Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Prostate ; 79(9): 994-1006, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31012142

RESUMO

BACKGROUND: Long noncoding RNA (lncRNA) plays a vital role in the development of many diseases. The abnormal expression of lncRNA is closely related to the occurrence and development of different kinds of tumors including prostate cancer (PCa). METHODS: Differentially expressed lncRNA LINC00304 was identified using a publicly available gene expression data set (GSE38241) and quantitative polymerase chain reaction validation. The Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis were used to predict the molecular function of LINC00304. A lncRNA microarray, bioinformatic analysis, and chromatin immunoprecipitation assay were carried out to verify the upstream androgen receptor (AR) signaling pathway. Subsequently, the function of LINC00304 was observed by a series of in vitro assays. RESULTS: We observed higher expression of LINC00304 in PCa cells and samples compared with normal prostate cells and tissues. Functional analysis of LINC00304 showed it was related to regulating cell cycle process, cellular developmental process, and focal adhesion. Further, we identified androgen-inhibited lncRNA, LINC00304 as a direct target of AR. A series of functional studies revealed that overexpression of LINC00304 could significantly promote cell proliferation and cell cycle progression in PCa cells. We also find that LINC00304 can significantly promote CCNA1 expression in PCa cells. CONCLUSIONS: Our results indicate that LINC00304 may represent a new diagnostic and therapeutic biomarker for PCa.


Assuntos
Ciclina A1/biossíntese , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Longo não Codificante/metabolismo , Androgênios/farmacologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Ciclina A1/genética , Humanos , Masculino , Células PC-3 , Neoplasias da Próstata/genética , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Androgênicos/metabolismo , Regulação para Cima
2.
Int J Oncol ; 49(6): 2442-2452, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27748911

RESUMO

Evidence suggests that RING1 and YY1 binding protein (RYBP) functions as a tumor suppressor. However, its role in breast cancer remains unclear. In the present study, the expression of RYBP was assessed in breast cancer patients and cell lines. Disease-free survival durations of breast cancer patients with high RYBP expression were determined based on the ATCG dataset. The effects of RYBP overexpression on cell growth, migration and invasive potency were also assessed. Nude mouse xenograft and lung metastasis models were also used to confirm the role of RYBP. The involvement of SRRM3 in RYBP-mediated breast cancer suppression was explored using SRRM3 siRNA. The potential relationship between RYBP, SRRM3, and REST-003 was examined by qPCR. The results showed that RYBP was downregulated in breast cancer patients and in several breast cancer cell lines. Breast cancer patients with high expression levels of RYBP displayed better disease-free survival. Overexpression of RYBP in MDA-MB-231 and SK-BR-3 cells significantly decreased cell proliferation, migration, and invasion ability, and increased the proportion of cells arrested in S-phase compared with the negative control cells. Additionally, upregulation of proliferation-related cell cycle proteins (cyclin A and cyclin B1) and E-cadherin, and downregulation of snail were observed in RYBP-overexpressing cells. Overexpression of RYBP reduced tumor volume and weight as well as metastatic foci in the lungs of nude mice. SRRM3 knockdown by siRNA, which is downregulated after RYBP overexpression, suppressed cell growth and metastasis in MDA-MB-231 and SK-BR-3 cells. Furthermore, qPCR analysis revealed that REST-003 ncRNA was downregulated in cells overexpressing RYBP and in SRRM3-inhibited cells. Moreover, cell invasion ability and growth were increased after SRRM3 upregulation in RYBP-overexpressing cells, but they were decreased following si-REST-003 transfection. In conclusion, overexpression of RYBP suppresses breast cancer growth and metastasis both in vitro and in vivo. SRRM3 and REST-003, which are downregulated in cells overexpressing RYBP, may be involved in RYBP-mediated breast cancer progression.


Assuntos
Neoplasias da Mama/patologia , Movimento Celular/genética , Proliferação de Células/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas/genética , Proteínas Repressoras/genética , Animais , Neoplasias da Mama/genética , Caderinas/biossíntese , Linhagem Celular Tumoral , Ciclina A1/biossíntese , Ciclina B1/biossíntese , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/secundário , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/genética , Complexo Repressor Polycomb 1/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição da Família Snail/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto , Fator de Transcrição YY1/metabolismo
3.
Tumour Biol ; 37(11): 14863-14872, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27644244

RESUMO

Recent studies have shown that the aqueous, ethanolic extracts and a monomer compound of Paris polyphylla exhibit anticancer activity toward several types of cancer cell lines, but the anticancer activity of (3ß,17α,25R)-spirost-5-ene-3,17-diol 3-O-α-L-rhamnopyranosyl-(1 â†’ 2)-ß-D-glucopyranoside, a monomer isolated from P. polyphylla (PP), named PP-22, has not been reported previously. In this study, we investigated the effect of PP-22 on human tongue squamous cell carcinoma SCC-15 cells in vitro. MTT assays showed that PP-22 inhibited the growth of SCC-15 cells and had no obvious inhibitory effects on human liver L02 cells. Flow cytometry assays showed that the percentages of apoptotic cells were increased. In addition, cleaved caspase-8, cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase (PARP) could be detected by Western blotting. Flow cytometry also showed that PP-22 triggered S and G2/M phases arrest in SCC-15 cells, and on the other hand, the expression of cyclin A, cyclin E2, cyclin B1, phospho-cell division cycle2 (p-cdc2)(Tyr15), p-Wee1, Myt1, and p53 was upregulated. Moreover, p-p38 levels increased, p-extracellular signal-regulated kinase (ERK) levels decreased, and cdc25B expression was inhibited. Furthermore, the p38/mitogen-activated protein kinase (MAPK) inhibitor SB203580 reversed the increase of the expression level of p38, p-cdc2 (Tyr15), cleaved caspase 3, cleaved PARP, p-p53, and p53 and reversed the decrease in cdc25B expression. In conclusion, these results demonstrated that PP-22 activated p38, inhibited cdc25B, increased p-cdc2 (Tyr15), and triggered S and G2/M phase arrest, as well as activated p53 through the p38-p53 pathway, inhibited the MAPK/ERK pathway, activated the caspase 8/caspase 3 pathway, and triggered the extrinsic apoptotic pathway in SCC-15 cells.


Assuntos
Caspase 3/metabolismo , Caspase 8/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Saponinas/farmacologia , Fosfatases cdc25/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Proteína Quinase CDC2 , Carcinoma de Células Escamosas/tratamento farmacológico , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina A1/biossíntese , Ciclina B1/biossíntese , Ciclinas/biossíntese , Proteínas de Ligação a DNA/biossíntese , Humanos , Imidazóis/farmacologia , Melanthiaceae/metabolismo , Proteínas Nucleares , Extratos Vegetais/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Tirosina Quinases , Piridinas/farmacologia , Neoplasias da Língua/tratamento farmacológico , Fatores de Transcrição/biossíntese , Proteína Supressora de Tumor p53/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
4.
Cancer Sci ; 106(10): 1333-40, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26250467

RESUMO

Human papillomavirus (HPV) oncoproteins drive distinctive promoter methylation patterns in cancer. However, the underlying mechanism remains to be elucidated. Cyclin A1 (CCNA1) promoter methylation is strongly associated with HPV-associated cancer. CCNA1 methylation is found in HPV-associated cervical cancers, as well as in head and neck squamous cell cancer. Numerous pieces of evidence suggest that E7 may drive CCNA1 methylation. First, the CCNA1 promoter is methylated in HPV-positive epithelial lesions after transformation. Second, the CCNA1 promoter is methylated at a high level when HPV is integrated into the human genome. Finally, E7 has been shown to interact with DNA methyltransferase 1 (Dnmt1). Here, we sought to determine the mechanism by which E7 increases methylation in cervical cancer by using CCNA1 as a gene model. We investigated whether E7 induces CCNA1 promoter methylation, resulting in the loss of expression. Using both E7 knockdown and overexpression approaches in SiHa and C33a cells, our data showed that CCNA1 promoter methylation decreases with a corresponding increase in expression in E7 siRNA-transfected cells. By contrast, CCNA1 promoter methylation was augmented with a corresponding reduction in expression in E7-overexpressing cells. To confirm whether the binding of the E7-Dnmt1 complex to the CCNA1 promoter induced methylation and loss of expression, ChIP assays were carried out in E7-, del CR3-E7 and vector control-overexpressing C33a cells. The data showed that E7 induced CCNA1 methylation by forming a complex with Dnmt1 at the CCNA1 promoter, resulting in the subsequent reduction of expression in cancers. It is interesting to further explore the genome-wide mechanism of E7 oncoprotein-mediated DNA methylation.


Assuntos
Ciclina A1/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/genética , Papillomavirus Humano 16/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Regiões Promotoras Genéticas/genética , Linhagem Celular Tumoral , Ciclina A1/biossíntese , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA Metiltransferase 3A , Feminino , Papillomavirus Humano 16/genética , Humanos , Dados de Sequência Molecular , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus/genética , Interferência de RNA , RNA Interferente Pequeno , Neoplasias do Colo do Útero/genética , DNA Metiltransferase 3B
5.
Cancer Sci ; 106(10): 1313-22, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26211905

RESUMO

Hematopoietic pre-B cell leukemia transcription factor interacting protein (HPIP) has been shown to play an important role in the development and progression of some cancers. However, the role of HPIP in gastric cancer (GC) is unclear. Here, we show that HPIP is upregulated in most GC patients and promotes GC cell proliferation, migration, and invasion. In GC patients, HPIP positively associates with tumor size and nodal metastasis, and negatively associates with tumor differentiation. Hematopoietic pre-B cell leukemia transcription factor interacting protein increases GC cell proliferation through activation of G1 /S and G2 /M cell cycle transitions, accompanied by a marked increase of the positive cell cycle regulators, including cyclin D1, cyclin A, and cyclin B1. Hematopoietic pre-B cell leukemia transcription factor interacting protein enhances GC cell migration and invasion, and modulates epithelial-mesenchymal transition, which plays a key role in cancer cell migration and invasion. These data underscore the critical role of HPIP in GC cell proliferation and progression and suggest that HPIP inhibition may be a useful therapeutic strategy for GC treatment.


Assuntos
Movimento Celular/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Neoplasias Gástricas/patologia , Linhagem Celular Tumoral , Ciclina A1/biossíntese , Ciclina B1/biossíntese , Ciclina D1/biossíntese , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , RNA Interferente Pequeno
6.
Clin Cancer Res ; 21(6): 1395-405, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25573381

RESUMO

PURPOSE: Although the overall cure rate for pediatric acute lymphoblastic leukemia (ALL) approaches 90%, infants with ALL harboring translocations in the mixed-lineage leukemia (MLL) oncogene (infant MLL-ALL) experience shorter remission duration and lower survival rates (∼50%). Mutations in the p53 tumor-suppressor gene are uncommon in infant MLL-ALL, and drugs that release p53 from inhibitory mechanisms may be beneficial. The purpose of this study was to assess the efficacy of the orally available nutlin, RG7112, against patient-derived MLL-ALL xenografts. EXPERIMENTAL DESIGN: Eight MLL-ALL patient-derived xenografts were established in immune-deficient mice, and their molecular features compared with B-lineage ALL and T-ALL xenografts. The sensitivity of MLL-ALL xenografts to RG7112 was assessed in vitro and in vivo, and the ability of RG7112 to induce p53, cell-cycle arrest, and apoptosis in vivo was evaluated. RESULTS: Gene-expression analysis revealed that MLL-ALL, B-lineage ALL, and T-ALL xenografts clustered according to subtype. Moreover, genes previously reported to be overexpressed in MLL-ALL, including MEIS1, CCNA1, and members of the HOXA family, were significantly upregulated in MLL-ALL xenografts, confirming their ability to recapitulate the clinical disease. Exposure of MLL-ALL xenografts to RG7112 in vivo caused p53 upregulation, cell-cycle arrest, and apoptosis. RG7112 as a single agent induced significant regressions in infant MLL-ALL xenografts. Therapeutic enhancement was observed when RG7112 was assessed using combination treatment with an induction-type regimen (vincristine/dexamethasone/L-asparaginase) against an MLL-ALL xenograft. CONCLUSIONS: The utility of targeting the p53-MDM2 axis in combination with established drugs for the management of infant MLL-ALL warrants further investigation.


Assuntos
Imidazolinas/uso terapêutico , Leucemia Aguda Bifenotípica/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina A1/biossíntese , Feminino , Histona-Lisina N-Metiltransferase/genética , Proteínas de Homeodomínio/biossíntese , Humanos , Lactente , Células Jurkat , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteína Meis1 , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Neoplasias/biossíntese , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Histopathology ; 66(2): 262-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25039670

RESUMO

AIMS: To evaluate the immunoexpression of cyclin A1 in pT1 urothelial carcinomas of the bladder (UC) from a cohort of patients treated by transurethral resection of the bladder (TURB), to determine its value in predicting tumour recurrence, tumour progression, or systemic metastases. METHODS AND RESULTS: Five tissue microarrays (TMAS) were constructed from representative paraffin blocks of high-grade pT1 UC from 149 consecutive patients. Cyclin A1 immunoexpression was evaluated as the percentage of tumour cells with positive nuclear staining estimated at each TMA spot. The cutoff for cyclin A1 positivity was set at 10% of cells. Outcome variables included tumour recurrence and tumour progression as the primary endpoints. Cyclin A1 positivity was associated with tumour progression but not with tumour recurrence or the presence of adjacent carcinoma in situ in the biopsy. Also, patients with pT1b at biopsy and cyclin A1 expression showed higher progression rates than patients with pT1a at biopsy and without cyclin A1 expression, respectively. Combining pT1 stage at biopsy and cyclin A1 expression more accurately predicted tumour progression than pT1 stage at biopsy alone and cyclin A1 expression alone. CONCLUSIONS: Cyclin A1 immunoexpression is of potential utility in predicting disease progression in patients with pT1 UC.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células de Transição/patologia , Ciclina A1/biossíntese , Neoplasias da Bexiga Urinária/patologia , Carcinoma de Células de Transição/mortalidade , Carcinoma de Células de Transição/cirurgia , Ciclina A1/análise , Progressão da Doença , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Estadiamento de Neoplasias , Modelos de Riscos Proporcionais , Análise Serial de Tecidos , Neoplasias da Bexiga Urinária/mortalidade , Neoplasias da Bexiga Urinária/cirurgia , Procedimentos Cirúrgicos Urológicos
8.
Int J Mol Sci ; 14(8): 16600-16, 2013 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-23941782

RESUMO

Industrialisation, the proximity of factories to cities, and human work activities have led to a disproportionate use of substances containing heavy metals, such as cadmium (Cd), which may have deleterious effects on human health. Carcinogenic effects of Cd and its relationship with breast cancer, among other tumours, have been reported. 5-Fluorouracil (5-FU) is a fluoropyrimidine anticancer drug used to treat solid tumours of the colon, breast, stomach, liver, and pancreas. The purpose of this work was to study the effects of Cd on cell cycle, apoptosis, and gene and protein expression in MCF-7 breast cancer cells treated with 5-FU. Cd altered the cell cycle profile, and its effects were greater when used either alone or in combination with 5-FU compared with 5-FU alone. Cd significantly suppressed apoptosis of MCF-7 cells pre-treated with 5-FU. Regarding gene and protein expression, bcl2 expression was mainly upregulated by all treatments involving Cd. The expression of caspase 8 and caspase 9 was decreased by most of the treatments and at all times evaluated. C-myc expression was increased by all treatments involving Cd, especially 5-FU plus Cd at the half time of treatment. Cd plus 5-FU decreased cyclin D1 and increased cyclin A1 expression. In conclusion, our results indicate that exposure to Cd blocks the anticancer effects of 5-FU in MCF-7 cells. These results could have important clinical implications in patients treated with 5-FU-based therapies and who are exposed to high levels of Cd.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Cloreto de Cádmio/farmacologia , Interações Medicamentosas , Fluoruracila/farmacologia , Antimetabólitos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Caspase 8/biossíntese , Caspase 9/biossíntese , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina A1/biossíntese , Ciclina D1/biossíntese , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-myc/biossíntese
9.
Gene ; 513(1): 101-10, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23123729

RESUMO

Bone mass is controlled through a delicate balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption. We show here that RNA editing enzyme adenosine deaminase acting on RNA 1 (ADAR1) is critical for proper control of bone mass. Postnatal conditional knockout of Adar1 (the gene encoding ADAR1) resulted in a severe osteopenic phenotype. Ablation of the Adar1 gene significantly suppressed osteoblast differentiation without affecting osteoclast differentiation in bone. In vitro deletion of the Adar1 gene decreased expression of osteoblast-specific osteocalcin and bone sialoprotein genes, alkaline phosphatase activity, and mineralization, suggesting a direct intrinsic role of ADAR1 in osteoblasts. ADAR1 regulates osteoblast differentiation by, at least in part, modulation of osterix expression, which is essential for bone formation. Further, ablation of the Adar1 gene decreased the proliferation and survival of bone marrow stromal cells and inhibited the differentiation of mesenchymal stem cells towards osteoblast lineage. Finally, shRNA knockdown of the Adar1 gene in MC-4 pre-osteoblasts reduced cyclin D1 and cyclin A1 expression and cell growth. Our results identify ADAR1 as a new key regulator of bone mass and suggest that ADAR1 functions in this process mainly through modulation of the intrinsic properties of osteoblasts (i.e., proliferation, survival and differentiation).


Assuntos
Adenosina Desaminase/metabolismo , Osso e Ossos/metabolismo , Osteoblastos/metabolismo , Osteogênese/fisiologia , Adenosina Desaminase/genética , Fosfatase Alcalina/metabolismo , Animais , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/metabolismo , Proliferação de Células , Sobrevivência Celular/genética , Células Cultivadas , Ciclina A1/biossíntese , Ciclina D1/biossíntese , Inativação Gênica , Sialoproteína de Ligação à Integrina/biossíntese , Masculino , Camundongos , Camundongos Transgênicos , Osteocalcina/biossíntese , Osteogênese/genética , Proteínas de Ligação a RNA , Fator de Transcrição Sp7 , Fatores de Transcrição/biossíntese
10.
J Int Med Res ; 40(2): 537-44, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22613414

RESUMO

OBJECTIVE: To investigate the role of interleukin-18 (IL-18) in regulating the growth of the human tongue squamous cell carcinoma cell line CRL-1623™. METHODS: The human IL18 gene was cloned and transfected into CRL-1623™ cells using the transfection vector pcDNA3.1(+). Investigations included analysis of cell viability, detection of apoptosis using annexin V-fluorescein isothiocyanate, assessment of caspase 3/7 activity and real-time reverse transcription-polymerase chain reaction to assess expression of the IL18, CCND1 (cyclin D(1)), CCNA1 (cyclin A(1)) and IFNG (interferon-γ) genes. RESULTS: Introduction of the IL18 gene inhibited cell proliferation at 24, 48 and 72 h after transfection compared with untransfected cells and cells transfected with blank pcDNA3.1(+) vector. Apoptotic cell numbers and caspase 3/7 activity were significantly enhanced by IL18 transfection. Levels of IL18 and IFNG mRNA were elevated and CCND1 mRNA was reduced after 48 h in IL18 transfected cells compared with wild-type cells. CONCLUSIONS: These findings suggest that IL-18 plays a role in the regulation of tongue squamous cell carcinoma and may represent a potential therapeutic target.


Assuntos
Apoptose , Carcinoma de Células Escamosas/metabolismo , Proliferação de Células , Interleucina-18/metabolismo , Neoplasias da Língua/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Caspase 3/biossíntese , Caspase 7/biossíntese , Linhagem Celular Tumoral , Ciclina A1/biossíntese , Ciclina D1/biossíntese , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos , Humanos , Interferon gama/biossíntese , Interleucina-18/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Neoplasias da Língua/genética , Neoplasias da Língua/patologia , Transfecção
11.
Carcinogenesis ; 33(3): 687-93, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22219178

RESUMO

The alteration of MHC class I (MHC-I) expression is a frequent event during cancer progression, allowing tumor cells to evade the immune system. We report that the loss of one major histocompatibility complex haplotype in human melanoma cells not only allowed them to evade immunosurveillance but also increased their intrinsic oncogenic potential. A second successive defect in MHC-I expression, MHC-I total downregulation, gave rise to melanoma cells that were more oncogenic per se in vivo and showed a higher proliferation rate and greater migratory and invasive potential in vitro. All these processes were reversed by restoring MHC-I expression via human leukocite antigen-A2 gene transfection. MHC-I cell surface expression was inversely correlated with intrinsic oncogenic potential. Modifications in the expression of various cell cycle genes were correlated with changes in MHC-I expression; the most important differences among the melanoma cell lines were in the transcriptional level of AP2-alpha, cyclin A1 and p21WAF1/CIP1. According to these results, altered MHC-I expression in malignant cells can directly increase their intrinsic oncogenic and invasive potential and modulate the expression of cell cycle genes. These findings suggest that human leukocite antigen class I molecules may act directly as tumor suppressor genes in melanoma.


Assuntos
Ciclo Celular/genética , Regulação Neoplásica da Expressão Gênica , Genes MHC Classe I , Genes Supressores de Tumor , Antígenos de Histocompatibilidade Classe I/metabolismo , Melanoma/genética , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Ciclina A1/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Proteínas de Ligação a Ácido Graxo/biossíntese , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Melanoma/imunologia , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Nus , Invasividade Neoplásica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA