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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(7)2024 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-38612901

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas , Anemia de Fanconi , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Neoplasias Bucais/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço , DNA
2.
Cancer Metastasis Rev ; 40(4): 1073-1091, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34932167

RESUMO

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.


Assuntos
Ácido Cítrico , Neoplasias , Citratos , Ácido Cítrico/metabolismo , Ciclo do Ácido Cítrico , Humanos , Neoplasias/metabolismo , Fosforilação Oxidativa , Microambiente Tumoral/fisiologia
3.
Biology (Basel) ; 12(1)2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36671795

RESUMO

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.
Sci Rep ; 11(1): 584, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436723

RESUMO

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.


Assuntos
Autofagia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Senescência Celular/genética , Fibroblastos/patologia , Fibroblastos/fisiologia , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Fator de Crescimento Transformador beta1/fisiologia , Autofagia/efeitos dos fármacos , Autofagia/genética , Autofagia/fisiologia , Diferenciação Celular/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Senescência Celular/efeitos dos fármacos , Humanos , Miofibroblastos/patologia , Invasividade Neoplásica/genética , Piridinas/farmacologia , Pirimidinonas/farmacologia
5.
Front Cell Dev Biol ; 8: 602476, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33425906

RESUMO

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.

6.
Exp Cell Res ; 314(13): 2434-47, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18589416

RESUMO

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.


Assuntos
Senescência Celular/genética , Queratinócitos/metabolismo , Telômero/fisiologia , Transcrição Gênica , Células 3T3 , Animais , Proliferação de Células , Células Cultivadas , Células Clonais , Dano ao DNA/genética , Perfilação da Expressão Gênica , Humanos , Recém-Nascido , Queratinócitos/patologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Telômero/patologia , Proteína 2 de Ligação a Repetições Teloméricas/genética , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Transgenes
7.
Cancers (Basel) ; 11(11)2019 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-31717573

RESUMO

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.
Oral Oncol ; 44(3): 261-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17475542

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias Bucais/patologia , Tolerância a Radiação , Telômero/ultraestrutura , Anáfase , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/radioterapia , Linhagem Celular Tumoral , Sobrevivência Celular , Senescência Celular , Raios gama , Humanos , Neoplasias Bucais/enzimologia , Neoplasias Bucais/radioterapia , Análise de Regressão , Estatísticas não Paramétricas , Telomerase/metabolismo , Falha de Tratamento
9.
J Oral Pathol Med ; 37(4): 211-20, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18205743

RESUMO

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.


Assuntos
Queratinócitos/patologia , Queratinas/biossíntese , Fibrose Oral Submucosa/patologia , Biomarcadores , Carcinoma de Células Escamosas , Estudos de Casos e Controles , Transformação Celular Neoplásica , Expressão Gênica , Humanos , Imuno-Histoquímica , Queratina-17/biossíntese , Queratinócitos/química , Queratinas/genética , Neoplasias Bucais/química , Neoplasias Bucais/patologia , Fenótipo , Fotografia Dentária , Lesões Pré-Cancerosas/química , Lesões Pré-Cancerosas/patologia , Índice de Gravidade de Doença
10.
Sci Rep ; 8(1): 12148, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30108296

RESUMO

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.


Assuntos
Envelhecimento/sangue , Senescência Celular/fisiologia , Fibroblastos/metabolismo , Metaboloma/fisiologia , Envelhecimento/metabolismo , Células Cultivadas , Ácido Cítrico/sangue , Ácido Cítrico/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Dano ao DNA/fisiologia , Humanos , Metabolômica
11.
BioDrugs ; 21(6): 375-85, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18020621

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Neoplasias/tratamento farmacológico , Telomerase/antagonistas & inibidores , Telômero/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , RNA/antagonistas & inibidores , RNA/genética , RNA/metabolismo , Telomerase/genética , Telomerase/metabolismo , Telômero/genética
12.
Mol Cell Biol ; 24(4): 1540-59, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14749371

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/patologia , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Invasividade Neoplásica/patologia , Proteínas Oncogênicas v-fos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Divisão Celular , Transformação Celular Neoplásica/genética , Dano ao DNA , Receptores ErbB/genética , Receptores ErbB/metabolismo , Perfilação da Expressão Gênica , Humanos , Cariotipagem , Camundongos , Invasividade Neoplásica/genética , Proteínas Oncogênicas v-fos/genética , Transdução de Sinais , Telomerase/genética , Telomerase/metabolismo , Proteína Supressora de Tumor p53/genética , Regulação para Cima , Cicatrização
14.
Int J Oncol ; 28(5): 1279-85, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16596245

RESUMO

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.


Assuntos
Neoplasias de Cabeça e Pescoço/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Linhagem Celular Tumoral , Humanos , Fosforilação , Plasmídeos , Proteínas Smad/efeitos dos fármacos , Transfecção , Fator de Crescimento Transformador beta1
15.
Cancer Res ; 63(2): 458-67, 2003 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-12543803

RESUMO

Human epithelial cells experience multiple barriers to cellular immortality in culture (mortality mechanisms 0, 1, and 2). Mortality mechanism 2 (M2) is termed crisis and involves telomere dysfunction due to lack of telomerase. However, proliferating normal keratinocytes in vivo can express telomerase, so it is unclear whether human squamous cell carcinomas (SCCs), which usually have high telomerase levels, develop from preexisting telomerase-positive precursors or by the activation of telomerase in telomerase-deficient somatic cells. We show that 6 of 29 oral SCCs show characteristics of M2 crisis in vivo, as indicated by a high anaphase bridge index (ABI), which is a good correlate of telomere dysfunction, and that 25 of 29 tumors possess some anaphase bridges. ABIs in excess of 0.2 in the primary tumor showed a decrease in the corresponding lymph node metastases. This suggests that high levels of telomere dysfunction (>0.2) and, by inference, M2 crisis bestow a selective disadvantage on SCCs during progression stages of the disease. Supporting this, SCCs with high levels of telomere dysfunction grow poorly in culture, and the ectopic expression of telomerase corrects this, together with other features of M2 crisis. Our data suggest that a substantial proportion of oral SCCs in vivo ultimately arise from telomerase-deficient keratinocytes rather than putative telomerase-proficient cells in the undifferentiated parts of the epithelium. Furthermore, the presence of significant levels of telomere dysfunction in a high proportion of SCCs at diagnosis but not in the normal epithelium implies that the therapeutic inhibition of telomerase should selectively compromise the growth of such tumors.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias Bucais/patologia , Telômero/fisiologia , Células 3T3 , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Proteínas de Ligação a DNA , Progressão da Doença , Humanos , Imuno-Histoquímica , Queratinócitos/metabolismo , Queratinócitos/patologia , Queratinócitos/fisiologia , Camundongos , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Telomerase/biossíntese , Telomerase/genética , Telomerase/metabolismo , Células Tumorais Cultivadas
16.
Cancer Res ; 62(16): 4757-66, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12183435

RESUMO

This study has identified molecular changes characteristic of early oral cancer progression. We reported previously that acquisition of the immortal phenotype is an early event in oral cancer development (F. McGregor et al., Cancer Res., 57: 3886-3889, 1997); our current data indicate that about half of oral dysplasia cultures are immortal, and this is associated with loss of expression of retinoic acid receptor (RAR)-beta and the cell cycle inhibitor p16(ink4a) (p16), p53 mutations, and increased levels of telomerase/human telomerase reverse transcriptase mRNA. In contrast, increased expression of the epidermal growth factor receptor, known to be a characteristic of oral cancer, does not occur until after the dysplasia stage in squamous cell carcinomas. Acquisition of invasive properties as judged by an in vitro Matrigel invasion assay also does not occur until the carcinoma stage and is further increased in metastases. Interestingly, one atypical mortal dysplasia with a considerably extended life span has lost expression of RAR-beta and p16, but it still expresses only wild-type p53 (albeit at a higher level than normal) and has not activated telomerase. RAR-beta and/or p16 re-expression can be induced by treatment with 5-aza-2-deoxycytidine (Aza-C) in some immortal dysplasias, and this has been shown to be due to silencing of gene expression by promoter methylation. Aza-C treatment also down-regulated telomerase activity and human telomerase reverse transcriptase mRNA. Interestingly, with one dysplasia, Aza-C was able to reverse its immortal phenotype, as judged by morphological criteria and expression of the senescence-associated acid beta-galactosidase activity during terminal growth arrest; this immortal dysplasia was the only one in which Aza-C treatment not only down-regulated telomerase activity but also induced re-expression of both RAR-beta and p16. The possibility of reversing the immortal phenotype of some dysplasias by Aza-C may be of clinical usefulness.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Mucosa Bucal/patologia , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/metabolismo , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Transformação Celular Neoplásica/genética , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/biossíntese , Ciclinas/genética , Proteínas de Ligação a DNA , Progressão da Doença , Receptores ErbB/biossíntese , Receptores ErbB/genética , Humanos , Mucosa Bucal/metabolismo , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Neoplasias Bucais/prevenção & controle , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Proteínas Quinases/biossíntese , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores do Ácido Retinoico/biossíntese , Receptores do Ácido Retinoico/genética , Telomerase/biossíntese , Telomerase/genética , Telomerase/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Quinase 1 Polo-Like
17.
Sci Rep ; 6: 38489, 2016 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-27924925

RESUMO

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.


Assuntos
Senescência Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Espaço Intracelular/metabolismo , Metaboloma , Metabolômica , NAD/metabolismo , Niacinamida/metabolismo , Ciclo Celular , Proliferação de Células , Células Cultivadas , Análise por Conglomerados , Homeostase , Humanos , Modelos Biológicos , Oxirredução , Análise de Componente Principal , Triptofano/metabolismo
18.
Oncogene ; 22(49): 7804-8, 2003 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-14586406

RESUMO

Our previous work showed that acquisition of immortality at the dysplasia stage of oral cancer progression was consistently associated with four changes: loss of retinoic acid receptor (RAR)-beta and p16INK4A expression, p53 mutations and activation of telomerase. One atypical dysplasia (D17) that underwent delayed senescence after an extended lifespan showed loss of RAR-beta and p16INK4A/p14ARF expression, but retained functional wild-type p53 and telomerase was not activated. We now demonstrate that retroviral delivery of hTERT results in telomere lengthening and immortalization of D17 without loss of functional wild-type p53 activity. In contrast, the expression of hTERT in two other typical mortal dyplasia cultures (that retain RAR-beta and p16INK4A expression) does not extend their lifespan, even though telomeres are lengthened.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Genes p53/genética , Mucosa Bucal/patologia , Neoplasias Bucais/patologia , Lesões Pré-Cancerosas/patologia , Receptores do Ácido Retinoico/fisiologia , Telomerase/genética , Senescência Celular , Proteínas de Ligação a DNA , Humanos , Mutação , Fosforilação , Retroviridae/genética , Telômero
19.
Oncogene ; 21(10): 1616-24, 2002 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-11896591

RESUMO

This study examined the role of TGF-beta1 in human keratinocyte malignancy. Two carcinoma-derived human oral keratinocyte cell lines, BICR 31 and H314, were selected on the basis of their known resistance to TGF-beta1-induced G(1) arrest, the presence of wild type TGF-beta cell surface receptors and normal Ras. Smad 4 protein was undetectable in both cell lines, but Smad 2 and Smad 3 were expressed at levels comparable with a fully TGF-beta responsive cell line, and treatment of the cells with TGF-beta1 resulted in the phosphorylation of Smad 2. Treatment with exogenous TGF-beta1 resulted in a failure to induce transcription from an artificial Smad-dependent promoter and a failure to down-regulate c-myc, but resulted in an up-regulation of AP-1 associated genes (Fra-1, JunB and fibronectin). Transient transfection of Smad 4 into BICR 31 restored TGF-beta1-induced growth inhibition and Smad-dependent transcriptional activation. Protracted treatment of cells with exogenous TGF-beta1 resulted in the attenuation of cell growth in vitro. To over-express TGF-beta1, both cell lines were transfected with latent TGF-beta1 cDNA; neutralization studies of conditioned media demonstrated that whilst the majority of the peptide was in the latent form, a small proportion was present as the active peptide. Cells that over-expressed endogenous TGF-beta1 grew more slowly in vitro compared to both the vector-only controls and cells that did not over-express the peptide. Orthotopic transplantation of cells that over-expressed endogenous TGF-beta1 to the floor of the mouth in athymic mice resulted in marked inhibition of primary tumor formation compared to controls. Expression of a dominant-negative TGF-beta type II receptor in cells that over-expressed endogenous TGF-beta1 resulted in enhanced cell growth in vitro and diminished the tumor suppressor effect of the ligand in vivo, indicating that the endogenous TGF-beta1 was acting in an autocrine capacity. The results demonstrate that over-expression of endogenous TGF-beta1 in human malignant oral keratinocytes leads to growth inhibition in vivo and tumor suppression in vitro by mechanisms that are independent of Smad 4 expression and TGF-beta1-induced G(1) arrest.


Assuntos
Carcinoma/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neoplasias Cutâneas/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Carcinoma/patologia , Divisão Celular , Proteínas de Ligação a DNA/genética , Fase G1 , Humanos , Queratinócitos/metabolismo , Cinética , Ligantes , Camundongos , Camundongos Nus , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Transplante de Neoplasias , RNA Neoplásico/biossíntese , Neoplasias Cutâneas/patologia , Proteína Smad4 , Transativadores/genética , Transcrição Gênica , Transfecção , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1 , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/farmacologia
20.
Oncogene ; 21(33): 5135-47, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12140764

RESUMO

Squamous cell carcinoma (SCC) immortality is associated with p53 and INK4A dysfunction, high levels of telomerase and loss of heterozygosity (LOH) of other chromosomes, including chromosome 4. To test for a functional cancer mortality gene on human chromosome 4 we introduced a complete or fragmented copy of the chromosome into SCC lines by microcell-mediated chromosome transfer (MMCT). Human chromosome 4 caused a delayed crisis, specifically in SCC lines with LOH on chromosome 4, but chromosomes 3, 6, 11 and 15 were without effect. The introduction of the telomerase reverse transcriptase into the target lines extended the average telomere terminal fragment length but did not affect the frequency of mortal hybrids following MMCT of chromosome 4. Furthermore, telomerase activity was still present in hybrids displaying the mortal phenotype. The MMCT of chromosomal fragments into BICR6 mapped the mortality gene to between the centromere and 4q23. Deletion analysis of the introduced chromosome in immortal segregants narrowed the candidate interval to 2.7 Mb spanning D4S423 and D4S1557. The results suggest the existence of a gene on human chromosome 4 whose dysfunction contributes to the continuous proliferation of SCC and that this gene operates independently from telomeres, p53 and INK4A.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Transformação Celular Neoplásica/genética , Cromossomos Humanos Par 4/genética , Perda de Heterozigosidade/genética , Animais , Sobrevivência Celular , Mapeamento Cromossômico , Coloração Cromossômica , Proteínas de Ligação a DNA , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Repetições de Microssatélites/genética , Fenótipo , Telomerase/antagonistas & inibidores , Telomerase/metabolismo , Telômero/metabolismo , Telômero/patologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA