Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 666
Filtrar
1.
Cancer Sci ; 112(12): 5088-5099, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34609775

RESUMO

Epstein-Barr virus-associated lymphoproliferative disease (EBV-LPD) is frequently fatal. Innate immunity plays a key role in protecting against pathogens and cancers. The stimulator of interferon genes (STING) is regarded as a key adaptor protein allowing DNA sensors recognizing exogenous cytosolic DNA to activate the type I interferon signaling cascade. In terms of EBV tumorigenicity, the role of STING remains elusive. Here we showed that treatment with the STING inhibitor, C-176, suppressed EBV-induced transformation in peripheral blood mononuclear cells. In an EBV-LPD mouse model, C-176 treatment also inhibited tumor formation and prolonged survival. Treatment with B cells alone did not affect EBV transformation, but suppression of EBV-induced transformation was observed in the presence of T cells. Even without direct B cell-T cell contact in a transwell system, the inhibitor reduced the transformation activity, indicating that intercellular communication by humoral factors was critical to prevent EBV-induced transformation. These findings suggest that inhibition of STING signaling pathway with C-176 could be a new therapeutic target of EBV-LPD.


Assuntos
Antineoplásicos/administração & dosagem , Transformação Celular Viral/efeitos dos fármacos , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Linfoma de Células B/prevenção & controle , Proteínas de Membrana/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Infecções por Vírus Epstein-Barr/imunologia , Células HEK293 , Herpesvirus Humano 4 , Humanos , Células Jurkat , Linfoma de Células B/imunologia , Linfoma de Células B/virologia , Camundongos , Análise de Sobrevida , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Virology ; 562: 1-8, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34242747

RESUMO

Bovine leukemia virus (BLV) infection has spread worldwide causing significant economic losses in the livestock industry. In countries with a high prevalence of BLV, minimizing economic losses is challenging; thus, research into various countermeasures is important for improving BLV control. Because anti-BLV drugs have not been developed, the present study explored a promising chemical compound with anti-BLV activity. Initially, screening of a chemical compound library revealed that violaceoid E (vioE), which is isolated from fungus, showed antiviral activity. Further analysis demonstrated that the antiviral effect of vioE inhibited transcriptional activation of BLV. Cellular thermal shift assay and pulldown assays provided evidence for a direct interaction between vioE and the viral transactivator protein, Tax. These data indicate that interference with Tax-dependent transcription could be a novel target for development of anti-BLV drugs. Therefore, it is suggested that vioE is a novel antiviral compound against BLV.


Assuntos
Antivirais/farmacologia , Vírus da Leucemia Bovina/efeitos dos fármacos , Animais , Antivirais/química , Gatos , Bovinos , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Produtos do Gene tax/antagonistas & inibidores , Humanos , Ativação Transcricional/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
3.
Virol J ; 18(1): 18, 2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33441159

RESUMO

Viral oncogenic transformation of healthy cells into a malignant state is a well-established phenomenon but took decades from the discovery of tumor-associated viruses to their accepted and established roles in oncogenesis. Viruses cause ~ 15% of know cancers and represents a significant global health burden. Beyond simply causing cellular transformation into a malignant form, a number of these cancers are augmented by a subset of viral factors that significantly enhance the tumor phenotype and, in some cases, are locked in a state of oncogenic addiction, and substantial research has elucidated the mechanisms in these cancers providing a rationale for targeted inactivation of the viral components as a treatment strategy. In many of these virus-associated cancers, the prognosis remains extremely poor, and novel drug approaches are urgently needed. Unlike non-specific small-molecule drug screens or the broad-acting toxic effects of chemo- and radiation therapy, the age of designer nucleases permits a rational approach to inactivating disease-causing targets, allowing for permanent inactivation of viral elements to inhibit tumorigenesis with growing evidence to support their efficacy in this role. Although many challenges remain for the clinical application of designer nucleases towards viral oncogenes; the uniqueness and clear molecular mechanism of these targets, combined with the distinct advantages of specific and permanent inactivation by nucleases, argues for their development as next-generation treatments for this aggressive group of cancers.


Assuntos
Carcinogênese/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/virologia , Vírus Oncogênicos/genética , Animais , Linhagem Celular , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Viral/efeitos dos fármacos , Transformação Celular Viral/genética , Humanos , Camundongos , Oncogenes , Vírus Oncogênicos/patogenicidade
4.
Blood ; 136(7): 871-884, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32391874

RESUMO

Adult T-cell leukemia-lymphoma (ATL) is an aggressive hematological malignancy of CD4+ T cells transformed by human T-cell lymphotropic virus-1 (HTLV-1). Most HTLV-1-infected individuals are asymptomatic, and only 3% to 5% of carriers develop ATL. Here, we describe the contribution of aberrant DNA methylation to ATL leukemogenesis. HTLV-1-infected T-cells and their uninfected counterparts were separately isolated based on CADM1 and CD7 expression status, and differentially methylated positions (DMPs) specific to HTLV-infected T cells were identified through genome-wide DNA methylation profiling. Accumulation of DNA methylation at hypermethylated DMPs correlated strongly with ATL development and progression. In addition, we identified 22 genes downregulated because of promoter hypermethylation in HTLV-1-infected T cells, including THEMIS, LAIR1, and RNF130, which negatively regulate T-cell receptor (TCR) signaling. Phosphorylation of ZAP-70, a transducer of TCR signaling, was dysregulated in HTLV-1-infected cell lines but was normalized by reexpression of THEMIS. Therefore, we hypothesized that DNA hypermethylation contributes to growth advantages in HTLV-1-infected cells during ATL leukemogenesis. To test this idea, we investigated the anti-ATL activities of OR-1200 and OR-2100 (OR21), novel decitabine (DAC) prodrugs with enhanced oral bioavailability. Both DAC and OR21 inhibited cell growth, accompanied by global DNA hypomethylation, in xenograft tumors established by implantation of HTLV-1-infected cells. OR21 was less hematotoxic than DAC, whereas tumor growth inhibition was almost identical between the 2 compounds, making it suitable for long-term treatment of ATL patient-derived xenograft mice. Our results demonstrate that regional DNA hypermethylation is functionally important for ATL leukemogenesis and an effective therapeutic target.


Assuntos
Antineoplásicos/administração & dosagem , Metilação de DNA/efeitos dos fármacos , Infecções por HTLV-I/tratamento farmacológico , Leucemia-Linfoma de Células T do Adulto/tratamento farmacológico , Piridinas/administração & dosagem , Administração Oral , Adulto , Idoso , Animais , Transformação Celular Viral/efeitos dos fármacos , Transformação Celular Viral/genética , Células Cultivadas , Metilação de DNA/genética , Desmetilação/efeitos dos fármacos , Drogas em Investigação/uso terapêutico , Feminino , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Infecções por HTLV-I/complicações , Infecções por HTLV-I/genética , Vírus Linfotrópico T Tipo 1 Humano/efeitos dos fármacos , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Humanos , Leucemia-Linfoma de Células T do Adulto/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Terapia de Alvo Molecular/métodos , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
5.
Biomolecules ; 9(9)2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31547402

RESUMO

The oncogenic gammaherpesvirus Epstein-Barr virus (EBV) immortalizes in vitro B lymphocytes into lymphoblastoid cell lines (LCLs), a model that gives the opportunity to explore the molecular mechanisms driving viral tumorigenesis. In this study, we addressed the potential of quercetin, a widely distributed flavonoid displaying antioxidant, anti-inflammatory, and anti-cancer properties, in preventing EBV-driven B cell immortalization. The results obtained indicated that quercetin inhibited thectivation of signal transducer and activator of transcription 3 (STAT3) induced by EBV infection and reduced molecules such as interleukin-6 (IL-6) and reactive oxidative species (ROS) known to be essential for the immortalization process. Moreover, we found that quercetin promoted autophagy and counteracted the accumulation of sequestosome1/p62 (SQSTM1/p62), ultimately leading to the prevention of B cell immortalization. These findings suggest that quercetin may have the potential to be used to counteract EBV-driven lymphomagenesis, especially if its stability is improved.


Assuntos
Linfócitos B/citologia , Infecções por Vírus Epstein-Barr/metabolismo , Interleucina-6/metabolismo , Quercetina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Autofagia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/virologia , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Células Cultivadas , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Retroalimentação Fisiológica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteína Sequestossoma-1/metabolismo
6.
Chem Biodivers ; 15(5): e1700560, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29569369

RESUMO

Three new compounds (1 - 3), including two euphane type triterpenes, 24,24-dimethoxy-25,26,27-trinoreuphan-3ß-ol (1) and (24S)-24-hydroperoxyeupha-8,25-dien-3ß-ol (2), and an ent-atisine diterpene, ent-atisane-3α,16α,17-triol (3), were isolated from an acetone extract of the stems of Euphorbia antiquorum, together with eight known diterpenes (4 - 11). The structures of compounds (1 - 11) were elucidated using NMR and MS spectroscopic methods. Compound 7 showed moderate activity against HIV-1 replication in vitro (EC50 = 1.38 µm).


Assuntos
Fármacos Anti-HIV/farmacologia , Euphorbia/química , HIV-1/efeitos dos fármacos , Terpenos/farmacologia , Fármacos Anti-HIV/química , Fármacos Anti-HIV/isolamento & purificação , Linhagem Celular Transformada , Transformação Celular Viral/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Relação Estrutura-Atividade , Terpenos/química , Terpenos/isolamento & purificação , Replicação Viral/efeitos dos fármacos
7.
J Cell Biochem ; 119(6): 4482-4495, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29231260

RESUMO

We aimed to understand whether metformin imposes the inhibitory effect on the HBV-associated tumorigenesis by regulating the HULC and its downstream signaling pathway. Luciferase assay, RT-PCR, and Western-blot, MTT and flow cytometry analysis were performed to understand and the mechanism, by which metformin enhance the inhibitory effect on the HBV-associated tumorigenesis by regulating the HULC and its downstream signaling pathway. HBX promoted viability of three types of cell lines, while metformin inhibited apoptosis of above two cells. ZEB1 was a direct downstream of miR-200a, which was further confirmed that miR-200a reduced luciferase activity of wild-type but not mutant ZEB1 3'UTR, and HULC was bound to region of miR-200a-3p using alignment prediction, but can't affect ZEB1 level. HULC transcription ability, HULC, ZEB1, and p18 levels were much higher in cell treated with HBX, while notably lower in cell treated with metformin, furthermore miR-200a level in cell showed an opposite trend as HULC, ZEB1, and p18 levels. HULC siRNA and miR-200a had no effect on HULC transcription ability, but decreased HULC, ZEB1, and p18 levels, and increased miR-200a expression. HBV (+) HCC +metformin exhibited a higher survival ratio and a lower recurrence rates than HBV (+) HCC group, HBV (-) HCC displayed an even higher survival ratio and an even lower recurrence rates than HBV (+) HCC + metformin groups. This study indicated that metformin imposed inhibitory effect on the HBV-associated HCC by negatively regulating the HULC/p18/miR-200a/ZEB1 signaling pathway.


Assuntos
Carcinoma Hepatocelular/metabolismo , Transformação Celular Viral/efeitos dos fármacos , Vírus da Hepatite B/metabolismo , Hepatite B/metabolismo , Neoplasias Hepáticas/metabolismo , Metformina/farmacologia , Proteínas de Neoplasias/metabolismo , RNA Longo não Codificante/biossíntese , RNA Neoplásico/biossíntese , Transativadores/metabolismo , Proteínas Virais/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/virologia , Feminino , Células Hep G2 , Hepatite B/genética , Hepatite B/patologia , Vírus da Hepatite B/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/virologia , Masculino , Proteínas de Neoplasias/genética , RNA Longo não Codificante/genética , RNA Neoplásico/genética , Transativadores/genética , Proteínas Virais/genética , Proteínas Virais Reguladoras e Acessórias
8.
J Antibiot (Tokyo) ; 70(9): 962-966, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28611469

RESUMO

Kaposi sarcoma herpesvirus (KSHV), also known as human herpesvirus 8, is the causative agent of Kaposi sarcoma; this malignant angiosarcoma is usually treated with conventional antitumor agents that can control disease evolution, but do not clear the latent KSHV episome that binds to cellular DNA. Some commercial antibacterial sulfonamides were tested for the ability to suppress latent KSHV. Quantitative PCR (qPCR) and cytofluorometry assays were used for detecting both viral DNA and the latency factor LANA (latency-associated nuclear antigen) in BC3 cells, respectively. The capacity of sulfonamides to impair MDM2-p53 complex formation was detected by an enzyme-linked immunosorbent assay method. The analysis of variance was performed according to one-way analysis of variance with Fisher as a post hoc test. Here we show that sulfonamide antibiotics are able to suppress the KSHV latent state in permanently infected BC3 lymphoma cells and interfere with the formation of the MDM2-p53 complex that KSHV seemingly needs to support latency and to trigger tumor cell transformation. These findings detected a new molecular target for the activity of sulfonamides and offer a new potential perspective for treating KSHV-induced lymphoproliferative diseases.


Assuntos
Antibacterianos/farmacologia , Antivirais/farmacologia , Herpesvirus Humano 8/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Sulfonamidas/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Antibacterianos/efeitos adversos , Antígenos Virais/metabolismo , Antivirais/efeitos adversos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Células Cultivadas , DNA Viral/metabolismo , Herpesvirus Humano 8/crescimento & desenvolvimento , Herpesvirus Humano 8/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/virologia , Humanos , Concentração Inibidora 50 , Proteínas Nucleares/metabolismo , Multimerização Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/química , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Sulfaguanidina/efeitos adversos , Sulfaguanidina/farmacologia , Sulfametoxazol/efeitos adversos , Sulfametoxazol/farmacologia , Sulfanilamida , Sulfanilamidas/efeitos adversos , Sulfanilamidas/farmacologia , Sulfatiazol , Sulfatiazóis/efeitos adversos , Sulfatiazóis/farmacologia , Sulfonamidas/efeitos adversos , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Oncotarget ; 7(41): 66740-66753, 2016 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-27556514

RESUMO

The Myc/Max/Mad network plays a critical role in cell proliferation, differentiation and apoptosis and c-Myc is overexpressed in many cancers, including HPV-positive cervical cancer cell lines. Despite the tolerance of cervical cancer keratinocytes to high Myc expression, we found that the solitary transduction of the Myc gene into primary cervical and foreskin keratinocytes induced rapid cell death. These findings suggested that the anti-apoptotic activity of E7 in cervical cancer cells might be responsible for negating the apoptotic activity of over-expressed Myc. Indeed, our earlier in vitro studies demonstrated that Myc and E7 synergize in the immortalization of keratinocytes. Since we previously postulated that E7 and the ROCK inhibitor, Y-27632, were members of the same functional pathway in cell immortalization, we tested whether Y-27632 would inhibit apoptosis induced by the over-expression of Myc. Our findings indicate that Y-27632 rapidly inhibited Myc-induced membrane blebbing and cellular apoptosis and, more generally, functioned as an inhibitor of extrinsic and intrinsic pathways of cell death. Most important, Y-27632 cooperated with Myc to immortalize keratinocytes efficiently, indicating that apoptosis is a major barrier to Myc-induced immortalization of keratinocytes. The anti-apoptotic activity of Y-27632 correlated with a reduction in p53 serine 15 phosphorylation and the consequent reduction in the expression of downstream target genes p21 and DAPK1, two genes involved in the induction of cell death.


Assuntos
Amidas/farmacologia , Apoptose/efeitos dos fármacos , Queratinócitos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Piridinas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Apoptose/genética , Transformação Celular Viral/efeitos dos fármacos , Transformação Celular Viral/genética , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas Quinases Associadas com Morte Celular/genética , Proteínas Quinases Associadas com Morte Celular/metabolismo , Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica/métodos , Humanos , Queratinócitos/metabolismo , Queratinócitos/virologia , Masculino , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
10.
PLoS One ; 11(6): e0155909, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27280728

RESUMO

A phenotypic high-throughput cell culture screen was performed to identify compounds that prevented proliferation of the human Papilloma virus type 16 (HPV-16) transformed cell line Ca Ski. A series of quinoxaline compounds exemplified by Compound 1 was identified. Testing against a panel of cell lines demonstrated that Compound 1 selectively inhibited replication of all HPV-16, HPV-18, and HPV-31 transformed cell lines tested with 50% Inhibitory Concentration (IC50) values of 2 to 8 µM relative to IC50 values of 28 to 73 µM in HPV-negative cell lines. Treatment with Compound 1 resulted in a cascade of multiple apoptotic events, including selective activation of effector caspases 3 and 7, fragmentation of cellular DNA, and PARP (poly(ADP-ribose) polymerase) cleavage in HPV-positive cells relative to HPV-negative cells. Unregulated proliferation of HPV transformed cells is dependent on the viral oncogenes, E6 and E7. Treatment with Compound 1 resulted in a decrease in HPV E7 protein in Ca Ski cells. However, the timing of this reduction relative to other effects of compound treatment suggests that this was a consequence, rather than a cause, of the apoptotic cascade. Likewise, compound treatment resulted in no obvious effects on the E6- and E7- mediated down regulation of p53 and Rb, or their downstream effectors, p21 or PCNA. Further investigation of apoptotic signals induced by Compound 1 revealed cleavage of Caspase-8 in HPV-positive cells as early as 2 hours post-treatment, suggesting the compound initiates apoptosis through the extrinsic, death receptor-mediated, pathway of cell death. These studies provide proof of concept that cells transformed by oncogenic Papillomaviruses can be selectively induced to undergo apoptosis by compound treatment.


Assuntos
Apoptose/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Papillomaviridae/efeitos dos fármacos , Infecções por Papillomavirus/patologia , Bibliotecas de Moléculas Pequenas/farmacologia , Neoplasias do Colo do Útero/patologia , Proteínas Reguladoras de Apoptose/metabolismo , Feminino , Humanos , Proteínas E7 de Papillomavirus/metabolismo , Infecções por Papillomavirus/tratamento farmacológico , Infecções por Papillomavirus/virologia , Proteína do Retinoblastoma/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/virologia
12.
Proc Natl Acad Sci U S A ; 113(6): E782-90, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26802124

RESUMO

Epstein-Barr virus (EBV) is an oncogenic herpesvirus that has been causally linked to the development of B-cell and epithelial malignancies. Early after infection, EBV induces a transient period of hyperproliferation that is suppressed by the activation of the DNA damage response and a G1/S-phase growth arrest. This growth arrest prevents long-term outgrowth of the majority of infected cells. We developed a method to isolate and characterize infected cells that arrest after this early burst of proliferation and integrated gene expression and metabolic profiling to gain a better understanding of the pathways that attenuate immortalization. We found that the arrested cells have a reduced level of mitochondrial respiration and a decrease in the expression of genes involved in the TCA cycle and oxidative phosphorylation. Indeed, the growth arrest in early infected cells could be rescued by supplementing the TCA cycle. Arrested cells were characterized by an increase in the expression of p53 pathway gene targets, including sestrins leading to activation of AMPK, a reduction in mTOR signaling, and, consequently, elevated autophagy that was important for cell survival. Autophagy was also critical to maintain early hyperproliferation during metabolic stress. Finally, in assessing the metabolic changes from early infection to long-term outgrowth, we found concomitant increases in glucose import and surface glucose transporter 1 (GLUT1) levels, leading to elevated glycolysis, oxidative phosphorylation, and suppression of basal autophagy. Our study demonstrates that oncogene-induced senescence triggered by a combination of metabolic and genotoxic stress acts as an intrinsic barrier to EBV-mediated transformation.


Assuntos
Linfócitos B/virologia , Transformação Celular Viral , Herpesvirus Humano 4/fisiologia , Estresse Fisiológico , Autofagia/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/patologia , Linfócitos B/ultraestrutura , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Respiração Celular/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Desoxiglucose/farmacologia , Dimetilformamida/farmacologia , Herpesvirus Humano 4/efeitos dos fármacos , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Metabolômica , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Complexos Multiproteicos/metabolismo , Oncogenes , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma/genética , Proteína Supressora de Tumor p53/metabolismo
13.
Bull Math Biol ; 78(1): 4-20, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26676766

RESUMO

The development of cervical cells from normal cells infected by human papillomavirus into invasive cancer cells can be modeled using population dynamics of the cells and free virus. The cell populations are separated into four compartments: susceptible cells, infected cells, precancerous cells and cancer cells. The model system of differential equations also has a free virus compartment in the system, which infect normal cells. We analyze the local stability of the equilibrium points of the model and investigate the parameters, which play an important role in the progression toward invasive cancer. By simulation, we investigate the boundary between initial conditions of solutions, which tend to stable equilibrium point, representing controlled infection, and those which tend to unbounded growth of the cancer cell population. Parameters affected by drug treatment are varied, and their effect on the risk of cancer progression is explored.


Assuntos
Modelos Biológicos , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/virologia , Neoplasias do Colo do Útero/virologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Colo do Útero/patologia , Colo do Útero/virologia , Simulação por Computador , Feminino , Humanos , Conceitos Matemáticos , Infecções por Papillomavirus/tratamento farmacológico , Infecções por Papillomavirus/patologia , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/patologia , Lesões Pré-Cancerosas/virologia , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/prevenção & controle
14.
Monoclon Antib Immunodiagn Immunother ; 34(3): 162-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26090593

RESUMO

During the past few decades, monoclonal antibodies (MAbs) have become an increasingly used tool in diagnostics, therapeutics, and biomedical research. Several methods have been employed to produce MAbs, one of which is the immortalization of B cells by Epstein-Barr virus (EBV). Despite its simplicity, this procedure was never routinely adopted due to its poor efficiency and short-lived antibody (Ab) production. Various adjustments to the basic procedure were introduced, including the addition of certain cytokines and CpG oligodeoxynucleotides, which were shown to improve EBV infectivity and cloning efficiency. The objective of this study was to manipulate culture conditions of the EBV-transformed human lymphocytes, lymphoblastoid cell lines (LCLs), by the timely addition of stimuli including CpG and various interleukins. Such manipulations are aimed at improving LCL proliferative activity and enhancing the cell lines' immortalization potential as well as their Ab production. To accomplish this, IgG(+) B cells were isolated from peripheral blood of a hepatitis B vaccinated, anti-HB Ab-positive volunteer. These cells were infected with EBV and incubated in the presence of CpG DNA 2006 motifs, recombinant human interleukin-2 (rhIL-2), rhIL-4, rhIL-6, and rhIL-21, individually and in combinations. Cells were then restimulated for 2 weeks with the same ILs. The effect of these ILs on anti-HB Ab production and the proliferation of the EBV-transformed lymphocytes were investigated. The current study demonstrates that treatment of LCL cultures with rhIL-2, rh-IL4, rhIL-6, and rhIL-21, individually and in combination, increased to varying degrees the proliferative activity and Ab production of these cells. The addition of IL-4 alone was able to sustain increase in anti-HB Ab despite IL-4 withdrawal. This study suggests that with further optimization ILs can have an enhancing effect on LCL immortalization potential and Ab production capacity.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Linfócitos B/virologia , Transformação Celular Viral/fisiologia , Infecções por Vírus Epstein-Barr/imunologia , Hepatite B/sangue , Herpesvirus Humano 4/fisiologia , Interleucinas/farmacologia , Linhagem Celular Transformada , Proliferação de Células , Transformação Celular Viral/efeitos dos fármacos , Células Cultivadas , Infecções por Vírus Epstein-Barr/patologia , Infecções por Vírus Epstein-Barr/virologia , Humanos
15.
J Pathol ; 237(2): 238-48, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26096068

RESUMO

Non-keratinizing nasopharyngeal carcinoma (NPC) is closely associated with Epstein-Barr virus (EBV) infection. The EBV-encoded latent membrane protein 1 (LMP1) is believed to play an important role in NPC pathogenesis by virtue of its ability to activate multiple cell signalling pathways which collectively promote cell proliferation, transformation, angiogenesis, and invasiveness, as well as modulation of energy metabolism. In this study, we report that LMP1 increases cellular uptake of glucose and glutamine, enhances LDHA activity and lactate production, but reduces pyruvate kinase activity and pyruvate concentrations. LMP1 also increases the phosphorylation of PKM2, LDHA, and FGFR1, as well as the expression of PDHK1, FGFR1, c-Myc, and HIF-1α, regardless of oxygen availability. Collectively, these findings suggest that LMP1 promotes aerobic glycolysis. With respect to FGFR1 signalling, LMP1 not only increases FGFR1 expression, but also up-regulates FGF2, leading to constitutive activation of the FGFR1 signalling pathway. Furthermore, two inhibitors of FGFR1 (PD161570 and SU5402) attenuate LMP1-mediated aerobic glycolysis, cellular transformation (proliferation and anchorage-independent growth), cell migration, and invasion in nasopharyngeal epithelial cells, identifying FGFR1 signalling as a key pathway in LMP1-mediated growth transformation. Immunohistochemical staining revealed that high levels of phosphorylated FGFR1 are common in primary NPC specimens and that this correlated with the expression of LMP1. In addition, FGFR1 inhibitors suppress cell proliferation and anchorage-independent growth of NPC cells. Our current findings demonstrate that LMP1-mediated FGFR1 activation contributes to aerobic glycolysis and transformation of epithelial cells, thereby implicating FGF2/FGFR1 signalling activation in the EBV-driven pathogenesis of NPC.


Assuntos
Transformação Celular Viral , Células Epiteliais/metabolismo , Glicólise , Herpesvirus Humano 4/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Nasofaringe/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Proteínas da Matriz Viral/metabolismo , Carcinoma , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transformação Celular Viral/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/virologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Glicólise/efeitos dos fármacos , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/virologia , Nasofaringe/patologia , Nasofaringe/virologia , Invasividade Neoplásica , Fosforilação , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Proteínas da Matriz Viral/genética
16.
Blood ; 125(16): 2530-43, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25742700

RESUMO

Epigenetic events that are essential drivers of lymphocyte transformation remain incompletely characterized. We used models of Epstein-Barr virus (EBV)-induced B-cell transformation to document the relevance of protein arginine methyltransferase 5 (PRMT5) to regulation of epigenetic-repressive marks during lymphomagenesis. EBV(+) lymphomas and transformed cell lines exhibited abundant expression of PRMT5, a type II PRMT enzyme that promotes transcriptional silencing of target genes by methylating arginine residues on histone tails. PRMT5 expression was limited to EBV-transformed cells, not resting or activated B lymphocytes, validating it as an ideal therapeutic target. We developed a first-in-class, small-molecule PRMT5 inhibitor that blocked EBV-driven B-lymphocyte transformation and survival while leaving normal B cells unaffected. Inhibition of PRMT5 led to lost recruitment of a PRMT5/p65/HDAC3-repressive complex on the miR96 promoter, restored miR96 expression, and PRMT5 downregulation. RNA-sequencing and chromatin immunoprecipitation experiments identified several tumor suppressor genes, including the protein tyrosine phosphatase gene PTPROt, which became silenced during EBV-driven B-cell transformation. Enhanced PTPROt expression following PRMT5 inhibition led to dephosphorylation of kinases that regulate B-cell receptor signaling. We conclude that PRMT5 is critical to EBV-driven B-cell transformation and maintenance of the malignant phenotype, and that PRMT5 inhibition shows promise as a novel therapeutic approach for B-cell lymphomas.


Assuntos
Linfócitos B/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Animais , Linfócitos B/metabolismo , Linfócitos B/virologia , Western Blotting , Linhagem Celular Transformada , Transformação Celular Viral/genética , Células Cultivadas , Herpesvirus Humano 4/fisiologia , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Linfoma/genética , Linfoma/metabolismo , Linfoma/virologia , Camundongos SCID , MicroRNAs/genética , MicroRNAs/metabolismo , Microscopia Confocal , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Interferência de RNA , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Bibliotecas de Moléculas Pequenas/farmacologia , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
17.
Cancer Lett ; 356(2 Pt B): 953-61, 2015 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-25444910

RESUMO

Human papillomavirus (HPV) DNA integrations may affect therapeutic responses in cancers through ATM network-related DNA damage response (DDR). We studied whether cisplatin-induced DDR was altered in human HK-2 renal tubular cells immortalized by HPV16 E6/E7 genes. Cytotoxicity assays utilized thiazolyl blue dye and DDR was identified by gene expression differences, double-strand DNA breaks, ATM promoter activity, and analysis of cell cycling and side population cells. After cisplatin, HK-2 cells showed greater ATM promoter activity indicating activation of this network, but DDR was muted, since little γH2AX was expressed, DNA strand breaks were absent and cells continued cycling. When HK-2 cells were treated with the MDM2 antagonist inducing p53, nutlin-3, or p53 transcriptional activator, tenovin-1, cell growth decreased but cisplatin toxicity was unaffected. By contrast, arsenic trioxide, which by inhibiting wild-type p53-induced phosphatase-1 that serves responses downstream of p53, and by depolymerizing tubulin, synergistically enhanced cisplatin cytotoxicity including loss of SP cells. Our findings demonstrated that HPV16 E6/E7 altered DDR through p53-mediated cell growth controls, which may be overcome by targeting of WIP1 and other processes, and thus should be relevant for treating renal cell carcinoma.


Assuntos
Apoptose/efeitos dos fármacos , Arsenicais/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Transformação Celular Viral/efeitos dos fármacos , Cisplatino/farmacologia , Túbulos Renais/efeitos dos fármacos , Proteínas Oncogênicas Virais/metabolismo , Óxidos/farmacologia , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Repressoras/metabolismo , Antineoplásicos/farmacologia , Trióxido de Arsênio , Proteínas Mutadas de Ataxia Telangiectasia/genética , Western Blotting , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/virologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transformação Celular Viral/genética , Ensaio Cometa , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Sinergismo Farmacológico , Histonas/genética , Histonas/metabolismo , Papillomavirus Humano 16/genética , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/virologia , Túbulos Renais/metabolismo , Túbulos Renais/virologia , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus/genética , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/virologia , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
18.
Blood ; 125(3): 474-82, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25395419

RESUMO

The human T-cell lymphotropic virus type I (HTLV-1) Tax transactivator initiates transformation in adult T-cell leukemia/lymphoma (ATL), a highly aggressive chemotherapy-resistant malignancy. The arsenic/interferon combination, which triggers degradation of the Tax oncoprotein, selectively induces apoptosis of ATL cell lines and has significant clinical activity in Tax-driven murine ATL or human patients. However, the role of Tax loss in ATL response is disputed, and the molecular mechanisms driving degradation remain elusive. Here we demonstrate that ATL-derived or HTLV-1-transformed cells are dependent on continuous Tax expression, suggesting that Tax degradation underlies clinical responses to the arsenic/interferon combination. The latter enforces promyelocytic leukemia protein (PML) nuclear body (NB) formation and partner protein recruitment. In arsenic/interferon-treated HTLV-1 transformed or ATL cells, Tax is recruited onto NBs and undergoes PML-dependent hyper-sumoylation by small ubiquitin-like modifier (SUMO)2/3 but not SUMO1, ubiquitination by RNF4, and proteasome-dependent degradation. Thus, the arsenic/interferon combination clears ATL through degradation of its Tax driver, and this regimen could have broader therapeutic value by promoting degradation of other pathogenic sumoylated proteins.


Assuntos
Arsenicais/farmacologia , Produtos do Gene tax/metabolismo , Interferons/farmacologia , Leucemia-Linfoma de Células T do Adulto/virologia , Proteínas Nucleares/metabolismo , Proteólise/efeitos dos fármacos , Proteína SUMO-1/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Quimioterapia Combinada , Citometria de Fluxo , Imunofluorescência , Células HeLa , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Humanos , Imunoprecipitação , Leucemia-Linfoma de Células T do Adulto/genética , Proteína da Leucemia Promielocítica , Reagentes de Sulfidrila/farmacologia , Sumoilação/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos
19.
Carcinogenesis ; 36(1): 151-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25416558

RESUMO

Interferons (IFNs) have been used to treat epithelial lesions caused by human papillomavirus (HPV) persistence. Here, we exposed primary human keratinocytes (HFKs) immortalized by persistently replicating HPV-16 plasmid genomes to increasing levels of IFN-γ. While untreated HFKs retained replicating HPV-16 plasmids for up to 60-120 population doublings, IFN led to rapid HPV-16 plasmid loss. However, treated cultures eventually gave rise to outgrowth of clones harboring integrated HPV-16 genomes expressing viral E6 and E7 oncogenes from chimeric virus-cell mRNAs similar to those in cervical and head and neck cancers. Surprisingly, every HPV-16 integrant that arose after IFN exposure stemmed from an independent integration event into a different cellular gene locus, even within parallel cultures started from small cell inocula and cultured separately for ≥ 25 doublings to permit the rise and expansion of spontaneous integrants. While IFN treatment conferred a growth advantage upon preexisting integrants added to mixed control cultures, our results indicate that IFN exposure directly or indirectly induces HPV-16 integration, rather than only selects preexisting, spontaneous integrants that appear to be much less frequent. We estimate that IFN exposure increased integration rates by ≥ 100-fold. IFN-induced HPV-16 integration involved a wide range of chromosomal loci with less apparent selection for recurrent insertions near genes involved in cancer-related pathways. We conclude that IFNs and other potential treatments targeting high-risk HPV persistence that disrupt viral genome replication may promote increased high-risk HPV integration as a step in cancer progression. Therapies against high-risk HPV persistence thus need to be evaluated for their integration-inducing potential.


Assuntos
Herança Extracromossômica , Genoma Viral/efeitos dos fármacos , Papillomavirus Humano 16/genética , Interferon gama/farmacologia , Queratinócitos/efeitos dos fármacos , Infecções por Papillomavirus/genética , Plasmídeos/genética , Integração Viral/genética , Antivirais/farmacologia , Transformação Celular Viral/efeitos dos fármacos , Células Cultivadas , DNA Viral/genética , Humanos , Queratinócitos/virologia , Infecções por Papillomavirus/virologia , Reação em Cadeia da Polimerase em Tempo Real
20.
Int J Med Mushrooms ; 16(2): 179-87, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24941039

RESUMO

In this study, we investigated the effects of the aqueous extracts of Lingzhi or Reishi medicinal mushroom, Ganoderma lucidum, obtained from three localities (China; and Morelos and Michoacan, Mexico) on cervical cells transformed by human papillomavirus (HeLa and SiHa) and C-33A cancer cells. The cells were plated in DMEM medium supplemented, and were incubated in the presence of different concentrations of G. lucidum for 24 h. Cell proliferation was determined by MTT colorimetric assay and viability by trypan blue assay. Inhibitory dose was determined (IC50) of the three different extracts of G. lucidum in the culture cell lines mentioned above. The apoptosis process was confirmed by nuclear DNA fragmentation and the cell cycle was determined by flow cytometry. The results showed that aqueous extracts G. lucidum obtained from three localities produced inhibition in the proliferation of VPH transformed cells; they also induced apoptosis and cell cycle arrest in HeLa, SiHa, and C-33A cancer cells. Therefore, it was found that aqueous extracts G. lucidum obtained from three different locations produced inhibitory effect on cancer cells and may have a potential therapeutic use for the prevention and treatment of this disease.


Assuntos
Antivirais/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Papillomavirus Humano 16/efeitos dos fármacos , Papillomavirus Humano 18/efeitos dos fármacos , Infecções por Papillomavirus/virologia , Extratos Vegetais/farmacologia , Reishi/química , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Transformada , Proliferação de Células/efeitos dos fármacos , Transformação Celular Viral/efeitos dos fármacos , Papillomavirus Humano 16/fisiologia , Papillomavirus Humano 18/fisiologia , Humanos , Infecções por Papillomavirus/tratamento farmacológico , Infecções por Papillomavirus/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...