Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-38612901

RESUMEN

We explore the possibility that defects in genes associated with the response and repair of DNA double strand breaks predispose oral potentially malignant disorders (OPMD) to undergo malignant transformation to oral squamous cell carcinoma (OSCC). Defects in the homologous recombination/Fanconi anemia (HR/FA), but not in the non-homologous end joining, causes the DNA repair pathway to appear to be consistent with features of familial conditions that are predisposed to OSCC (FA, Bloom's syndrome, Ataxia Telangiectasia); this is true for OSCC that occurs in young patients, sometimes with little/no exposure to classical risk factors. Even in Dyskeratosis Congenita, a disorder of the telomerase complex that is also predisposed to OSCC, attempts at maintaining telomere length involve a pathway with shared HR genes. Defects in the HR/FA pathway therefore appear to be pivotal in conditions that are predisposed to OSCC. There is also some evidence that abnormalities in the HR/FA pathway are associated with malignant transformation of sporadic cases OPMD and OSCC. We provide data showing overexpression of HR/FA genes in a cell-cycle-dependent manner in a series of OPMD-derived immortal keratinocyte cell lines compared to their mortal counterparts. The observations in this study argue strongly for an important role of the HA/FA DNA repair pathway in the development of OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Anemia de Fanconi , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Neoplasias de la Boca/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , ADN
2.
Biology (Basel) ; 12(1)2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36671795

RESUMEN

An escape from cellular senescence through the development of unlimited growth potential is one of the hallmarks of cancer, which is thought to be an early event in carcinogenesis. In this review, we propose that the molecular effectors of senescence, particularly the inactivation of TP53 and CDKN2A, together with telomere attrition and telomerase activation, all lead to aneuploidy in the keratinocytes from oral potentially malignant disorders (OPMD). Premalignant keratinocytes, therefore, not only become immortal but also develop genotypic and phenotypic cellular diversity. As a result of these changes, certain clonal cell populations likely gain the capacity to invade the underlying connective tissue. We review the clinical implications of these changes and highlight a new PCR-based assay to identify aneuploid cell in fluids such as saliva, a technique that is extremely sensitive and could facilitate the regular monitoring of OPMD without the need for surgical biopsies and may avoid potential biopsy sampling errors. We also draw attention to recent studies designed to eliminate aneuploid tumour cell populations that, potentially, is a new therapeutic approach to prevent malignant transformations in OPMD.

4.
Cancer Metastasis Rev ; 40(4): 1073-1091, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34932167

RESUMEN

It is well established that cancer cells acquire energy via the Warburg effect and oxidative phosphorylation. Citrate is considered to play a crucial role in cancer metabolism by virtue of its production in the reverse Krebs cycle from glutamine. Here, we review the evidence that extracellular citrate is one of the key metabolites of the metabolic pathways present in cancer cells. We review the different mechanisms by which pathways involved in keeping redox balance respond to the need of intracellular citrate synthesis under different extracellular metabolic conditions. In this context, we further discuss the hypothesis that extracellular citrate plays a role in switching between oxidative phosphorylation and the Warburg effect while citrate uptake enhances metastatic activities and therapy resistance. We also present the possibility that organs rich in citrate such as the liver, brain and bones might form a perfect niche for the secondary tumour growth and improve survival of colonising cancer cells. Consistently, metabolic support provided by cancer-associated and senescent cells is also discussed. Finally, we highlight evidence on the role of citrate on immune cells and its potential to modulate the biological functions of pro- and anti-tumour immune cells in the tumour microenvironment. Collectively, we review intriguing evidence supporting the potential role of extracellular citrate in the regulation of the overall cancer metabolism and metastatic activity.


Asunto(s)
Ácido Cítrico , Neoplasias , Citratos , Ácido Cítrico/metabolismo , Ciclo del Ácido Cítrico , Humanos , Neoplasias/metabolismo , Fosforilación Oxidativa , Microambiente Tumoral/fisiología
5.
Sci Rep ; 11(1): 584, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436723

RESUMEN

Many of the characteristics ascribed to cancer-associated fibroblasts (CAFs) are shared by activated, autophagic and senescent fibroblasts. Whilst most oral squamous cell carcinomas (OSCCs) are genetically unstable (GU-OSCC), genetically stable variants (GS-OSCC) have been described and, notably, CAF activation (myofibroblast differentiation) and senescence are characteristics particularly associated with GU-OSCCs. However, it is not known whether autophagy is disrupted in these cells or whether autophagy regulates the development of the myofibroblast and senescent phenotypes. In this study, we show that senescent CAFs from GU-OSCCs contained more autophagosomes than normal human oral fibroblasts (NHOFs) and CAFs from GS-OSCCs possibly due to autophagic impairment. Further, we show that deregulation of autophagy in normal fibroblasts, either by inhibition with autophagy inhibitor, SAR405, or activation with TGF-ß1, induced fibroblast activation and senescence: In response to TGF-ß1, autophagy was induced prior to the development of the activated and senescent phenotypes. Lastly, we show that both SAR405- and TGF-ß1-treated NHOFs enhance OSCC cell migration but only TGF-ß1-treated cells increase OSCC invasion through Matrigel, indicating that TGF-ß1 has additional effects that are independent of fibroblast activation/senescence. These results suggest a functional role for autophagy in the development of myofibroblast and CAF phenotypes.


Asunto(s)
Autofagia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Senescencia Celular/genética , Fibroblastos/patología , Fibroblastos/fisiología , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Factor de Crecimiento Transformador beta1/fisiología , Autofagia/efectos de los fármacos , Autofagia/genética , Autofagia/fisiología , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Senescencia Celular/efectos de los fármacos , Humanos , Miofibroblastos/patología , Invasividad Neoplásica/genética , Piridinas/farmacología , Pirimidinonas/farmacología
6.
Front Cell Dev Biol ; 8: 602476, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33425906

RESUMEN

Cancer cells need excess energy and essential nutrients/metabolites not only to divide and proliferate but also to migrate and invade distant organs for metastasis. Fatty acid and cholesterol synthesis, considered a hallmark of cancer for anabolism and membrane biogenesis, requires citrate. We review here potential pathways in which citrate is synthesized and/or supplied to cancer cells and the impact of extracellular citrate on cancer cell metabolism and growth. Cancer cells employ different mechanisms to support mitochondrial activity and citrate synthesis when some of the necessary substrates are missing in the extracellular space. We also discuss the different transport mechanisms available for the entry of extracellular citrate into cancer cells and how citrate as a master metabolite enhances ATP production and fuels anabolic pathways. The available literature suggests that cancer cells show an increased metabolic flexibility with which they tackle changing environmental conditions, a phenomenon crucial for cancer cell proliferation and metastasis.

7.
Cancers (Basel) ; 11(11)2019 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-31717573

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and includes squamous cell carcinomas of the oropharynx and oral cavity. Patient prognosis has remained poor for decades and molecular targeted therapies are not in routine use. Here we showed that the overall expression of collagen subunit genes was higher in cancer-associated fibroblasts (CAFs) than normal fibroblasts. Focusing on collagen8A1 and collagen11A1, we showed that collagen is produced by both CAFs and tumour cells, indicating that HNSCCs are collagen-rich environments. We then focused on discoidin domain receptor 1 (DDR1), a collagen-activated receptor tyrosine kinase, and showed that it is over-expressed in HNSCC tissues. Further, we demonstrated that collagen promoted the proliferation and migration of HNSCC cells and attenuated the apoptotic response to cisplatin. Knockdown of DDR1 in HNSCC cells demonstrated that these tumour-promoting effects of collagen are mediated by DDR1. Our data suggest that specific inhibitors of DDR1 might provide novel therapeutic opportunities to treat HNSCC.

8.
Sci Rep ; 8(1): 12148, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30108296

RESUMEN

Cellular senescence is often associated with irreparable DNA double strand breaks (IrrDSBs) which accumulate with chronological age (IrrDSBsen). The removal of senescent cells ameliorates several age-related diseases in mice but the translation of these findings into a clinical setting would be aided by the characterisation of non-invasive biomarkers of senescent cells. Several serum metabolites are independent indicators of chronological age and some of these accumulate outside senescent fibroblasts independently of cell cycle arrest, repairable DNA breaks and cell size (the extracellular senescence metabolome, or ESM). The post-mitotic phase of senescence is dynamic, making the detection of senescent cells in vivo difficult. An unbiased metabolomic screen of the IrrDSBsen fibroblast ESM also showed differences in the times of initiation and maintenance of different metabolites but generally the ESM altered progressively over the 20 day study period unlike the reported transcriptional profiles. This more detailed analysis of IrrDSBsen identified several new ESM metabolites that are associated with chronological ageing. Targeted analysis of citrate confirmed the dynamic nature of this metabolite in two cell lines and revealed its independence from the senescence effector p16INK4A. These data will aid our understanding of metabolic signatures of ageing and their relationship to cellular senescence and IrrDSBs.


Asunto(s)
Envejecimiento/sangre , Senescencia Celular/fisiología , Fibroblastos/metabolismo , Metaboloma/fisiología , Envejecimiento/metabolismo , Células Cultivadas , Ácido Cítrico/sangre , Ácido Cítrico/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN/fisiología , Humanos , Metabolómica
9.
Sci Rep ; 6: 38489, 2016 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-27924925

RESUMEN

Cellular senescence occurs by proliferative exhaustion (PEsen) or following multiple cellular stresses but had not previously been subject to detailed metabolomic analysis. Therefore, we compared PEsen fibroblasts with proliferating and transiently growth arrested controls using a combination of different mass spectroscopy techniques. PEsen cells showed many specific alterations in both the NAD+ de novo and salvage pathways including striking accumulations of nicotinamide mononucleotide (NMN) and nicotinamide riboside (NR) in the amidated salvage pathway despite no increase in nicotinamide phosphoribosyl transferase or in the NR transport protein, CD73. Extracellular nicotinate was depleted and metabolites of the deamidated salvage pathway were reduced but intracellular NAD+ and nicotinamide were nevertheless maintained. However, sirtuin 1 was downregulated and so the accumulation of NMN and NR was best explained by reduced flux through the amidated arm of the NAD+ salvage pathway due to reduced sirtuin activity. PEsen cells also showed evidence of increased redox homeostasis and upregulated pathways used to generate energy and cellular membranes; these included nucleotide catabolism, membrane lipid breakdown and increased creatine metabolism. Thus PEsen cells upregulate several different pathways to sustain their survival which may serve as pharmacological targets for the elimination of senescent cells in age-related disease.


Asunto(s)
Senescencia Celular , Fibroblastos/citología , Fibroblastos/metabolismo , Espacio Intracelular/metabolismo , Metaboloma , Metabolómica , NAD/metabolismo , Niacinamida/metabolismo , Ciclo Celular , Proliferación Celular , Células Cultivadas , Análisis por Conglomerados , Homeostasis , Humanos , Modelos Biológicos , Oxidación-Reducción , Análisis de Componente Principal , Triptófano/metabolismo
10.
PLoS One ; 10(7): e0133745, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26196672

RESUMEN

BACKGROUND: Limited options for the treatment of cartilage damage have driven the development of tissue engineered or cell therapy alternatives reliant on ex vivo cell expansion. The study of chondrogenesis in primary cells is difficult due to progressive cellular aging and senescence. Immortalisation via the reintroduction of the catalytic component of telomerase, hTERT, could allow repeated, longitudinal studies to be performed while bypassing senescent phenotypes. METHODS: Three human cell types: bone marrow-derived stromal cells (BMA13), embryonic stem cell-derived (1C6) and chondrocytes (OK3) were transduced with hTERT (BMA13H, 1C6H and OK3H) and proliferation, surface marker expression and tri-lineage differentiation capacity determined. The sulphated glycosaminoglycan (sGAG) content of the monolayer and spent media was quantified in maintenance media (MM) and pro-chondrogenic media (PChM) and normalised to DNA. RESULTS: hTERT expression was confirmed in transduced cells with proliferation enhancement in 1C6H and OK3H cells but not BMA13H. All cells were negative for leukocyte markers (CD19, CD34, CD45) and CD73 positive. CD14 was expressed at low levels on OK3 and OK3H and HLA-DR on BMA13 (84.8%). CD90 was high for BMA13 (84.9%) and OK3 (97.3%) and moderate for 1C6 (56.7%), expression was reduced in BMA13H (33.7%) and 1C6H (1.6%). CD105 levels varied (BMA13 87.7%, 1C6 8.2%, OK3 43.3%) and underwent reduction in OK3H (25.1%). 1C6 and BMA13 demonstrated osteogenic and adipogenic differentiation but mineralised matrix and lipid accumulation appeared reduced post hTERT transduction. Chondrogenic differentiation resulted in increased monolayer-associated sGAG in all primary cells and 1C6H (p<0.001), and BMA13H (p<0.05). In contrast OK3H demonstrated reduced monolayer-associated sGAG in PChM (p<0.001). Media-associated sGAG accounted for ≥55% (PChM-1C6) and ≥74% (MM-1C6H). CONCLUSION: In conclusion, hTERT transduction could, but did not always, prevent senescence and cell phenotype, including differentiation potential, was affected in a variable manner. As such, these cells are not a direct substitute for primary cells in cartilage regeneration research.


Asunto(s)
Diferenciación Celular , Condrocitos/metabolismo , Glicosaminoglicanos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Fenotipo , Telomerasa/metabolismo , Línea Celular , Proliferación Celular , Condrocitos/citología , Condrocitos/inmunología , Condrogénesis , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Telomerasa/genética
11.
Oral Oncol ; 46(12): 840-53, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21050803

RESUMEN

Senescence of somatic cells in vitro can occur through the gradual erosion of the chromosomal telomeres following multiple rounds of cell division, or more acutely following cellular stresses connected with oncogene activation, tumour suppressor loss, ageing and migration. These various forms of senescence are associated with the activation of DNA damage checkpoints, the over-expression of p16(INK4A) and the secretion of cytokines, all of which are detected in pre-malignant lesions but muted upon malignant conversion. The various senescence signals are integrated by p16(INK4A) and p53 to produce the permanent cell cycle arrest associated with senescence. Both pRB/p16(INK4A) and p53 are dysfunctional in many cancers, including the most common type of oral cancer, squamous cell carcinoma (OSCC) and other evidence is accumulating in support of the idea that senescence acts as a barrier to tumour development and/or progression. However, senescence of the non-epithelial component of developing human tumours has been shown to enhance growth and invasion of the pre-malignant epithelial component and so senescence may well enhance cancer as well as suppress it depending on the context.


Asunto(s)
Carcinoma de Células Escamosas/patología , Senescencia Celular/fisiología , Genes p16/fisiología , Neoplasias de la Boca/patología , Proteína p53 Supresora de Tumor/fisiología , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Senescencia Celular/genética , Femenino , Humanos , Masculino , Neoplasias de la Boca/genética , Proteína p53 Supresora de Tumor/genética
12.
Exp Cell Res ; 314(13): 2434-47, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18589416

RESUMEN

The uncapping of telomeres has been shown to precipitate senescence in normal human fibroblasts and apoptosis in lymphocytes and p53-competent cancer cell lines. However, the effects of telomere uncapping on normal epithelial cells have not previously been examined. We have used the well characterised telomere repeat binding factor 2 (TRF2) dominant-negative mutant, TRF2(DeltaBDeltaM), to deplete Normal Human Epidermal Keratinocytes (NHEK) telomeres of TRF2. We observed only a two fold increase in both phosphorylation of p53 at serine 15 and 53BP1 DNA damage foci and no detectable increase in p21(WAF). Despite the weak DNA damage response, the keratinocytes growth arrest, demonstrate reduced colony formation and senescence. The small, abortive senescent colonies did not incorporate Brd-U within 48 h and expressed senescence-associated beta galactosidase (SA-beta-gal). Transcriptional profiling of TRF2-depleted keratinocytes showed a reproducible up-regulation of several genes. These included histones, genes associated with DNA damage and keratinocyte terminal differentiation. Several of the same genes were also shown to be up-regulated when keratinocytes undergo natural telomere-mediated senescence and down-regulated by ectopic telomerase expression. This study has thus revealed highly sensitive and specific candidate indicators of telomere dysfunction that may find use in identifying telomere-mediated keratinocyte senescence in ageing, cancer and other diseases.


Asunto(s)
Senescencia Celular/genética , Queratinocitos/metabolismo , Telómero/fisiología , Transcripción Genética , Células 3T3 , Animales , Proliferación Celular , Células Cultivadas , Células Clonales , Daño del ADN/genética , Perfilación de la Expresión Génica , Humanos , Recién Nacido , Queratinocitos/patología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Telómero/patología , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Transgenes
13.
J Oral Pathol Med ; 37(4): 211-20, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18205743

RESUMEN

Oral submucous fibrosis (OSF) is characterized by abnormal collagen metabolism in the submucosal connective tissue. Its influence on the overlying epithelium is not known but about 14% of OSF cases undergo malignant transformation to squamous cell carcinoma indicating association with abnormality of the epithelium. Here, we have defined the keratin expression profile, by immunohistochemistry and quantitative image analysis, using a panel of 22 anti-keratin monoclonal antibodies on 28 OSF samples. We observed an increase of K1 and K10 in the suprabasal layers, induction of K6 in the basal layer and complete loss of K19 in the epithelium. Furthermore, there was increased K17 expression in the suprabasal layers, which correlated with disease severity. In a subset of the most severe OSF cases (14%), K17 expression was completely lost in the basal layer which might define them to be at most risk to undergo malignant transformation. There was no detectable expression of K8, K18, K7 and K9 and the expression of K4, K13, K14, K15 and K16 did not change in OSF. We propose that the altered keratin profiles could be useful as histological diagnostic markers and provide important insights into the pathogenesis of the disease and its predisposition to malignancy.


Asunto(s)
Queratinocitos/patología , Queratinas/biosíntesis , Fibrosis de la Submucosa Bucal/patología , Biomarcadores , Carcinoma de Células Escamosas , Estudios de Casos y Controles , Transformación Celular Neoplásica , Expresión Génica , Humanos , Inmunohistoquímica , Queratina-17/biosíntesis , Queratinocitos/química , Queratinas/genética , Neoplasias de la Boca/química , Neoplasias de la Boca/patología , Fenotipo , Fotografía Dental , Lesiones Precancerosas/química , Lesiones Precancerosas/patología , Índice de Severidad de la Enfermedad
14.
Oral Oncol ; 44(3): 261-9, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17475542

RESUMEN

Evidence from telomerase-deficient mice strongly suggests that dysfunctional short telomeres affect cellular radio-sensitivity but this idea has yet to be extensively tested in relevant human cancer types such as oral squamous cell carcinomas (OSCCs), which are frequently treated by radiotherapy. The OSCC line BICR7 has low levels of telomerase activity, short telomeres and high levels of telomere dysfunction (judged by a high level of anaphase bridges); whereas the BICR6 line has high levels of telomerase and is more radio-resistant. Ectopic expression of the human TElomerase Reverse Transcriptase (hTERT) reduced telomere dysfunction and increased radio-resistance in BICR7 cells, but not BICR6. Furthermore, the radio-resistance of GM847 cells, which use telomerase-independent mechanisms of telomere maintenance, and of telomerase-negative normal human fibroblasts with long telomeres are similarly unaffected by ectopic expression of telomerase. We tested whether telomere function, as measured by the Anaphase Bridge Index (ABI), was found to be a useful predictor of radio-resistance in a panel of OSCC lines. Using inverse regression analysis, we found a strong inverse relationship between the ABI and radio-resistance (P<0.001), as measured by the Surviving Fraction at 4Gy (SF4). These results suggest that telomerase inhibitors could sensitise a subset of oral SCCs with short telomeres to radiotherapy and for the first time demonstrate that the tumour ABI may assist the selection of cancers that would be suitable for such sensitisation therapy.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias de la Boca/patología , Tolerancia a Radiación , Telómero/ultraestructura , Anafase , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/radioterapia , Línea Celular Tumoral , Supervivencia Celular , Senescencia Celular , Rayos gamma , Humanos , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/radioterapia , Análisis de Regresión , Estadísticas no Paramétricas , Telomerasa/metabolismo , Insuficiencia del Tratamiento
15.
BioDrugs ; 21(6): 375-85, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18020621

RESUMEN

Normal human somatic cells undergo telomeric attrition causing replicative senescence. Most immortal cancer cells cope with this by upregulating the active form of telomerase. Long-term inhibition of telomerase results in telomeric attrition and highly specific killing of cancer cells, in which the maintenance of telomere length is reliant on telomerase activity. Unfortunately, telomere erosion requires many cell divisions, possibly opening the way for acquired drug resistance. Recent attempts to solve this problem include the development of drugs that are more potent catalytic inhibitors, deny telomerase access to the telomere in situ, or affect telomere structure; some of these drugs have entered clinical trials. Combinations of these approaches may ultimately produce the best clinical results. This article reviews the latest results in both basic and applied telomere research that indicate the most promising avenues for future anticancer drug development in this area.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Telomerasa/antagonistas & inhibidores , Telómero/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , ARN/antagonistas & inhibidores , ARN/genética , ARN/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética
16.
Int J Oncol ; 28(5): 1279-85, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16596245

RESUMEN

This study examined Smad2- and Smad3-dependent transcription in 12 human head and neck squamous cell carcinoma (HNSCC) cell lines following treatment with transforming growth factor-beta1 (TGF-beta1). A markedly elevated level of TGF-beta1-induced Smad3 signalling was observed in one cell line (H357), whilst four cell lines (BICR31, H314, BICR56, BICR19) demonstrated absence of Smad3-dependent transcription that correlated with loss of TGF-beta1 growth inhibition; TGF-beta1-induced Smad2-dependent transcription was retained in two of these cell lines (H314, BICR31). Using transient expression of TGF-beta signalling components and a Smad3-dependent reporter assay, we show that BICR31 and H314 had defects of Smad4, BICR56 had abnormal TbetaR-II and BICR19 overexpressed Smad7. The results demonstrate that deregulated TGF-beta1-induced Smad signalling is common in HNSCC cell lines and can occur as a result of a variety of defects in the TGF-beta signal transduction pathway.


Asunto(s)
Neoplasias de Cabeza y Cuello/fisiopatología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/fisiología , Factor de Crecimiento Transformador beta/farmacología , Línea Celular Tumoral , Humanos , Fosforilación , Plásmidos , Proteínas Smad/efectos de los fármacos , Transfección , Factor de Crecimiento Transformador beta1
17.
Head Neck ; 28(5): 432-40, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16320362

RESUMEN

BACKGROUND: As part of ongoing studies aimed at identifying the molecular events involved in head and neck squamous cell carcinoma progression, we recently isolated a novel serine protease, DESC1. This study was conducted to further characterize DESC1. METHODS: Specimens of normal, dysplastic, and carcinomatous oropharyngeal mucosa (n = 31) were evaluated for DESC1 immunoreactivity using standard streptavidin-biotin immunoperoxidase techniques. Between-lesion stain intensity values were analyzed using multiple Wilcoxon tests. DESC1 expression was also evaluated in cultured human keratinocytes after induction of differentiation by calcium challenge, with subsequent real-time reverse transcriptase-polymerase chain reaction quantification. RESULTS: DESC1 immunoreactivity decreased as lesions approached a malignant phenotype. Post hoc testing comparing the different lesion types and DESC1 staining values showed significance between "normal" and "carcinoma" (p = .0017) groups. Induction of normal keratinocyte differentiation by calcium challenge was accompanied by an increase in DESC1 expression (p = .002). CONCLUSIONS: These results suggest downregulation of DESC1 during squamous cell carcinoma progression and upregulation during normal epithelial differentiation.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/patología , Lesiones Precancerosas/metabolismo , Serina Endopeptidasas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Diferenciación Celular , Femenino , Humanos , Queratinocitos/citología , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/patología , ARN Mensajero/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Serina Endopeptidasas/genética
18.
Methods Mol Biol ; 281: 333-48, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15220541

RESUMEN

Telomeres are the structures at the ends of chromosomes, composed of repetitive sequences and associated proteins, which cap chromosome ends to maintain genomic stability. These structures are maintained by the enzyme complex telomerase in germ cells and some stem cells, but are absent in the majority of somatic cells. The consequence of this lack of telomerase in normal somatic cells is the shortening of the telomeric repeat, which results in a limited replicative life span. However, in cancer cells, which grow indefinitely, telomerase activity has been detected in a large number of different cancer cell types. This has lead to a great deal of interest in establishing techniques to measure telomerase activity, telomere length, and telomere function in both normal and cancer cells/tissue. Here we describe the TRAP (telomeric repeat amplification protocol) assay, a technique to measure telomerase activity in cells, TRF (terminal restriction fragment length) analysis to estimate telomere length, and both the anaphase bridge index and the frequency of dicentric chromosomes as indicators of telomere dysfunction.


Asunto(s)
Neoplasias/enzimología , Telomerasa/metabolismo , Telómero/fisiología , Aberraciones Cromosómicas , Humanos , Secuencias Repetitivas de Ácidos Nucleicos/genética
19.
Exp Cell Res ; 295(2): 525-38, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15093749

RESUMEN

We show here that histone deacetylase inhibitors (HDACIs) sodium dibutyrate (SDB) and trichostatin A (TSA) induce a phenotype that has similarities to replicative senescence in human fibroblasts. There was no evidence that SDB accelerated a constitutive cell division counting mechanism as previously suggested because cells pretreated with SDB for three mean population doublings (MPDs) exhibited a similar overall proliferative life span to controls once SDB was withdrawn. SDB-treated cells upregulated the cell cycle inhibitors p21(WAF1) and p16(INK4A), but not p14(ARF), in the same sequential order as in senescence and the cells developed biochemical markers of senescence. However, the mechanism of senescence did not involve telomere dysfunction and there was no evidence for any posttranslational modification of p53. The expression of human papillomavirus (HPV) 16 E6 in human fibroblasts or targeted disruption of the p53 and p21(WAF) genes only weakly antagonized HDACI-induced senescence. However, expression of the E7 gene, which inhibits the function of pRb, cooperated with E6 to block SDB-induced senescence completely and human cells deficient in p16(INK4A) (but not p14(ARF)) were also resistant to SDB-induced senescence, suggesting that the p16(INK4A)/pRb pathway is the major mediator of HDACI-induced senescence in human cells. However, p53-/- mouse fibroblasts were resistant to HDACI-induced senescence, identifying p53 as the major pathway to senescence in this species.


Asunto(s)
División Celular/fisiología , Senescencia Celular/fisiología , Inhibidores Enzimáticos/farmacología , Fibroblastos/metabolismo , Inhibidores de Histona Desacetilasas , Animales , Ácido Butírico/farmacología , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Diploidia , Feto , Fibroblastos/enzimología , Regulación de la Expresión Génica , Histona Desacetilasas/efectos de los fármacos , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Cariotipificación , Ratones , Papillomaviridae/metabolismo , Pruebas de Precipitina , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Piel/citología , Especificidad de la Especie , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
20.
Mol Cell Biol ; 24(4): 1540-59, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14749371

RESUMEN

Invasion is generally perceived to be a late event during the progression of human cancer, but to date there are no consistent reports of alterations specifically associated with malignant conversion. We provide evidence that the v-Fos oncogene induces changes in gene expression that render noninvasive normal human diploid fibroblasts highly invasive, without inducing changes in growth factor requirements or anchorage dependence for proliferation. Furthermore, v-Fos-stimulated invasion is independent of the pRb/p16(INK4a) and p53 tumor suppressor pathways and telomerase. We have performed microarray analysis using Affymetrix GeneChips, and the gene expression profile of v-Fos transformed cells supports its role in the regulation of invasion, independent from proliferation. We also demonstrate that invasion, but not proliferation, is dependent on the activity of the up-regulated epidermal growth factor receptor. Taken together, these results indicate that AP-1-directed invasion could precede deregulated proliferation during tumorigenesis and that sustained activation of AP-1 could be the epigenetic event required for conversion of a benign tumor into a malignant one, thereby explaining why many malignant human tumors present without an obvious premalignant hyperproliferative dysplastic lesion.


Asunto(s)
Transformación Celular Neoplásica/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Invasividad Neoplásica/patología , Proteínas Oncogénicas v-fos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , División Celular , Transformación Celular Neoplásica/genética , Daño del ADN , Receptores ErbB/genética , Receptores ErbB/metabolismo , Perfilación de la Expresión Génica , Humanos , Cariotipificación , Ratones , Invasividad Neoplásica/genética , Proteínas Oncogénicas v-fos/genética , Transducción de Señal , Telomerasa/genética , Telomerasa/metabolismo , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...