Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Carcinog ; 61(1): 85-98, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34710250

RESUMEN

Poly(ADP-ribose) polymerases (PARP) act as DNA damage sensors that produce poly(ADP-ribose) (PAR) chains at double-strand breaks, facilitating the recruitment of repair factors. Cancers with homologous recombination defects are sensitive to small molecule PARP inhibitors. Despite PARP5B gene copy number changes in many cancers, the effects of this genetic alteration on tumor phenotype are largely unknown. To better understand this clinical finding, we characterized a PARP5B null mutation in a carcinogen-induced in vivo head and neck squamous cell carcinoma (SCC) model. Reduced PARP5B expression inhibited tumor growth, induced primary tumor differentiation and apoptosis, and inhibited cell proliferation and metastasis. Loss of PARP5B expression-induced ataxia telangiectasia and Rad3 related (ATR) activation and depleted the cancer stem cell fraction. PARP5B null tumor cells lacked 53BP1+ double-strand break foci, ATM activation, and p53 induction compared to PARP5B+/+ cancers. PARP5B null SCC expresses a multiprotein complex containing PML, pRPA, Rad50, Rad51, XRCC1, proliferating cell nuclear antigen (PCNA), and Mcm2, suggesting an HR-mediated repair mechanism at DNA replication foci. Low doses of etoposide combined with the PARP5B inhibitor XAV939 induced senescence and apoptosis in human SCC lines. NBS1 overexpression in these cells inhibited the effects of low-dose etoposide/XAV939 treatment. Our results indicate that PARP5B inhibition is new targeted cancer therapy.


Asunto(s)
Carcinógenos/toxicidad , Regulación hacia Abajo , Neoplasias de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Tanquirasas/genética , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Etopósido/administración & dosificación , Etopósido/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/inducido químicamente , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Mutación con Pérdida de Función , Ratones , Invasividad Neoplásica , Carcinoma de Células Escamosas de Cabeza y Cuello/inducido químicamente , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Tanquirasas/metabolismo
2.
Int J Cancer ; 144(9): 2239-2253, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30450584

RESUMEN

Mammary gland luminal cells are maintained by the proliferation of ER- luminal progenitor (LP) cells. Human breast LP cells exhibit telomere DNA damage, which is associated with mammographic density and increased cancer risk. Telomeric repeat factor 2 (TRF2) protects telomeres from DNA damage response. TRF2 expression is reduced in human breast cancers. We deleted TRF2 expression in mammary gland epithelium. Mammary glands lacking TRF2 expression exhibited increased telomere DNA damage response, histopathological and functional degeneration, and prominent ductal fibrosis. TRF2-deficient mammary tumors exhibited rapid onset and increased proliferation. Tumor derived LP cells failed to form tumors after transplantation. The MSC population was highly tumorigenic and maintained telomeres via the ALT mechanism. Telomere DNA damage response in mammary tumors resulted in p53 dependent ER+ cellular differentiation and sensitivity to anti-estrogen therapy. Our results provide a new in vivo model of mammographic density, stem cell differentiation, cancer risk, and therapeutic sensitivity.


Asunto(s)
Neoplasias de la Mama/patología , Fibrosis/patología , Glándulas Mamarias Animales/patología , Telómero/fisiología , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Daño del ADN/genética , Reparación del ADN/genética , Femenino , Humanos , Glándulas Mamarias Animales/citología , Ratones , Ratones Transgénicos , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Anticancer Res ; 44(8): 3231-3242, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39060071

RESUMEN

BACKGROUND/AIM: The most frequently altered epigenetic modifier in head and neck squamous carcinoma (HNSC) is the histone methyltransferase KMT2D. KMT2D catalyzes methylation of histone H3K4 resulting in open chromatin and the activation of target genes. Tumor-associated macrophages (TAMs) promote cancer growth by causing T lymphocyte exhaustion. C-C motif chemokine ligand 2 (CCL2) is a potent TAM chemotactic factor. In HNSC, TAMs have been associated with unfavorable patient outcomes and metastasis. The aim of this study was to determine the role of KMT2D in HNSC using genetically engineered in vivo models. MATERIALS AND METHODS: KMT2D protein expression was correlated with lymph node metastasis in human HNSC using immunohistochemistry. Genetically engineered KMT2D and CCL2 knockout models of HNSC were created in vivo. HNSC was characterized using qRT-PCR, histopathology, and immunohistochemistry/immunofluorescence microscopy. We also analyzed the effects of KMT2D expression on the proliferation and migration of human HNSC lines. The regulation of the CCL2 gene by KMT2D was characterized using chromatin immunoprecipitation-sequencing assay of transposase accessible chromatin-sequencing, and chromatin conformation capture-sequencing. RESULTS: Human HNSC cases with high KMT2D expression exhibited significantly increased lymph node metastasis. Reduced KMT2D expression in our genetically engineered model correlated with reduced lymph node metastasis, longer latency, and slow tumor growth. CCL2 expression was decreased in KMT2D deficient HNSC, which correlated with a reduced TAM gene expression signature. Genomic experiments demonstrated that KMT2D directly targeted the CCL2 gene. A new genetically engineered in vivo model of CCL2-null HNSC was created, recapitulating the KMT2D deficient phenotype and showing a decreased T lymphocyte exhaustion signature. CONCLUSION: KMT2D regulates CCL2-mediated immune response and metastasis in HNSC.


Asunto(s)
Quimiocina CCL2 , Neoplasias de Cabeza y Cuello , Humanos , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Animales , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Ratones , Línea Celular Tumoral , Metástasis Linfática , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/inmunología , Femenino , Movimiento Celular
4.
Mol Carcinog ; 52(2): 103-17, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22086874

RESUMEN

Telomerase is a ribonucleoprotein that maintains the ends of chromosomes (telomeres). In normal cells lacking telomerase activity, telomeres shorten with each cell division because of the inability to completely synthesize the lagging strand. Critically shortened telomeres elicit DNA damage responses and limit cellular division and lifespan, providing an important tumor suppressor function. Most human cancer cells express telomerase which contributes significantly to the tumor phenotype. In human breast cancer, telomerase expression is predictive of clinical outcomes such as lymph node metastasis and survival. In mouse models of mammary cancer, telomerase expression is also upregulated. Telomerase overexpression resulted in spontaneous mammary tumor development in aged female mice. Increased mammary cancer also was observed when telomerase deficient mice were crossed with p53 null mutant animals. However, the effects of telomerase and telomere length on oncogene driven mammary cancer have not been completely characterized. To address these issues we characterized neu proto-oncogene driven mammary tumor formation in G1 Terc-/- (telomerase deficient with long telomeres), G3 Terc-/- (telomerase deficient with short telomeres), and Terc+/+ mice. Telomerase deficiency reduced the number of mammary tumors and increased tumor latency regardless of telomere length. Decreased tumor formation correlated with increased apoptosis in Terc deficient tumors. Short telomeres dramatically increased lung metastasis which correlated with increased genomic instability, and specific alterations in DNA copy number and gene expression. We concluded that short telomeres promote metastasis in the absence of telomerase activity in neu oncogene driven mammary tumors.


Asunto(s)
Genes erbB-2 , Inestabilidad Genómica , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Telomerasa/metabolismo , Telómero , Animales , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proto-Oncogenes Mas , ARN/genética , ARN/metabolismo , Telomerasa/genética
5.
Oncogene ; 42(31): 2402-2414, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37393340

RESUMEN

The International Agency for Research on Cancer determined that obesity is the primary preventable cause of breast cancer. The nuclear receptor peroxisome proliferator activated receptor γ (PPARγ) binds inflammatory mediators in obesity and its expression is reduced in human breast cancer. We created a new model to better understand how the obese microenvironment alters nuclear receptor function in breast cancer. The obesity related cancer phenotype was PPARγ dependent; deletion of PPARγ in mammary epithelium which is a tumor suppressor in lean mice unexpectedly increased tumor latency, reduced the luminal progenitor (LP) tumor cell fraction, and increased autophagic and senescent cells. Loss of PPARγ expression in mammary epithelium of obese mice increased expression of 2-aminoadipate semialdehyde synthase (AASS) which regulates lysine catabolism to acetoacetate. PPARγ-associated co-repressors and activators regulated AASS expression via a canonical response element. AASS expression was significantly reduced in human breast cancer, and AASS overexpression or acetoacetate treatment inhibited proliferation and induced autophagy and senescence in human breast cancer cell lines. Genetic or pharmacologic HDAC inhibition promoted autophagy and senescence in mammary tumor cells in vitro and in vivo. We concluded that lysine metabolism is a novel metabolic tumor suppressor pathway in breast cancer.


Asunto(s)
Neoplasias de la Mama , Ratones , Humanos , Animales , Femenino , Neoplasias de la Mama/patología , PPAR gamma/genética , Lisina , Acetoacetatos , Obesidad , Microambiente Tumoral
6.
Anticancer Res ; 41(11): 5393-5403, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34732408

RESUMEN

BACKGROUND: Head and neck cancer (HNC) is common worldwide. Given poor outcomes for patients with HNC, research into targeted therapies is needed. Ataxia telangiectasia mutated (ATM) is a DNA damage kinase which is activated by double-strand DNA breaks. We tested the effects of a novel ATM inhibitor on HNC cell lines and xenografts. MATERIALS AND METHODS: p53-Binding protein 1 and phosphorylated ATM were localized in cultured cells by immunofluorescence microscopy. Protein expression was determined by western blot. Tumor xenografts were established by injecting HNC lines into immunocompromised mice. Tumor sections were characterized by immunohistochemistry. Apoptotic cells were determined by terminal transferase-mediated dUTP nick-end labeling assay. RESULTS: ATM inhibition increased double-strand DNA breaks at replication foci in HNC cell lines. ATM inhibition affected cell-cycle regulatory protein expression, blocked cell-cycle progression at the G2/M phase and resulted in apoptosis. CONCLUSION: ATM inhibition may be therapeutically useful in treating HNC.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Roturas del ADN de Doble Cadena , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Ratones Desnudos , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
BMC Cancer ; 10: 629, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21080969

RESUMEN

BACKGROUND: The peroxisome proliferator activated receptor (PPAR) subgroup of the nuclear hormone receptor superfamily is activated by a variety of natural and synthetic ligands. PPARs can heterodimerize with retinoid X receptors, which have homology to other members of the nuclear receptor superfamily. Ligand binding to PPAR/RXRs results in recruitment of transcriptional coactivator proteins such as steroid receptor coactivator 1 (SRC-1) and CREB binding protein (CBP). Both SRC-1 and CBP are histone acetyltransferases, which by modifying nucleosomal histones, produce more open chromatin structure and increase transcriptional activity. Nuclear hormone receptors can recruit limiting amounts of coactivators from other transcription factor binding sites such as AP-1, thereby inhibiting the activity of AP-1 target genes. PPAR and RXR ligands have been used in experimental breast cancer therapy. The role of coactivator expression in mammary tumorigenesis and response to drug therapy has been the subject of recent studies. METHODS: We examined the effects of loss of SRC-1 on MMTV-neu mediated mammary tumorigenesis. RESULTS: SRC-1 null mutation in mammary tumor prone mice increased the tumor latency period, reduced tumor proliferation index and metastasis, inhibited response to PPAR and RXR ligands, and induced genes involved in mammary gland differentiation. We also examined human breast cancer cell lines overexpressing SRC-1 or CBP. Coactivator overexpression increased cellular proliferation with resistance to PPAR and RXR ligands and remodeled chromatin of the proximal epidermal growth factor receptor promoter. CONCLUSIONS: These results indicate that histone acetyltransferases play key roles in mammary tumorigenesis and response to anti-proliferative therapies.


Asunto(s)
Antineoplásicos/farmacología , Genes erbB-2 , Neoplasias Mamarias Experimentales/prevención & control , Virus del Tumor Mamario del Ratón/genética , Coactivador 1 de Receptor Nuclear/deficiencia , Receptores Activados del Proliferador del Peroxisoma/agonistas , Animales , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Clofibrato/farmacología , Ácidos Grasos Insaturados/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Invasividad Neoplásica , Coactivador 1 de Receptor Nuclear/genética , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Regiones Promotoras Genéticas , Receptores X Retinoide/agonistas , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Tetrahidronaftalenos/farmacología , Tiazolidinedionas/farmacología , Factores de Tiempo , Transfección , Carga Tumoral/efectos de los fármacos
8.
BMC Cancer ; 10: 365, 2010 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-20618948

RESUMEN

BACKGROUND: Lymph node metastasis is a critical event in the progression of tongue squamous cell carcinoma (TSCC). The identification of biomarkers associated with the metastatic process would provide critical prognostic information to facilitate clinical decision making. Previous studies showed that deregulation of manganese superoxide dismutase (SOD2) expression is a frequent event in TSCC and may be associated with enhanced cell invasion. The purpose of this study is to further evaluate whether the expression level of SOD2 is correlated with the metastatic status in TSCC patients. METHODS: We first examined the SOD2 expression at mRNA level on 53 TSCC and 22 normal control samples based on pooled-analysis of existing microarray datasets. To confirm our observations, we examined the expression of SOD2 at protein level on an additional TSCC patient cohort (n = 100), as well as 31 premalignant dysplasias, 15 normal tongue mucosa, and 32 lymph node metastatic diseases by immunohistochemistry (IHC). RESULTS: The SOD2 mRNA level in primary TSCC tissue is reversely correlated with lymph node metastasis in the first TSCC patient cohort. The SOD2 protein level in primary TSCC tissue is also reversely correlated with lymph node metastasis in the second TSCC patient cohort. Deregulation of SOD2 expression is a common event in TSCC and appears to be associated with disease progression. Statistical analysis revealed that the reduced SOD2 expression in primary tumor tissue is associated with lymph node metastasis in both TSCC patient cohorts examined. CONCLUSIONS: Our study suggested that the deregulation of SOD2 in TSCC has potential predictive values for lymph node metastasis, and may serve as a therapeutic target for patients at risk of metastasis.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Superóxido Dismutasa/genética , Neoplasias de la Lengua/genética , Neoplasias de la Lengua/patología , Lengua/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/metabolismo , Estudios de Cohortes , Femenino , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Masculino , Persona de Mediana Edad , Pronóstico , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/metabolismo , Tasa de Supervivencia , Lengua/patología , Neoplasias de la Lengua/metabolismo
9.
Mol Cancer Res ; 18(9): 1326-1339, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32467172

RESUMEN

Telomere shortening has been demonstrated in benign prostatic hypertrophy (BPH), which is associated with prostate epithelial cell senescence. Telomere shortening is the most frequently observed genetic alteration in prostatic intraepithelial neoplasia, and is associated with poor clinical outcomes in prostate cancer. Gene expression database analysis revealed decreased TRF2 expression during malignant progression of the prostate gland. We reasoned that reduced TRF2 expression in prostate epithelium, by activating the telomere DNA damage response, would allow us to model both benign and malignant prostate disease. Prostate glands with reduced epithelial TRF2 expression developed age- and p53-dependent hypertrophy, senescence, ductal dilation, and smooth muscle hyperplasia similar to human BPH. Prostate tumors with reduced TRF2 expression were classified as high-grade androgen receptor-negative adenocarcinomas, which exhibited decreased latency, increased proliferation, and distant metastases. Prostate cancer stem cells with reduced TRF2 expression were highly tumorigenic and maintained telomeres both by telomerase and alternative lengthening (ALT). Telomerase inhibition in prostate glands with reduced TRF2 expression produced significant reduction in prostate tumor incidence by halting progression at intraepithelial neoplasia (PIN). These lesions were highly differentiated, exhibited low proliferation index, and high apoptotic cell fraction. Prostate tumors with reduced TRF2 expression and telomerase inhibition failed to metastasize and did not exhibit ALT. IMPLICATIONS: Our results demonstrate that the telomere DNA damage response regulates BPH, PIN, and prostate cancer and may be therapeutically manipulated to prevent prostate cancer progression.


Asunto(s)
Daño del ADN/genética , Telómero/metabolismo , Animales , Carcinogénesis , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Transducción de Señal
10.
Int J Oncol ; 34(5): 1449-53, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19360358

RESUMEN

Breast cancer is composed of heterogeneous cell populations with different biological properties. The capacity to form tumors resides in a small group of cells termed tumor initiating cells or cancer stem cells. Tumor initiating cells have been identified in a variety of cancers by sorting of subpopulations based on cell surface markers and transplantation into animal models. Tumor initiating cells have the important feature of self renewal, which is a property in common with stem cells. We examined established breast cancer lines for cells with tumor initiating properties. A dye efflux side population in MCF7 and T47D lines expressed markers of breast cancer stem cells. The side population represents a distinct morphologic and functional subpopulation within the human breast cancer cell lines MCF7 and T47D. The side population from human breast cancer cell lines was able to initiate tumors in vivo. The side population cells from human breast cancer cell lines were more resistant to ionizing radiation than the non-side population. We concluded that tumor initiating cells exist in established human breast cancer cell lines.


Asunto(s)
Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Madre Neoplásicas/patología , Animales , Proliferación Celular/efectos de la radiación , Separación Celular , Supervivencia Celular/efectos de la radiación , Femenino , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias/patología , Células Madre Neoplásicas/efectos de la radiación , Radiación Ionizante , Trasplante Heterólogo
11.
BMC Cancer ; 9: 167, 2009 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-19497117

RESUMEN

BACKGROUND: Recent proteomic studies identified Hsp27 as a highly over-expressed protein in oral squamous cell carcinoma (OSCC). Clinical studies that attempted to evaluate the prognostic values of Hsp27 yielded inconsistent results, which may be due to inclusion of OSCC cases from multiple anatomic sites. In this study, to determine the utility of Hsp27 for prognosis, we focused on oral tongue SCC (OTSCC), one of the most aggressive forms of OSCC. METHODS: Archival clinical samples of 15 normal oral tongue mucosa, 31 dysplastic lesions, 80 primary OTSCC, and 32 lymph node metastases were examined for Hsp27 expression by immunohistochemistry (IHC). Statistical analyses were carried out to assess the prognostic value of Hsp27 expression for patients with this disease. RESULTS: Dysregulation of Hsp27 expression was observed in dysplastic lesions, primary OTSCC, and lymph node metastases, and appears to be associated with disease progression. Statistical analysis revealed that the reduced Hsp27 expression in primary tumor tissue was associated with poor differentiation. Furthermore, the higher expression of Hsp27 was correlated with better overall survival. CONCLUSION: Our study confirmed that the dysregulation of Hsp27 expression is a frequent event during the progression of OTSCC. The expression of Hsp27 appears to be an independent prognostic marker for patients with this disease.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Proteínas de Choque Térmico HSP27/biosíntesis , Neoplasias de la Lengua/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Femenino , Expresión Génica , Proteínas de Choque Térmico HSP27/genética , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Neoplasias de la Lengua/genética , Neoplasias de la Lengua/patología , Adulto Joven
12.
Mol Cancer Res ; 17(7): 1480-1492, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31043491

RESUMEN

Dyskeratosis congenita is a telomere DNA damage syndrome characterized by defective telomere maintenance, bone marrow failure, and increased head and neck cancer risk. The Pot1b-/-;Terc+/- mouse exhibits some features of dyskeratosis congenita, but head and neck cancer was not reported in this model. To model the head and neck cancer phenotype, we created unique Pot1b- and p53-null-mutant models which allow genetic lineage tracing of two distinct stem cell populations. Loss of Pot1b expression depleted stem cells via ATR/Chk1/p53 signaling. Tumorigenesis was inhibited in Pot1b-/-;p53+/+ mice due to cellular senescence. Pot1b-/-;p53-/- tumors also exhibited senescence, but proliferated and metastasized with expansion of Lgr6+ stem cells indicative of senescence-associated secretory phenotype. Selective depletion of the small K15+ stem cell fraction resulted in reduction of Lgr6+ cells and inhibition of tumorigenesis via senescence. Gene expression studies revealed that K15+ cancer stem cells regulate Lgr6+ cancer stem cell expansion via chemokine signaling. Genetic ablation of the chemokine receptor Cxcr2 inhibited cancer stem cell expansion and tumorigenesis via senescence. The effects of chemokines were primarily mediated by PI3K signaling, which is a therapeutic target in head and neck cancer. IMPLICATIONS: Paracrine interactions of cancer stem cell populations impact therapeutic options and patient outcomes.


Asunto(s)
Proteínas de Unión al ADN/genética , Neoplasias de Cabeza y Cuello/genética , Receptores Acoplados a Proteínas G/genética , Proteína p53 Supresora de Tumor/genética , Animales , Carcinogénesis/genética , Linaje de la Célula/genética , Proliferación Celular/genética , Senescencia Celular/genética , Daño del ADN/genética , Disqueratosis Congénita/complicaciones , Disqueratosis Congénita/genética , Disqueratosis Congénita/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/complicaciones , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Comunicación Paracrina/genética , ARN/genética , Receptores de Interleucina-8B/genética , Telomerasa/genética , Telómero/genética , Homeostasis del Telómero/genética
13.
Mol Cancer Res ; 5(4): 351-62, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17426250

RESUMEN

Squamous cell carcinoma of the head and neck (HNSCC) is the sixth most frequent cancer worldwide. Because HNSCC is largely acquired by environmental carcinogen exposure rather than through germ line mutations, there are no known familial forms of the disease in humans nor are there inbred rodent strains prone to spontaneous head and neck tumors. Transgenic animals with inactivation of tumor suppressor genes commonly mutated in human cases of HNSCC provide attractive models for studying the pathogenesis of head and neck cancer. p53 is the most frequently inactivated tumor suppressor gene in HNSCC. We used a chemical induction protocol in mice heterozygous for the p53 gene to evaluate how p53 inactivation contributed to head and neck carcinogenesis the mouse model. Metastatic squamous cell carcinomas developed in 100% of animals. Histopathologically, the tumors ranged from well to poorly differentiated and showed many molecular features of human HNSCC. Mice carrying only one p53 allele developed tumors with significantly reduced latency compared with wild-type controls (average, 18 versus 22 weeks). Metastatic cancer cells showed complete loss of p53 expression when compared with primary tumors. Transcriptional profiling showed not only distinct genetic differences between primary and metastatic tumors, but also when cancers from heterozygous null and wild-type animals were compared. Our results provide novel insights into the molecular genetics of tumor progression in head and neck cancer.


Asunto(s)
Carcinoma de Células Escamosas/patología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína p53 Supresora de Tumor/metabolismo , Animales , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/metabolismo , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Neoplasias de Cabeza y Cuello/inducido químicamente , Neoplasias de Cabeza y Cuello/etiología , Neoplasias de Cabeza y Cuello/metabolismo , Ganglios Linfáticos/patología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Estadificación de Neoplasias , Fenotipo , Transcripción Genética , Proteína p53 Supresora de Tumor/genética
14.
Cancer Genomics Proteomics ; 5(2): 85-94, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18460737

RESUMEN

Metastasis is a critical event in oral squamous cell carcinoma (OSCC) progression. To identify proteomic biomarkers for OSCC metastasis, 3 paired OSCC cell lines (UM1/UM2, 1386Tu/1386Ln, 686Tu/686Ln) with different metastatic potential were examined. Among those 3 cell lines, UM1, 1386Ln and 686Ln exhibited a higher degree of metastatic potential than their paired cell lines UM2, 1386Tu and 686Tu, respectively, as measured using an in vitro cell invasion assay. A total of 40 differentially expressed proteins were identified using 2D-PAGE/MS proteomic approach. Selected protein candidates (superoxide dismutase 2 and heat shock protein 27) were further investigated by immuno-histochemistry (IHC) method using independent OSCC patient tissue samples. The statistically significantly increases in IHC staining for manganese superoxide dismutase 2 (SOD2) were observed in lymph node metastatic disease when compared with the paired primary OSCC. Our results thus indicated that elevated SOD2 levels is associated with lymph node metastasis in OSCC and may provide predictive values for diagnosis of metastasis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/enzimología , Neoplasias de la Boca/enzimología , Proteómica , Superóxido Dismutasa/metabolismo , Adulto , Anciano , Western Blotting , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Factores Quimiotácticos/farmacología , Electroforesis en Gel Bidimensional , Femenino , Proteínas de Choque Térmico HSP27 , Proteínas de Choque Térmico/metabolismo , Humanos , Inmunohistoquímica , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Chaperonas Moleculares , Neoplasias de la Boca/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Superóxido Dismutasa/química
15.
J Endocrinol ; 193(1): 147-55, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17400812

RESUMEN

Steroid hormones such as 17beta-estradiol (E2) are critical to diverse cellular processes including tumorigenesis. A number of cofactors such as nuclear receptor corepressor (NCoR), CREB-binding protein (CBP), and steroid receptor coactivator 1 (SRC-1) interact with estrogen receptors (ERs) to regulate transcriptional repression or activation of target genes. Estrogen signaling in non-reproductive tract tissues such as skin is less well characterized and the effectiveness of anti-estrogen therapy for cancer arising from these tissues is unknown. We show that tamoxifen (TAM) treatment inhibited cell cycle progression and proliferation of human cancer lines derived from stratified squamous epithelium squamous cell carcinoma (SCC). E2 had no effect on proliferation of these lines despite low levels of ERalpha expression. The E2 treatment promoted displacement of the NCoR from ERalpha and recruitment of CBP to the receptor. SRC-1 expression was not detected in these SCC lines; however, transient transfection of SRC-1, CBP, or both coactivators enhanced transactivation of an estrogen responsive promoter in cancer cells treated with E2 or TAM. In stable clones expressing SRC-1, the coactivator was recruited to ERalpha along with CBP in E2 but not in TAM-treated cells. SRC-1 expression restored the E2-mediated proliferative response to human SCC lines. This increased proliferation correlated with increased extracellular signal regulated kinase 1 (ERK1) expression. SRC-1 and CBP were recruited to the proximal ERK1 promoter region in E2 but not in TAM-treated cells. We concluded that SRC-1 was a key molecular determinant of estrogen-mediated proliferation in human SCC lines.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Histona Acetiltransferasas/metabolismo , Tamoxifeno/farmacología , Factores de Transcripción/metabolismo , Western Blotting/métodos , Neoplasias de la Mama , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Histona Acetiltransferasas/genética , Humanos , Lípidos/administración & dosificación , Lípidos/genética , Coactivador 1 de Receptor Nuclear , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Transfección/métodos
16.
Int J Oncol ; 30(5): 1279-87, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17390032

RESUMEN

Reports have suggested that spindle cell carcinoma of the head and neck occurs following radiation therapy of incompletely resected SCC, representing anaplastic progression of the primary tumor. Examination of differences between spindle cell carcinoma and SCC may provide important information about anaplastic progression, clinical behavior, and response to therapy. We created a mouse model that developed spindle cell carcinoma. Spindle cell carcinoma was characterized by marked downregulation of epithelial differentiation markers and cell adhesion genes. Expression of growth factors and receptors important for epithelial proliferation was inhibited while those which regulate fibroblast and mesenchymal cell proliferation were increased. By far the largest class of upregulated genes in spindle cell carcinomas was chemokine receptors and ligands which are involved in tumor cell invasion and metastasis. These changes in gene expression clearly show loss of epithelial characteristics, acquisition of mesenchymal phenotypes, and increased propensity for invasion and metastasis by spindle cell carcinomas.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/patología , Sarcoma/patología , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Invasividad Neoplásica
17.
Oncotarget ; 8(46): 80139-80155, 2017 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-29113290

RESUMEN

Chromosome ends are protected by telomeres that prevent DNA damage response and degradation. When telomeres become critically short, the DNA damage response is activated at chromosome ends which induces cellular senescence or apoptosis. Telomeres are protected by the double stranded DNA binding protein TRF2 and maintained by telomerase or a recombination based mechanism known as alternative lengthening of telomeres (ALT). Telomerase is expressed in the basal layer of the epidermis, and stem cells in epidermis have longer telomeres than proliferating populations. Stem cell expansion has been associated with epithelial-mesenchymal transition (EMT) in cancer. EMT is a critical process in cancer progression in which cells acquire spindle morphology, migrate from the primary tumor, and spread to distant anatomic sites. Our previous study demonstrated that loss of TRF2 expression observed in human squamous cell carcinomas expanded metastatic cancer stem cells during mouse skin carcinogenesis. To determine if telomerase inhibition could block the TRF2-null mediated expansion of metastatic clones, we characterized skin carcinogenesis in a conditional TRF2/Terc double null mutant mouse. Loss of TRF2 and Terc expression resulted in telomere DNA damage, severely depleted CD34 + and Lgr6+ cancer stem cells, and induced terminal differentiation of metastatic cancer cells. However a novel cancer stem cell population evolved in primary tumors exhibiting genomic instability, ALT, and EMT. Surprisingly we discovered that metastatic clones evolved prior to histopathologic onset of primary tumors. These results have important implications for understanding the evolution and treatment of metastatic cancer.

18.
Breast Cancer Res ; 8(1): R1, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16417649

RESUMEN

INTRODUCTION: The breast cancer susceptibility gene BRCA1 is involved in the repair of double-strand breaks induced by ionizing radiation and chemotherapy drugs. BRCA1 interacts with coactivators such as p300 and CREB-binding protein (CBP) to activate target gene transcription. Estrogen and retinoic acid receptors (ER and RAR) also require coactivator proteins for their ligand-dependent functions. Few studies have suggested a role for nuclear hormone receptors in DNA repair. METHODS: DNA damage and repair activity were quantified with the use of single-cell gel electrophoresis and plasmid end-joining assays. Cell cycle progression and apoptosis were determined by bromodeoxyuridine and TdT-mediated dUTP nick end labelling assays. Stable transfection was accomplished with the lipofection procedure. Protein interaction and expression were determined by immunoprecipitation and western blotting. RESULTS: 17beta-estradiol (E2) and all-trans retinoic acid (RA) had opposing effects on DNA damage and breast cancer cell survival after double-strand break damage. Treatment with E2, but not with RA, resulted in complex formation between ERalpha, CBP, and BRCA1 in ER-positive cell lines. Mutant BRCA1 reduced the expression and activity of DNA damage repair proteins but did not block nuclear hormone-dependent effects. Mutant BRCA1 failed to form complexes with ERalpha and CBP, which correlated with its ability to exert E2-independent effects on DNA repair. Mutant BRCA1 inhibited cell cycle progression and produced increased survival in cells with double-strand breaks. Ectopic ERalpha expression reproduced the E2-mediated effects on DNA damage, repair, and survival. CONCLUSION: The present study proposes a new mechanism by which ER and RAR regulate BRCA1-mediated DNA repair by means of CBP.


Asunto(s)
Neoplasias de la Mama/genética , Proteína de Unión a CREB/fisiología , Reparación del ADN , Receptor alfa de Estrógeno/fisiología , Genes BRCA1 , Antineoplásicos/farmacología , Western Blotting , Neoplasias de la Mama/patología , Supervivencia Celular , Daño del ADN , Estradiol/fisiología , Femenino , Humanos , Inmunoprecipitación , Tretinoina/farmacología , Células Tumorales Cultivadas
19.
Biochem J ; 392(Pt 3): 589-99, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16050810

RESUMEN

RARs (retinoic acid receptors) mediate the effect of their ligand RA (retinoic acid) on gene expression. We previously showed that RA inhibited cellular proliferation in part by decreasing expression of the mitogen activated protein kinase ERK1 (extracellular signal regulated kinase 1). However, the mechanism by which RA regulates ERK1 expression is largely uncharacterized. The present study characterizes coactivator-mediated regulation of RA target gene expression by analysing ERK1 promoter activation. CBP (CREB-binding protein) and PCAF (p300/CBP associated factor) are transcriptional coactivators that interact with nuclear hormone receptors such as RARs. CBP and PCAF differentially regulated ERK1 expression in stable clones. CBP clones expressed higher ERK1 protein levels, proliferated faster in culture and were resistant to RA-mediated growth inhibition. PCAF clones expressed lower levels of ERK1 protein and cells grew more slowly than controls. CBP and PCAF regulation of the ERK1 promoter was dependent on two Sp1 (specificity protein 1) sites located between -86 and -115 bp. Immunoprecipitation and yeast two-hybrid analysis revealed that PCAF interacted with Sp1 via CBP. A putative p53 binding site at -360 bp functioned as a major repressor of ERK1 promoter activity even in the absence of exogenous p53 expression. CBP and PCAF occupancy of the proximal ERK1 promoter was dramatically decreased by RA treatment. PCAF mediated inhibition of ERK1 expression was due to decreased stability of the kinase mRNA. We conclude that CBP and PCAF coactivators mediate ERK1 gene expression at both the transcriptional and post-transcriptional level.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Humanos , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tretinoina/metabolismo , Factores de Transcripción p300-CBP
20.
Mol Cancer Res ; 1(7): 532-40, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12754300

RESUMEN

The chemotherapeutic agent retinoic acid (RA) and its derivatives have been used to treat many tumor types. The antitumor effects of retinoids are in part due to their ability to inhibit proliferation of cancer cells. However, smokers receiving dietary vitamin A and beta carotene in chemoprevention studies had a higher incidence of lung cancer. These studies imply that lower doses of retinoids may have tumor-promoting activity. The effects of RA are mediated by a family of ligand-dependent transcription factors, the retinoic acid receptors (RARs) and the retinoid X receptors (RXR). We examined the effects of low- and high-dose RA treatment on proliferation of human squamous cell carcinoma lines in vitro. Low concentrations of RA (20 nM) increased proliferation of SCC lines by epidermal growth factor (EGF) activation of the mitogen-activated protein kinase ERK1. These changes were accompanied by increased expression of S- and G(2) phase cyclins and cyclin-dependent kinases (cdk), increased Rb phosphorylation, and increased E2F-1 DNA binding activity. In contrast, higher doses of RA (40 nM to 1 micro M) inhibited ERK1 expression, caused accumulation of G(1) phase cyclins and cdks, decreased Rb phosphorylation, and increased Rb/E2F-1 association. Overexpression of ERK1 or dominant negative ERK1 was sufficient to reproduce the effects of low- and high-dose RA, respectively. Treatment with receptor selective retinoids revealed that both RARalpha and RARgamma mediated the effects of RA on SCC lines. We concluded that low-dose RA induced proliferation by increased EGF signaling while higher concentrations inhibited cell division by decreasing ERK1 activation.


Asunto(s)
Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Tretinoina/farmacología , Carcinoma de Células Escamosas , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA