/metabolismo , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/genética , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Alquinos/farmacología , Proteína 3 que Contiene Repeticiones IAP de Baculovirus/antagonistas & inhibidores , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Anemia de Fanconi/complicaciones , Anemia de Fanconi/inmunología , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/complicaciones , Neoplasias de Cabeza y Cuello/inmunología , Humanos , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oligopéptidos/farmacología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Proteínas Señalizadoras YAP/genética , Proteínas Señalizadoras YAP/metabolismo
7.
Am J Pathol ; 174(1): 71-81, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19036808

RESUMEN

Overexpression of the DEK gene is associated with multiple human cancers, but its specific roles as a putative oncogene are not well defined. DEK transcription was previously shown to be induced by the high-risk human papillomavirus (HPV) E7 oncogene via E2F and Rb pathways. Transient DEK overexpression was able to inhibit both senescence and apoptosis in cultured cells. In at least the latter case, this mechanism involved the destabilization of p53 and the decreased expression of p53 target genes. We show here that DEK overexpression disrupts the normal differentiation program in a manner that is independent of either p53 or cell death. DEK expression was distinctly repressed upon the differentiation of cultured primary human keratinocytes, and stable DEK overexpression caused epidermal thickening in an organotypic raft model system. The observed hyperplasia involved a delay in keratinocyte differentiation toward a more undifferentiated state, and expansion of the basal cell compartment was due to increased proliferation, but not apoptosis. These phenotypes were accompanied by elevated p63 expression in the absence of p53 destabilization. In further support of bona fide oncogenic DEK activities, we report here up-regulated DEK protein levels in both human papilloma virus-positive hyperplastic murine skin and a subset of human squamous cell carcinomas. We suggest that DEK up-regulation may contribute to carcinoma development at least in part through increased proliferation and retardation of differentiation.


Asunto(s)
Diferenciación Celular/genética , Transformación Celular Neoplásica/genética , Proteínas Cromosómicas no Histona/biosíntesis , Células Epiteliales/citología , Queratinocitos/citología , Proteínas Oncogénicas/biosíntesis , Animales , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/virología , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Epitelio/metabolismo , Epitelio/patología , Técnica del Anticuerpo Fluorescente , Prepucio/citología , Expresión Génica , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/virología , Queratinocitos/patología , Queratinocitos/virología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas de Unión a Poli-ADP-Ribosa , Proto-Oncogenes Mas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
8.
Sci Rep ; 10(1): 20107, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208894

RESUMEN

Our understanding of how projected climatic warming will influence the world's biota remains largely speculative, owing to the many ways in which it can directly and indirectly affect individual phenotypes. Its impact is expected to be especially severe in the tropics, where organisms have evolved in more physically stable conditions relative to temperate ecosystems. Lake Tanganyika (eastern Africa) is one ecosystem experiencing rapid warming, yet our understanding of how its diverse assemblage of endemic species will respond is incomplete. Herein, we conducted a laboratory experiment to assess how anticipated future warming would affect the mirror-elicited aggressive behaviour of Julidochromis ornatus, a common endemic cichlid in Lake Tanganyika. Given linkages that have been established between temperature and individual behaviour in fish and other animals, we hypothesized that water warming would heighten average individual aggression. Our findings support this hypothesis, suggesting the potential for water warming to mediate behavioural phenotypic expression through negative effects associated with individual health (body condition). We ultimately discuss the implications of our findings for efforts aimed at understanding how continued climate warming will affect the ecology of Lake Tanganyika fishes and other tropical ectotherms.


Asunto(s)
Agresión/fisiología , Cíclidos , Animales , Conducta Animal , Tamaño Corporal , Cíclidos/fisiología , Cambio Climático , Femenino , Lagos , Masculino , Tanzanía , Temperatura
10.
Clin Cancer Res ; 22(8): 2062-73, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26603260

RESUMEN

PURPOSE: Head and neck squamous cell carcinoma (HNSCC) remains a devastating disease, and Fanconi anemia (FA) gene mutations and transcriptional repression are common. Invasive tumor behavior is associated with poor outcome, but relevant pathways triggering invasion are poorly understood. There is a significant need to improve our understanding of genetic pathways and molecular mechanisms driving advanced tumor phenotypes, to develop tailored therapies. Here we sought to investigate the phenotypic and molecular consequences of FA pathway loss in HNSCC cells. EXPERIMENTAL DESIGN: Using sporadic HNSCC cell lines with and without FA gene knockdown, we sought to characterize the phenotypic and molecular consequences of FA deficiency. FA pathway inactivation was confirmed by the detection of classic hallmarks of FA following exposure to DNA cross-linkers. Cells were subjected to RNA sequencing with qRT-PCR validation, followed by cellular adhesion and invasion assays in the presence and absence of DNA-dependent protein kinase (DNA-PK) and Rac1 inhibitors. RESULTS: We demonstrate that FA loss in HNSCC cells leads to cytoskeletal reorganization and invasive tumor cell behavior in the absence of proliferative gains. We further demonstrate that cellular invasion following FA loss is mediated, at least in part, through NHEJ-associated DNA-PK and downstream Rac1 GTPase activity. CONCLUSIONS: These findings demonstrate that FA loss stimulates HNSCC cell motility and invasion, and implicate a targetable DNA-PK/Rac1 signaling axis in advanced tumor phenotypes.


Asunto(s)
Proteína Quinasa Activada por ADN/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Transducción de Señal , Proteína de Unión al GTP rac1/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Biología Computacional/métodos , Citoesqueleto/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ontología de Genes , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Fenotipo , Carcinoma de Células Escamosas de Cabeza y Cuello
11.
Clin Cancer Res ; 21(8): 1962-72, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25609062

RESUMEN

PURPOSE: Fanconi anemia is an inherited disorder associated with a constitutional defect in the Fanconi anemia DNA repair machinery that is essential for resolution of DNA interstrand crosslinks. Individuals with Fanconi anemia are predisposed to formation of head and neck squamous cell carcinomas (HNSCC) at a young age. Prognosis is poor, partly due to patient intolerance of chemotherapy and radiation requiring dose reduction, which may lead to early recurrence of disease. EXPERIMENTAL DESIGN: Using HNSCC cell lines derived from the tumors of patients with Fanconi anemia, and murine HNSCC cell lines derived from the tumors of wild-type and Fancc(-/-) mice, we sought to define Fanconi anemia-dependent chemosensitivity and DNA repair characteristics. We utilized DNA repair reporter assays to explore the preference of Fanconi anemia HNSCC cells for non-homologous end joining (NHEJ). RESULTS: Surprisingly, interstrand crosslinker (ICL) sensitivity was not necessarily Fanconi anemia-dependent in human or murine cell systems. Our results suggest that the increased Ku-dependent NHEJ that is expected in Fanconi anemia cells did not mediate relative ICL resistance. ICL exposure resulted in increased DNA damage sensing and repair by PARP in Fanconi anemia-deficient cells. Moreover, human and murine Fanconi anemia HNSCC cells were sensitive to PARP inhibition, and sensitivity of human cells was attenuated by Fanconi anemia gene complementation. CONCLUSIONS: The observed reliance upon PARP-mediated mechanisms reveals a means by which Fanconi anemia HNSCCs can acquire relative resistance to the ICL-based chemotherapy that is a foundation of HNSCC treatment, as well as a potential target for overcoming chemoresistance in the chemosensitive individual.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Anemia de Fanconi/complicaciones , Anemia de Fanconi/genética , Neoplasias de Cabeza y Cuello/etiología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular , Cisplatino/farmacología , Daño del ADN/efectos de los fármacos , Reparación del ADN por Unión de Extremidades , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Técnicas de Inactivación de Genes , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Xenoinjertos , Humanos , Autoantígeno Ku , Ratones , Ratones Noqueados , Fenotipo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Esferoides Celulares , Células Tumorales Cultivadas
12.
Cancer Epidemiol Biomarkers Prev ; 24(5): 864-72, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25809863

RESUMEN

BACKGROUND: Fanconi anemia is a rare genetic disorder resulting in a loss of function of the Fanconi anemia-related DNA repair pathway. Individuals with Fanconi anemia are predisposed to some cancers, including oropharyngeal and gynecologic cancers, with known associations with human papillomavirus (HPV) in the general population. As individuals with Fanconi anemia respond poorly to chemotherapy and radiation, prevention of cancer is critical. METHODS: To determine whether individuals with Fanconi anemia are particularly susceptible to oral HPV infection, we analyzed survey-based risk factor data and tested DNA isolated from oral rinses from 126 individuals with Fanconi anemia and 162 unaffected first-degree family members for 37 HPV types. RESULTS: Fourteen individuals (11.1%) with Fanconi anemia tested positive, significantly more (P = 0.003) than family members (2.5%). While HPV prevalence was even higher for sexually active individuals with Fanconi anemia (17.7% vs. 2.4% in family; P = 0.003), HPV positivity also tended to be higher in the sexually inactive (8.7% in Fanconi anemia vs. 2.9% in siblings). Indeed, having Fanconi anemia increased HPV positivity 4.9-fold (95% CI, 1.6-15.4) considering age and sexual experience, but did not differ by other potential risk factors. CONCLUSION: Our studies suggest that oral HPV is more common in individuals with Fanconi anemia. It will be essential to continue to explore associations between risk factors and immune dysfunction on HPV incidence and persistence over time. IMPACT: HPV vaccination should be emphasized in those with Fanconi anemia as a first step to prevent oropharyngeal cancers, although additional studies are needed to determine whether the level of protection it offers in this population is adequate.


Asunto(s)
Anemia de Fanconi/virología , Enfermedades de la Boca/virología , Papillomaviridae/aislamiento & purificación , Infecciones por Papillomavirus/virología , Adolescente , Adulto , Niño , Preescolar , Femenino , Neoplasias de Cabeza y Cuello/virología , Humanos , Incidencia , Lactante , Masculino , Persona de Mediana Edad , Mucosa Bucal/virología , Adulto Joven
13.
Microrna ; 2(1): 2-13, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25070710

RESUMEN

Human papillomaviruses (HPVs) are associated with the pathogenesis of a variety of human cancers, including cervical and oropharyngeal cancers. The HPV E6 and E7 oncogenes are usually expressed to high levels in these cancers. Previous studies have shown dysregulation of cellular microRNAs (miRNAs) in HPV-positive cell lines and cancer tissues and recent studies have identified a few miRNAs whose levels are altered in the presence of the viral E6 and E7 proteins. In order to identify all the cellular miRNAs whose expression may be affected by these oncoproteins, we carried out microarray analysis using human foreskin keratinocytes (HFKs) expressing either or both of these two proteins. These studies showed that 90 and 60 miRNAs were dysregulated in the presence of the E6 or the E7 protein, respectively. Of these, 43 miRNAs were similarly affected in HFK-E6 and/or HFK-E7 when compared to control cells. The joint expression of E6 and E7 proteins in HFKs caused changes in the levels of 64 miRNAs, of which 24 were similarly affected in HFK-E6 and/or HFK-E7 cells relative to controls. The microarray experiments were validated by quantitative real-time RT-PCR analysis of several differentially expressed miRNAs. Several miRNAs dysregulated by the E6 and/or E7 proteins are known to be altered in a variety of human cancers. Furthermore, previously known cellular targets of these miRNAs are involved in processes such as cell cycle regulation, apoptosis, cell-cell adhesion, cell mobility and proliferation, and alterations in their levels may contribute to HPV-associated carcinogenesis. Taken together, the results of our studies suggest that high risk HPV E6 and E7 proteins share the ability to regulate a subset of cellular miRNAs.


Asunto(s)
Papillomavirus Humano 16/metabolismo , Queratinocitos/metabolismo , MicroARNs/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Represoras/metabolismo , Células Cultivadas , Prepucio/citología , Humanos , Queratinocitos/citología , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus/genética , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , Proteínas Represoras/genética
14.
Head Neck ; 33(4): 504-12, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20652977

RESUMEN

BACKGROUND: Human papillomavirus (HPV)-positive cases of squamous cell carcinoma of the head and neck (SCCHN) have a much better disease outcome compared to SCCHN cases lacking HPV. Differences in microRNA (miRNA) expression may affect their clinical outcomes. METHODS: The miRNA expression was studied using microarrays and quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) in HPV-16-positive and HPV-negative SCCHN cell lines. The role of HPV-16 E6 and E7 oncogenes in altering miRNA expression was investigated using human foreskin keratinocytes (HFKs). RESULTS: The miRNAs miR-363, miR-33, and miR-497 were upregulated, whereas miR-155, miR-181a, miR-181b, miR-29a, miR-218, miR-222, miR-221, and miR-142-5p were downregulated in HPV-positive cells compared to both HPV-negative SCCHN and normal oral keratinocytes. HPV-16 E6 oncogene altered miRNA expression in HFKs and in an HPV-16-positive cell line with E6 knockdown using siRNA. CONCLUSION: miRNAs differentially expressed in the presence of HPV-16 may provide biomarkers for SCCHN and identify cellular pathways targeted by this virus.


Asunto(s)
Carcinoma de Células Escamosas/virología , Neoplasias de Cabeza y Cuello/virología , Papillomavirus Humano 16 , MicroARNs/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Queratinocitos/metabolismo , Infecciones por Papillomavirus/complicaciones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Cancer Res ; 69(17): 7022-9, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19706760

RESUMEN

The human papillomavirus (HPV) 16 E7 oncoprotein has been reported previously to stimulate DNA damage and to activate host cell DNA damage checkpoints. How HPV-16 E7 maintains proliferation despite activated DNA damage checkpoints is incompletely understood. Here, we provide evidence that cells expressing the HPV-16 E7 oncoprotein can enter mitosis in the presence of DNA damage. We show that this activity of HPV-16 E7 involves attenuation of DNA damage checkpoint control by accelerating the proteolytic turnover of claspin. Claspin mediates the activation of CHK1 by ATR in response to replication stress, and its degradation plays a critical role in DNA damage checkpoint recovery. Expression of a nondegradable mutant of claspin was shown to inhibit mitotic entry in HPV-16 E7-expressing cells. Multiple components of the SCF(beta-TrCP)-based claspin degradation machinery were found deregulated in the presence of HPV-16 E7, including cullin 1, beta-TrCP, Aurora A, and Polo-like kinase-1 (PLK1). In contrast, no difference in the expression level of the claspin deubiquitinating enzyme USP7 was detected. Levels of Aurora A and PLK1 as well as phosphorylated PLK1 at threonine 210, a prerequisite for DNA damage checkpoint recovery, remained detectable following replication stress in HPV-16 E7-expressing cells but not in control cells. In summary, our results suggest that the HPV-16 E7 oncoprotein alleviates DNA damage checkpoint responses and promotes mitotic entry by accelerating claspin degradation through a mechanism that involves deregulation of components of the SCF(beta-TrCP)-based claspin degradation machinery.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Daño del ADN , Papillomavirus Humano 16/genética , Mitosis/genética , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Aurora Quinasas , Proteínas de Ciclo Celular/metabolismo , Proteínas Cullin/metabolismo , Regulación Viral de la Expresión Génica , Humanos , Hidrólisis , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Quinasa Tipo Polo 1
16.
Cancer Res ; 68(23): 9954-63, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047177

RESUMEN

Expression of the high-risk human papillomavirus (HPV-16) E7 oncoprotein extends the life span of primary human keratinocytes and partially restores telomere length in the absence of telomerase. The molecular basis of this activity is incompletely understood. Here, we show that HPV-16 E7 induces an increased formation of alternative lengthening of telomeres (ALT)-associated promyelocytic leukemia bodies (APBs) in early passage primary human keratinocytes as well as HPV-negative tumor cells. This activity was found to require sequences of HPV-16 E7 involved in degradation of the retinoblastoma tumor suppressor protein as well as regions in the COOH terminus. HPV-16 E7-induced APBs contained ssDNA and several proteins that are involved in the response to DNA replication stress, most notably the Fanconi anemia D2 protein (FANCD2) as well as BRCA2 and MUS81. In line with these results, we found that FANCD2-containing APBs form in an ATR-dependent manner in HPV-16 E7-expressing cells. To directly show a role of FANCD2 in ALT, we provide evidence that knockdown of FANCD2 rapidly causes telomere dysfunction in cells that rely on ALT to maintain telomeres. Taken together, our results suggest a novel link between replication stress and recombination-based telomere maintenance that may play a role in HPV-16 E7-mediated extension of host cell life span and immortalization.


Asunto(s)
Cuerpos de Inclusión Intranucleares/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Proteínas Oncogénicas Virales/biosíntesis , Telómero/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Replicación del ADN , ADN de Neoplasias/biosíntesis , ADN de Neoplasias/genética , ADN de Cadena Simple/genética , ADN de Cadena Simple/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/biosíntesis , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Células HeLa , Humanos , Cuerpos de Inclusión Intranucleares/genética , Cuerpos de Inclusión Intranucleares/virología , Queratinocitos/patología , Queratinocitos/virología , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/virología , Proteínas E7 de Papillomavirus , ARN Interferente Pequeño/genética , Telómero/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA