Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Exp Cell Res ; 386(1): 111690, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31678172

RESUMO

Abundant with organelle-like membranous structures, the tumor microenvironment is composed of cancer cells that secrete exosomes. Studies have shown that these secreted exosomes transport RNA and active molecules to other cells to reshape the tumor microenvironment and promote tumor growth. In fact, we found that exosomes derived from melanoma cells drive pre-malignant transition in primary melanocytes. However, there is little available in the scientific literature on how exosomes modulate melanocytes in the microenvironment to optimize conditions for tumor progression and metastasis. We therefore focused this current study on identifying these conditions genetically. Through RNA sequencing, we analyzed gene expression levels of melanocytes driven by exosomes derived from melanoma and lung cancer cells compared with those without exosome controls. Significant differences were found in gene expression patterns of melanocytes driven by exosomes derived from melanoma and lung cancer cells. In the melanocytes responding to exosomes derived from melanoma cells, genes of lipopolysaccharide and regulation of leukocyte chemotaxis were predominant. In the melanocytes responding to exosomes derived from lung cancer cells, genes of DNA replication and mitotic nuclear division played an important role. These results provide further mechanistic understanding of tumor progression promoted by tumor-derived exosomes. This will also help identify potential therapeutic targets for melanoma progression.


Assuntos
Exossomos/metabolismo , Melanócitos/metabolismo , Melanoma/genética , Transcriptoma , Células A549 , Células Cultivadas , Exossomos/genética , Exossomos/patologia , Perfilação da Expressão Gênica , Humanos , Microambiente Tumoral
2.
Ann Surg Oncol ; 25(5): 1296-1303, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29497912

RESUMO

BACKGROUND: Current risk assessment tools to estimate the risk of nonsentinel lymph node metastases after completion lymphadenectomy for a positive sentinel lymph node (SLN) biopsy in cutaneous melanoma are based on clinical and pathologic factors. We identified a novel genetic signature that can predict non-SLN metastases in patients with cutaneous melanoma staged with a SLN biopsy. METHODS: RNA was collected for tumor-positive SLNs in patients staged by SLN biopsy for cutaneous melanoma. All patients with a tumor-positive SLN biopsy underwent completion lymphadenectomy. A 1:10 case:control series of positive and negative non-SLN patients was analyzed by microarray and quantitative RT-PCR. Candidate differentially expressed genes were validated in a 1:3 case:control separate cohort of positive and negative non-SLN patients. RESULTS: The 1:10 case:control discovery set consisted of 7 positive non-SLN cases matched to 70 negative non-SLN controls. The cases and controls were similar with regards to important clinicopathologic factors, such as gender, primary tumor site, age, ulceration, and thickness. Microarray and RT-PCR identified six potential differentially expressed genes for validation. In the 40-patient separate validation set, 10 positive non-SLN patients were matched to 30 negative non-SLN controls based on gender, ulceration, age, and thickness. Five of the six genes were differentially expressed. The five gene panel identified patients at low (7.1%) and high risk (66.7%) for non-SLN metastases. CONCLUSIONS: A novel, non-SLN gene score based on differential expressed genes in a tumor-positive SLN can identify patients at high and low risk for non-SLN metastases.


Assuntos
Melanoma/genética , Melanoma/secundário , Linfonodo Sentinela , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Transcriptoma , Adulto , Área Sob a Curva , Estudos de Casos e Controles , Feminino , Humanos , Excisão de Linfonodo , Linfonodos/patologia , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Curva ROC , Linfonodo Sentinela/patologia , Linfonodo Sentinela/cirurgia , Biópsia de Linfonodo Sentinela
3.
Ann Surg Oncol ; 24(1): 108-116, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27663566

RESUMO

PURPOSE: Melanoma patients with a single microscopically-positive sentinel lymph node (SLN) are classified as stage III and are often advised to undergo expensive and substantially toxic adjuvant therapy. However, the 5-year survival rate for these patients, with or without adjuvant therapy, varies from 14 to 85 %, representing a heterogeneous biological population with a variable prognosis. We aimed to identify an SLN gene signature to aid in risk stratification of patients with tumor-positive SLNs. METHODS: Microarray experiments were performed to screen SLN genes in recurrence (N = 39) versus non-recurrence (N = 58) groups in the training dataset. Quantitative reverse-transcriptase polymerase chain reaction (RT-PCR) assay was applied to confirm the expression of selected SLN genes, which were further verified using an independent validation cohort (N = 30). Area under the receiver operating characteristic curve (AUC) was calculated to evaluate prognostic accuracy of the selected SLN gene panel, and the prognostic value of our SLN gene signature was also compared with the current American Joint Committee on Cancer (AJCC) staging system. RESULTS: We identified two SLN genes (PIGR and TFAP2A) that provided high prognostic accuracy in SLN-positive melanoma patients (AUC = 0.864). These two SLN genes, along with clinicopathological features, can differentiate the high- and low-risk groups in node-positive melanoma patients in this cohort. CONCLUSION: The two SLN genes, when combined with clinicopathological features, may offer a new tool for personalized patient risk assessment.


Assuntos
Metástase Linfática/patologia , Melanoma/genética , Melanoma/patologia , Receptores de Imunoglobulina Polimérica/genética , Linfonodo Sentinela/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Fator de Transcrição AP-2/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medição de Risco , Biópsia de Linfonodo Sentinela , Taxa de Sobrevida
4.
Exp Mol Pathol ; 90(2): 215-25, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21236253

RESUMO

Hypoxia confers resistance to chemoradiation therapy and promotes metastasis in head and neck squamous cell carcinomas (HNSCC). We investigated the effects of hypoxia in tumor phenotype using immunocompetent murine HNSCC models. Balb/c mice were injected intraorally with murine squamous cell carcinoma cells LY-2 and B4B8. Intratumoral hypoxia fraction was evaluated by the immunohistochemical detection of hypoxic probe pimonidazole and carbonic anhydrase IX (CAIX). Tumor cell apoptosis and autophagy in hypoxic areas of these tumors were examined immunohistochemically. Hypoxia-induced apoptotic and autophagic responses in vitro were examined by treating LY2 cells with CoCl(2). B4B8 tumors exhibited a non-aggressive phenotype characterized by its slow growth rate and the lack of metastatic spread. LY2 tumors demonstrated an aggressive phenotype characterized by rapid growth rate with regional and distant metastasis. Intratumoral hypoxia fraction in B4B8 tumors was significantly lower than in LY2 tumors. The hypoxic areas in B4B8 tumors exhibited increased apoptosis rate than that of LY2 tumors. In contrast, the hypoxic areas in LY2 tumors revealed autophagy. The induction of hypoxia in vitro elicited autophagy and not apoptosis in LY2 cells. The induction of autophagy coupled with blockage of apoptosis in hypoxic areas promotes tumor cell survival and confers aggressive phenotype in immunocompetent murine HNSCC models.


Assuntos
Autofagia , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/patologia , Imunocompetência , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Carcinoma de Células Escamosas/enzimologia , Caspase 3/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Neoplasias de Cabeça e Pescoço/enzimologia , Imuno-Histoquímica , Camundongos , Neoplasias Bucais/patologia , Metástase Neoplásica , Fenótipo , Regulação para Cima
5.
Nephron Exp Nephrol ; 119(1): e8-20, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21606656

RESUMO

AIM: To define renal gene expression during the development of severe albuminuria in OVE26 diabetic mice. METHODS: Kidney microarray analysis was performed at 2, 4 and 8 months. Data were validated by RT-PCR, in situ hybridization and immunohistochemistry. RESULTS: Gene expression differences between control and diabetic mice increased 10-fold from 2 to 8 months. This change was most obvious for inflammatory genes. Three inflammatory genes, complement C3, VCAM1 and CD44 were upregulated more than 4-fold. Inflammatory gene expression correlated with albuminuria and C3 and CD44 increased in tubules that accumulated albumin. VCAM1 was induced in different tubules that were neither dilated nor accumulated albumin. Six of 8 genes previously reported to be markers of human advanced diabetic nephropathy and the NF-κB_IFN_x promoter module were elevated in the oldest diabetic mice. Vitamin D inhibits diabetic renal inflammation. Vitamin D and mRNA for vitamin D synthetic enzyme CYP2B1 were elevated in kidneys of young OVE26 mice. CONCLUSIONS: OVE26 mice induce inflammatory genes consistent with advanced renal disease, associated with severe albuminuria and to a greater extent than reported in other diabetic models. They provide an excellent model of diabetic nephropathy to assess the effect of induction of inflammatory proteins.


Assuntos
Quimiocinas/genética , Complemento C3/genética , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/fisiopatologia , Receptores de Hialuronatos/genética , Inflamação/genética , Albuminúria/fisiopatologia , Animais , Citocromo P-450 CYP2B1/genética , Feminino , Expressão Gênica , Camundongos , Camundongos Endogâmicos , Análise Serial de Proteínas , Regulação para Cima
6.
Sci Rep ; 11(1): 15715, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34344959

RESUMO

Key elements for viral pathogenesis include viral strains, viral load, co-infection, and host responses. Several studies analyzing these factors in the function of disease severity of have been published; however, no studies have shown how all of these factors interplay within a defined cohort. To address this important question, we sought to understand how these four key components interplay in a cohort of COVID-19 patients. We determined the viral loads and gene expression using high throughput sequencing and various virological methods. We found that viral loads in the upper respiratory tract in COVID-19 patients at an early phase of infection vary widely. While the majority of nasopharyngeal (NP) samples have a viral load lower than the limit of detection of infectious viruses, there are samples with an extraordinary amount of SARS-CoV-2 RNA and a high viral titer. No specific viral factors were identified that are associated with high viral loads. Host gene expression analysis showed that viral loads were strongly correlated with cellular antiviral responses. Interestingly, however, COVID-19 patients who experience mild symptoms have a higher viral load than those with severe complications, indicating that naso-pharyngeal viral load may not be a key factor of the clinical outcomes of COVID-19. The metagenomics analysis revealed that the microflora in the upper respiratory tract of COVID-19 patients with high viral loads were dominated by SARS-CoV-2, with a high degree of dysbiosis. Finally, we found a strong inverse correlation between upregulation of interferon responses and disease severity. Overall our study suggests that a high viral load in the upper respiratory tract may not be a critical factor for severe symptoms; rather, dampened antiviral responses may be a critical factor for a severe outcome from the infection.


Assuntos
COVID-19/patologia , Interferons/metabolismo , SARS-CoV-2/genética , Adulto , Idoso , COVID-19/virologia , Disbiose/etiologia , Feminino , Humanos , Masculino , Metagenômica , Microbiota/genética , Pessoa de Meia-Idade , Nasofaringe/virologia , RNA Viral/análise , Reação em Cadeia da Polimerase em Tempo Real , Sistema Respiratório/microbiologia , Sistema Respiratório/virologia , SARS-CoV-2/isolamento & purificação , Índice de Gravidade de Doença , Transcriptoma , Regulação para Cima , Carga Viral
7.
Pathogens ; 10(10)2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34684220

RESUMO

Throughout the course of the ongoing SARS-CoV-2 pandemic there has been a need for approaches that enable rapid monitoring of public health using an unbiased and minimally invasive means. A major way this has been accomplished is through the regular assessment of wastewater samples by qRT-PCR to detect the prevalence of viral nucleic acid with respect to time and location. Further expansion of SARS-CoV-2 wastewater monitoring efforts to include the detection of variants of interest/concern through next-generation sequencing has enhanced the understanding of the SARS-CoV-2 outbreak. In this report, we detail the results of a collaborative effort between public health and metropolitan wastewater management authorities and the University of Louisville to monitor the SARS-CoV-2 pandemic through the monitoring of aggregate wastewater samples over a period of 28 weeks. Through the use of next-generation sequencing approaches the polymorphism signatures of Variants of Concern/Interest were evaluated to determine the likelihood of their prevalence within the community on the basis of their relative dominance within sequence datasets. Our data indicate that wastewater monitoring of water quality treatment centers and smaller neighborhood-scale catchment areas is a viable means by which the prevalence and genetic variation of SARS-CoV-2 within a metropolitan community of approximately one million individuals may be monitored, as our efforts detected the introduction and emergence of variants of concern in the city of Louisville. Importantly, these efforts confirm that regional emergence and spread of variants of interest/concern may be detected as readily in aggregate wastewater samples as compared to the individual wastewater sheds. Furthermore, the information gained from these efforts enabled targeted public health efforts including increased outreach to at-risk communities and the deployment of mobile or community-focused vaccination campaigns.

8.
Aging (Albany NY) ; 12(24): 24914-24939, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33373316

RESUMO

Age is an important factor for determining the outcome of melanoma patients. Sentinel lymph node (SLN) status is also a strong predictor of survival for melanoma. Paradoxically, older melanoma patients have a lower incidence of SLN metastasis but a higher mortality rate when compared with their younger counterparts. The mechanisms that underlie this phenomenon remain unknown. This study uses three independent datasets of RNA samples from patients with melanoma metastatic to the SLN to identify age-related transcriptome changes in SLNs and their association with outcome. Microarray was applied to the first dataset of 97 melanoma patients. NanoString was performed in the second dataset to identify the specific immune genes and pathways that are associated with recurrence in younger versus older patients. qRT-PCR analysis was used in the third dataset of 36 samples to validate the differentially expressed genes (DEGs) from microarray and NanoString. These analyses show that FOS, NR4A, and ITGB1 genes were significantly higher in older melanoma patients with positive SLNs. IRAK3- and Wnt10b-related genes are the major pathways associated with recurrent melanoma in younger and older patients with tumor-positive SLNs, respectively. This study aims to elucidate age-related differences in SLNs in the presence of nodal metastasis.


Assuntos
Melanoma/genética , Linfonodo Sentinela/patologia , Neoplasias Cutâneas/genética , Adulto , Fatores Etários , Idoso , Proteínas Relacionadas à Autofagia/genética , Moléculas de Adesão Celular/genética , Feminino , Humanos , Integrina beta1/genética , Quinases Associadas a Receptores de Interleucina-1/genética , Lectinas Tipo C/genética , Metástase Linfática , Masculino , Melanoma/patologia , Glicoproteínas de Membrana/genética , Pessoa de Meia-Idade , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-fos/genética , Receptores Imunológicos/genética , Transdução de Sinais , Neoplasias Cutâneas/patologia , Transcriptoma , Proteínas Wnt/genética
9.
BMC Cancer ; 7: 108, 2007 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-17592646

RESUMO

BACKGROUND: TRAIL plays an important role in host immunosurveillance against tumor progression, as it induces apoptosis of tumor cells but not normal cells, and thus has great therapeutic potential for cancer treatment. TRAIL binds to two cell-death-inducing (DR4 and DR5) and two decoy (DcR1, and DcR2) receptors. Here, we compare the expression levels of TRAIL and its receptors in normal oral mucosa (NOM), oral premalignancies (OPM), and primary and metastatic oral squamous cell carcinomas (OSCC) in order to characterize the changes in their expression patterns during OSCC initiation and progression. METHODS: DNA microarray, immunoblotting and immunohistochemical analyses were used to examine the expression levels of TRAIL and its receptors in oral epithelial cell lines and in archival tissues of NOM, OPM, primary and metastatic OSCC. Apoptotic rates of tumor cells and tumor-infiltrating lymphocytes (TIL) in OSCC specimens were determined by cleaved caspase 3 immunohistochemistry. RESULTS: Normal oral epithelia constitutively expressed TRAIL, but expression was progressively lost in OPM and OSCC. Reduction in DcR2 expression levels was noted frequently in OPM and OSCC compared to respective patient-matched uninvolved oral mucosa. OSCC frequently expressed DR4, DR5 and DcR1 but less frequently DcR2. Expression levels of DR4, DR5 and DcR1 receptors were not significantly altered in OPM, primary OSCC and metastatic OSCC compared to patient-matched normal oral mucosa. Expression of proapoptotic TRAIL-receptors DR4 and DR5 in OSCC seemed to depend, at least in part, on whether or not these receptors were expressed in their parental oral epithelia. High DR5 expression in primary OSCC correlated significantly with larger tumor size. There was no significant association between TRAIL-R expression and OSSC histology grade, nodal status or apoptosis rates of tumor cells and TIL. CONCLUSION: Loss of TRAIL expression is an early event during oral carcinogenesis and may be involved in dysregulation of apoptosis and contribute to the molecular carcinogenesis of OSCC. Differential expressions of TRAIL receptors in OSCC do not appear to play a crucial role in their apoptotic rate or metastatic progression.


Assuntos
Carcinoma de Células Escamosas/fisiopatologia , Transformação Celular Neoplásica/patologia , Neoplasias Bucais/fisiopatologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Adulto , Biópsia por Agulha , Carcinoma de Células Escamosas/genética , Estudos de Casos e Controles , Morte Celular/genética , Morte Celular/fisiologia , Transformação Celular Neoplásica/genética , Progressão da Doença , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mucosa Bucal/metabolismo , Mucosa Bucal/patologia , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Prognóstico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Valores de Referência , Sensibilidade e Especificidade , Ligante Indutor de Apoptose Relacionado a TNF/genética , Células Tumorais Cultivadas
10.
Toxicol Lett ; 170(2): 134-45, 2007 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-17399918

RESUMO

Cigarette smoke, which contains several carcinogens known to initiate and promote tumorigenesis and metastasis, is the major cause of oral cancer. Lysosomal cathepsin proteases play important roles in tumor progression, invasion and metastasis. In the present work we investigated the effects of cigarette smoke condensate (CSC) on cathepsin (B, D and L) expression and protease-mediated invasiveness in human oral squamous cell carcinoma (OSCC) cells. Our results show that treatment of OSCC cells (686Tu and 101A) with CSC activated cathepsins B, D and L in a dose-dependent manner. Both expression and activity of these cathepsins were up-regulated in CSC-exposed versus non-exposed cells. Although cathepsin L had the lowest basal level, it had the highest induction in exposed cells compared to cathepsins B and D. Suppression of CSC-induced cathepsin B and L activities by specific chemical inhibitors decreased the invasion process, suggesting that these proteases are involved in the invasion process. Overall, our results indicate that CSC activates cathepsin B and L proteolytic activity and enhances invasiveness in OSCC cells, a response that may play a role in CSC-mediated tumor progression and metastasis dissemination.


Assuntos
Carcinoma de Células Escamosas/patologia , Catepsinas/metabolismo , Neoplasias Bucais/patologia , Nicotiana , Alcatrões/toxicidade , Idoso , Apoptose/efeitos dos fármacos , Western Blotting , Carcinoma de Células Escamosas/enzimologia , Catepsinas/antagonistas & inibidores , Catepsinas/genética , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Bucais/enzimologia , Invasividade Neoplásica
11.
Genes (Basel) ; 8(5)2017 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-28467356

RESUMO

Smoking has been established as a major risk factor for developing oral squamous cell carcinoma (OSCC), but less attention has been paid to the effects of smokeless tobacco products. Our objective is to identify potential biomarkers to distinguish the biological effects of combustible tobacco products from those of non-combustible ones using oral cell lines. Normal human gingival epithelial cells (HGEC), non-metastatic (101A) and metastatic (101B) OSCC cell lines were exposed to different tobacco product preparations (TPPs) including cigarette smoke total particulate matter (TPM), whole-smoke conditioned media (WS-CM), smokeless tobacco extract in complete artificial saliva (STE), or nicotine (NIC) alone. We performed microarray-based gene expression profiling and found 3456 probe sets from 101A, 1432 probe sets from 101B, and 2717 probe sets from HGEC to be differentially expressed. Gene Set Enrichment Analysis (GSEA) revealed xenobiotic metabolism and steroid biosynthesis were the top two pathways that were upregulated by combustible but not by non-combustible TPPs. Notably, aldo-keto reductase genes, AKR1C1 and AKR1C2, were the core genes in the top enriched pathways and were statistically upregulated more than eight-fold by combustible TPPs. Quantitative real time polymerase chain reaction (qRT-PCR) results statistically support AKR1C1 as a potential biomarker for differentiating the biological effects of combustible from non-combustible tobacco products.

12.
Cell Rep ; 18(8): 1930-1945, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28228259

RESUMO

Embryonic stem cell (ESC) abnormalities in genome methylation hamper the utility of their therapeutic derivatives; however, the underlying mechanisms are unknown. Here, we show that the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase, Sirt1, selectively prevents abnormal DNA methylation of some developmental genes in murine ESCs by antagonizing Dnmt3l. Transcriptome and DNA methylome analyses demonstrated that Sirt1-null (Sirt1-/-) ESCs repress expression of a subset of imprinted and germline genes concomitant with increased DNA methylation of regulatory elements. Dnmt3l was highly expressed in Sirt1-/- ESCs, and knockdown partially rescued abnormal DNA methylation of the Sirt1 target genes. The Sirt1 protein suppressed transcription of Dnmt3l and physically interacted with the Dnmt3l protein, deacetylating and destabilizing Dnmt3l protein. Sirt1 deficiency delayed neurogenesis and spermatogenesis. These differentiation delays were significantly or partially abolished by reintroduction of Sirt1 cDNA or Dnmt3l knockdown. This study sheds light on mechanisms that restrain DNA methylation of developmentally vital genes operating in ESCs.


Assuntos
Diferenciação Celular/fisiologia , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Metilação de DNA/fisiologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Sirtuína 1/metabolismo , Animais , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , NAD/metabolismo , Neurogênese/fisiologia , Espermatogênese/fisiologia , Transcrição Gênica/fisiologia
13.
Environ Health Perspect ; 114(9): 1401-6, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16966095

RESUMO

BACKGROUND: Arsenic is both a human carcinogen and a chemotherapeutic agent, but the mechanism of neither arsenic-induced carcinogenesis nor tumor selective cytotoxicity is clear. Using a model cell line in which p53 expression is regulated exogenously in a tetracycline-off system (TR9-7 cells) , our laboratory has shown that arsenite disrupts mitosis and that p53-deficient cells [p53(-)], in contrast to p53-expressing cells [p53(+)], display greater sensitivity to arsenite-induced mitotic arrest and apoptosis. OBJECTIVE: Our goal was to examine the role p53 plays in protecting cells from arsenite-induced mitotic arrest. METHODS: p53(+) and p53(-) cells were synchronized in G2 phase using Hoechst 33342 and released from synchrony in the presence or absence of 5 microM sodium arsenite. RESULTS: Mitotic index analysis demonstrated that arsenite treatment delayed exit from G2 in p53(+) and p53(-) cells. Arsenite-treated p53(+) cells exited mitosis normally, whereas p53(-) cells exited mitosis with delayed kinetics. Microarray analysis performed on mRNAs of cells exposed to arsenite for 0 and 3 hr after release from G2 phase synchrony showed that arsenite induced inhibitor of DNA binding-1 (ID1) differentially in p53(+) and p53(-) cells. Immunoblotting confirmed that ID1 induction was more extensive and sustained in p53(+) cells. CONCLUSIONS: p53 promotes mitotic exit and leads to more extensive ID1 induction by arsenite. ID1 is a dominant negative inhibitor of transcription that represses cell cycle regulatory genes and is elevated in many tumors. ID1 may play a role in the survival of arsenite-treated p53(+) cells and contribute to arsenic carcinogenicity.


Assuntos
Apoptose/efeitos dos fármacos , Arsenitos/toxicidade , Ciclo Celular/efeitos dos fármacos , Genes p53/efeitos dos fármacos , Mitose/efeitos dos fármacos , Apoptose/fisiologia , Benzimidazóis/farmacologia , Ciclo Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Fase G2/efeitos dos fármacos , Fase G2/fisiologia , Genes p53/fisiologia , Humanos , Proteína 1 Inibidora de Diferenciação/metabolismo , Cinética , Análise em Microsséries , Mitose/fisiologia , RNA Mensageiro/metabolismo , Compostos de Sódio/toxicidade , Fatores de Tempo
14.
J Cancer Res Clin Oncol ; 132(3): 171-83, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16362335

RESUMO

PURPOSE: The death ligand TRAIL (tumor necrosis factor-related apoptosis inducing ligand) triggers apoptosis in a variety of cancer cells, which implies the potential for therapeutic applications. The purpose of this study was to investigate the role of the lysosomal protease cathepsin B (CB) in mediating TRAIL-induced cell death in oral squamous cell carcinoma (OSCC) cells. METHODS: OSCC cell lines from primary tumor and lymph node metastasis were examined for expression of apoptosis markers by Western blots, enzyme activity assays, nuclear fragmentation assays, and FACS analysis. Gene-specific ribozymes or chemical inhibitors were used to inhibit CB or caspases in target cells. RESULTS: TRAIL-induced activation of caspase-3, cleavage of Bid and poly-ADP-ribose polymerase, release of cytochrome c, and DNA fragmentation were blocked either by a pan-caspase inhibitor (zVAD-fmk) or a CB inhibitor (CA074Me), consistent with the involvement of TRAIL as well as CB in cell death. The primary tumor cells were more susceptible to apoptosis than their corresponding lymph node metastatic cells. Stable transfection of a ribozyme which inhibited CB expression also decreased the apoptotic process. CONCLUSIONS: We conclude that TRAIL-induced apoptotic cell death in OSCC cells is mediated through CB or through caspase activation. Our data point to a new tumor-suppressive role for CB in OSCC which is opposed to the invasion- and metastasis-promoting functions of lysosomal proteases.


Assuntos
Apoptose/fisiologia , Carcinoma de Células Escamosas/patologia , Catepsina B/metabolismo , Neoplasias Bucais/patologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Idoso , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Citometria de Fluxo , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Neoplasias Bucais/metabolismo , Células Tumorais Cultivadas
15.
Life Sci ; 78(8): 898-907, 2006 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-16150465

RESUMO

Cystatins are inhibitors of lysosomal cysteine proteinases. Cystatin M demonstrates more diverse tissue distribution, target specificity and biological function than other cystatins from the same family. We utilized small interference RNAs (siRNA) to silence cystatin M gene expression in a metastatic oral cancer cell line (MDA-686Ln) that expresses a high level of cystatin M. We tested four different siRNAs targeted to different sites of the cystatin M mRNA, and found three out of the four siRNAs were effective in suppressing cystatin M expression by >50% at both mRNA and protein levels, as measured by quantitative real-time RT-PCR and Western blotting. We used siRNA-#1, which demonstrated highest efficiency of silencing cystatin M, to evaluate the phenotypic outcome of silencing cystatin M in MDA-686Ln cells. Cystatin M inhibition significantly increased the enzymatic activities of cathepsins B and L and legumain while reducing cysteine protease inhibitor activity both in the media and intracellularly. MDA-686Ln cells treated with siRNA#1 demonstrated markedly increased proliferation rate, in vitro motility and Matrigel invasiveness. Collectively, our data show that silencing of cystatin M in tumor cells not only increases their invasion and motility via cysteine-proteinase-dependent pathways, but also renders them hyperproliferative through a currently unknown mechanism.


Assuntos
Carcinoma de Células Escamosas/genética , Cistatinas/genética , Cisteína Endopeptidases/genética , Neoplasias Bucais/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/secundário , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cistatina M , Cistatinas/metabolismo , Cisteína Endopeptidases/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Bucais/enzimologia , Neoplasias Bucais/patologia , Invasividade Neoplásica/genética , RNA Interferente Pequeno/administração & dosagem
16.
Toxicol Lett ; 165(2): 182-94, 2006 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-16713138

RESUMO

Our objective is to identify molecular factors which contribute to the increased risk of smokers for oral squamous cell carcinoma (OSCC). In the present study, we investigated the effects of cigarette smoke condensate (CSC) on gene expression profiles in different human oral cell phenotypes: normal epidermal keratinocytes (NHEK), oral dysplasia cell lines (Leuk1 and Leuk2), and a primary oral carcinoma cell line (101A). We determined differential gene expression patterns in CSC-exposed versus non-exposed cells using high-density microarray RNA expression profiling and validation by quantitative real-time RT-PCR. A set of 35 genes was specifically up- or down-regulated following CSC treatment (25microg/ml for 24h) by at least 2-fold in any one cell type. Notably, five genes of the cytochrome P450 (CYP1A1, CYP1B1) and aldo-keto reductase (AKR1C1, AKR1C3, AKR1B10) families were highly increased in expression, some of them 15- to 30-fold. The timing and extent of induction for these genes differed among the four cell phenotypes. A potential biological interaction network for the CSC response in oral cells was derived from these data, proposing novel putative response pathways. These CSC-responsive genes presumably participate in the prevention or repair of carcinogen-induced DNA damage in tobacco-related oral carcinogenesis, and may potentially be exploited for determining the severity of exposure and for correcting mutagenic damage in exposed tissues of the oral cavity.


Assuntos
Oxirredutases do Álcool/genética , Carcinoma de Células Escamosas/genética , Sistema Enzimático do Citocromo P-450/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Neoplasias Bucais/genética , Nicotiana/toxicidade , Fumaça/efeitos adversos , Idoso , Oxirredutases do Álcool/biossíntese , Aldeído Redutase , Aldo-Ceto Redutases , Carcinoma de Células Escamosas/enzimologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/biossíntese , Indução Enzimática/efeitos dos fármacos , Feminino , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/enzimologia , Pessoa de Meia-Idade , Neoplasias Bucais/enzimologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Nucleic Acids Res ; 31(17): 4981-8, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12930947

RESUMO

In yeast, mutations induced by UV radiation are dependent on the function of the Rev1 gene product, a Y-family DNA polymerase that assists in translesion replication with potentially mutagenic consequences. Human REV1 has been cloned, but its role in mutagenesis and carcinogenesis remains obscure. To examine the role of REV1 in UV mutagenesis in human cells and to evaluate its potential as a therapeutic target to prevent such mutations, we developed a ribozyme that cleaves human REV1 mRNA in vitro. Stable expression of the ribozyme in human cells reduced the target REV1 mRNA up to 90%. We examined the cytotoxic and mutagenic response to UV of seven independent clones that had reduced levels of endogenous REV1 mRNA. In each case, the clonogenic survival after UV was not different from that of the parental cell strains. In contrast, the UV-induced mutant frequencies at the endogenous HPRT locus were reduced up to 75% in cells with reduced levels of REV1 mRNA. The data support the idea that targeting the mutagenic translesion DNA replication pathway can greatly reduce the frequency of induced mutations.


Assuntos
Hipoxantina Fosforribosiltransferase/genética , Mutagênese/efeitos da radiação , Nucleotidiltransferases/metabolismo , RNA Catalítico/metabolismo , Sequência de Bases , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Frequência do Gene , Humanos , Masculino , Proteínas Nucleares , Conformação de Ácido Nucleico , Nucleotidiltransferases/genética , RNA Catalítico/química , RNA Catalítico/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Saccharomyces cerevisiae , Raios Ultravioleta
18.
Nucleic Acids Res ; 32(19): 5820-6, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15523096

RESUMO

The REV1 gene encodes a Y-family DNA polymerase that has been postulated to have both catalytic and structural functions in translesion replication past UV photoproducts in mammalian cells. To examine if REV1 is implicated in DNA damage tolerance mechanisms after exposure of human cells to a chemical carcinogen, we generated a plasmid expressing REV1 protein fused at its C-terminus with green fluorescent protein (GFP). In transient transfection experiments, virtually all of the transfected cells had a diffuse nuclear pattern in the absence of carcinogen exposure. In contrast, in cells exposed to benzo[a]pyrenediolepoxide, the fusion protein accumulated in a focal pattern in the nucleus in 25% of the cells, and co-localized with PCNA. These data support the idea that REV1 is present at stalled replication forks. We also examined the mutagenic response at the HPRT locus of human cells that had greatly reduced levels of REV1 mRNA due to the stable expression of gene-specific ribozymes, and compared them to wild-type cells. The mutant frequency was greatly reduced in the ribozyme-expressing cells. These data indicate that REV1 is implicated in the mutagenic DNA damage tolerance response to BPDE and support the development of strategies to target this protein to prevent such mutations.


Assuntos
Núcleo Celular/enzimologia , Dano ao DNA , Mutagênese , Nucleotidiltransferases/análise , Nucleotidiltransferases/fisiologia , 7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/toxicidade , Linhagem Celular , Humanos , Mutagênicos/toxicidade , Proteínas Nucleares , Nucleotidiltransferases/genética , Proteínas Recombinantes de Fusão/análise
19.
Gene ; 344: 67-77, 2005 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-15656974

RESUMO

It has been shown that adenovirus-mediated overexpression of E2F-1 can efficiently induce apoptosis in cancer cells with little effect on normal cells. However, the mechanisms by which E2F-1 induces apoptosis remains poorly understood. The goal of this study was to evaluate changes in gene expression in response to E2F-1 in order to help elucidate the mechanisms by which E2F-1 causes apoptosis. Therefore, we used a quantitative microarray assay to identify the genes regulated by E2F-1 in melanoma cells. By gene expression profiling, we first screened a proprietary list of about 12,000 genes. Overexpression of E2F-1 in melanoma cells resulted in two-fold or greater alteration in the level of expression of 452 genes compared to vehicle-treated control cells. Most of the affected genes were not known to be responsive to E2F-1 prior to this study. E2F-1 adenoviral infection of these cells was found to affect the expression of a diverse range of genes, including oncogenes, transcription factors and genes involved in signal transduction, cell cycle regulation, cell proliferation and apoptosis, as well as other genes with unknown function. Changes in expression of 17 of these genes were confirmed by quantitative real-time polymerase chain reaction (PCR). This is first application of the microarray technique in the study of the global profile of genes regulated by E2F-1 in melanoma cells. This study leads to an increased understanding of the biochemical pathways involved in E2F-1-induced apoptosis and possibly to the identification of new therapeutic targets.


Assuntos
Apoptose/genética , Proteínas de Ciclo Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Perfilação da Expressão Gênica , Fatores de Transcrição/fisiologia , Adenoviridae/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Análise por Conglomerados , Proteínas de Ligação a DNA/genética , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Vetores Genéticos/genética , Humanos , Melanoma/genética , Melanoma/patologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fatores de Transcrição/genética , Transfecção
20.
J Transl Med ; 3: 44, 2005 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-16375766

RESUMO

BACKGROUND: The goal of this study was to evaluate changes in gene expression in SW-620 cells in response to SN-38 in order to further elucidate the mechanisms by which SN-38 causes apoptosis and cell cycle arrest. METHODS: We used a quantitative gene expression microarray assay to identify the genes regulated by SN-38 treatment in colon cancer cells and confirmed our results with RT-PCR. By gene expression profiling, we first screened a proprietary list of about 22,000 genes. RESULTS: Treatment with SN-38 cells resulted in two-fold or greater alteration in the level of expression of 192 genes compared to control treatment. Most of the affected genes were not known to be responsive to SN-38 prior to this study. SN-38 treatment of these cells was found to affect the expression of various genes involved in DNA replication, transcription, signal transduction, growth factors, cell cycle regulation, and apoptosis, as well as other genes with unknown function. Changes in expression of 14 genes were confirmed by quantitative real-time polymerase chain reaction (RT-PCR). CONCLUSION: This study leads to an increased understanding of the biochemical pathways involved in SN-38-induced apoptosis and possibly to the identification of new therapeutic targets.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA