Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 622
Filtrar
1.
J Virol ; 95(24): e0139921, 2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34586865

RESUMO

Targeting host factors is a promising strategy to develop broad-spectrum antiviral drugs. Drugs targeting anti-apoptotic Bcl-2 family proteins that were originally developed as tumor suppressors have been reported to inhibit multiplication of different types of viruses. However, the mechanisms whereby Bcl-2 inhibitors exert their antiviral activity remain poorly understood. In this study, we have investigated the mechanisms by which obatoclax (OLX) and ABT-737 Bcl-2 inhibitors exhibited a potent antiviral activity against the mammarenavirus lymphocytic choriomeningitis virus (LCMV). OLX and ABT-737 potent anti-LCMV activity was not associated with their proapoptotic properties but rather with their ability to induce cell arrest at the G0/G1 phase. OLX- and ABT-737-mediated inhibition of Bcl-2 correlated with reduced expression levels of thymidine kinase 1 (TK1), cyclin A2 (CCNA2), and cyclin B1 (CCNB1) cell cycle regulators. In addition, small interfering RNA (siRNA)-mediated knockdown of TK1, CCNA2, and CCNB1 resulted in reduced levels of LCMV multiplication. The antiviral activity exerted by Bcl-2 inhibitors correlated with reduced levels of viral RNA synthesis at early times of infection. Importantly, ABT-737 exhibited moderate efficacy in a mouse model of LCMV infection, and Bcl-2 inhibitors displayed broad-spectrum antiviral activities against different mammarenaviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our results suggest that Bcl-2 inhibitors, actively being explored as anticancer therapeutics, might be repositioned as broad-spectrum antivirals. IMPORTANCE Antiapoptotic Bcl-2 inhibitors have been shown to exert potent antiviral activities against various types of viruses via mechanisms that are currently poorly understood. This study has revealed that Bcl-2 inhibitors' mediation of cell cycle arrest at the G0/G1 phase, rather than their proapoptotic activity, plays a critical role in blocking mammarenavirus multiplication in cultured cells. In addition, we show that Bcl-2 inhibitor ABT-737 exhibited moderate antimammarenavirus activity in vivo and that Bcl-2 inhibitors displayed broad-spectrum antiviral activities against different mammarenaviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our results suggest that Bcl-2 inhibitors, actively being explored as anticancer therapeutics, might be repositioned as broad-spectrum antivirals.


Assuntos
Apoptose , Arenaviridae/efeitos dos fármacos , Tratamento Farmacológico da COVID-19 , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células A549 , Animais , Antivirais/farmacologia , Proteínas Reguladoras de Apoptose/farmacologia , Compostos de Bifenilo/farmacologia , COVID-19/virologia , Ciclo Celular , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/virologia , Chlorocebus aethiops , Ciclina A2/biossíntese , Ciclina B1/biossíntese , Fase G1 , Humanos , Indóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Nitrofenóis/farmacologia , Piperazinas/farmacologia , Pirróis/farmacologia , Fase de Repouso do Ciclo Celular , SARS-CoV-2 , Sulfonamidas/farmacologia , Timidina Quinase/biossíntese , Células Vero
2.
PLoS One ; 16(8): e0253008, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34370752

RESUMO

Glioblastoma is a malignant brain tumor with poor prognosis that rapidly acquires resistance to available clinical treatments. The herpes simplex virus thymidine kinase/ganciclovir (HSVtk/GCV) system produces the selective elimination of HSVtk-positive cells and is a candidate for preclinical testing against glioblastoma via its ability to regulate proliferation and differentiation. Therefore, in this study, we aimed to establish a plasmid encoding the HSVtk/GCV system driven by a glial fibrillary acidic protein (GFAP) promoter and verify its possibility of neural differentiation of glioblastoma cell line under the GCV challenge. Four stable clones-N2A-pCMV-HSVtk, N2A-pGFAP-HSVtk, U251-pCMV-HSVtk, and U251-pGFAP-HSVtk-were established from neuronal N2A and glioblastoma U251 cell lines. In vitro GCV sensitivity was assessed by MTT assay for monitoring time- and dosage-dependent cytotoxicity. The capability for neural differentiation in stable glioblastoma clones during GCV treatment was assessed by performing immunocytochemistry for nestin, GFAP, and ßIII-tubulin. Under GFAP promoter control, the U251 stable clone exhibited GCV sensitivity, while the neuronal N2A clones were nonreactive. During GCV treatment, cells underwent apoptosis on day 3 and dying cells were identified after day 5. Nestin was increasingly expressed in surviving cells, indicating that the population of neural stem-like cells was enriched. Lower levels of GFAP expression were detected in surviving cells. Furthermore, ßIII-tubulin-positive neuron-like cells were identified after GCV treatment. This study established pGFAP-HSVtk-P2A-EGFP plasmids that successfully ablated GFAP-positive glioblastoma cells, but left neuronal N2A cells intact. These data suggest that the neural differentiation of glioblastoma cells can be promoted by treatment with the HSVtk/GCV system.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Ganciclovir/farmacologia , Proteína Glial Fibrilar Ácida/genética , Glioblastoma/metabolismo , Proteínas de Neoplasias/genética , Simplexvirus/genética , Timidina Quinase , Proteínas Virais , Animais , Diferenciação Celular/genética , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Proteína Glial Fibrilar Ácida/metabolismo , Glioblastoma/genética , Glioblastoma/terapia , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/metabolismo , Simplexvirus/enzimologia , Timidina Quinase/antagonistas & inibidores , Timidina Quinase/biossíntese , Timidina Quinase/genética , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/biossíntese , Proteínas Virais/genética
3.
Nanomedicine ; 23: 102115, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31655205

RESUMO

Together, medulloblastoma (MB) and atypical teratoid/rhabdoid tumors (AT/RT) represent two of the most prevalent pediatric brain malignancies. Current treatment involves radiation, which has high risks of developmental sequelae for patients under the age of three. New safer and more effective treatment modalities are needed. Cancer gene therapy is a promising alternative, but there are challenges with using viruses in pediatric patients. We developed a library of poly(beta-amino ester) (PBAE) nanoparticles and evaluated their efficacy for plasmid delivery of a suicide gene therapy to pediatric brain cancer models-specifically herpes simplex virus type I thymidine kinase (HSVtk), which results in controlled apoptosis of transfected cells. In vivo, PBAE-HSVtk treated groups had a greater median overall survival in mice implanted with AT/RT (P = 0.0083 vs. control) and MB (P < 0.0001 vs. control). Our data provide proof of principle for using biodegradable PBAE nanoparticles as a safe and effective nanomedicine for treating pediatric CNS malignancies.


Assuntos
Neoplasias Encefálicas , Terapia Genética , Herpesvirus Humano 1 , Nanopartículas , Timidina Quinase , Proteínas Virais , Animais , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Criança , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Nanopartículas/química , Nanopartículas/uso terapêutico , Timidina Quinase/biossíntese , Timidina Quinase/genética , Proteínas Virais/biossíntese , Proteínas Virais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Tumour Biol ; 39(6): 1010428317706479, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28651488

RESUMO

Cancer is a disease with abnormally proliferating cells and therefore proliferation rate is an important index for assessing tumour growth. Ki-67 is a commonly used proliferation marker considered to be an unfavourable prognostic marker in some tumors, while Thymidine kinase 1 (TK1) is an interesting proliferation marker because its levels are highly dependent on the growth stage of cells. To define the immunohistochemistry (IHC) expression of the TK1 in patients with ovarian serous adenocarcinoma and establish its potential role as a new biomarker for progressive disease, we analyzed the expression patterns of TK1 and Ki-67 in 109 patients with ovarian serous adenocarcinoma. TK1 and Ki-67 expression both showed a statistically significant correlation to MD Anderson Cancer Center (MDACC) grade, but not to age, tumour size, lymph node metastasis or pathological TNM (pTNM) stages. TK1 expression, MDACC grades, pathological stages and lymph node metastasis correlate to relapse incident rate and overall survival, but Ki-67 does not. Although TK1 expression, MDACC grade, pTNM stage and lymph node metastasis significantly correlate to relapse in the Cox univariate analysis, in the multivariate Cox analysis only TK1 expression and lymph node metastasis were independent prognostic factors. The overall survival also correlated significantly to TK1 expression, MDACC grade, pTNM stage and lymph node metastasis in the Cox univariate analysis. However, only the pTNM stage was found to be an independent prognostic factor for survival in the Cox multivariate analysis. Therefore, though TK1 expression was an independent prognostic factor for relapse, but not for survival, TK1 is a more informative expression than Ki-67 for LI, relapse and overall survival rates. Thus, when TK1 is combined with MDACC grading, pTNM staging and lymph node metastasis, IHC determination of TK1 expression may improve the overall prediction of prognosis in patients with ovarian cancer.


Assuntos
Adenocarcinoma/genética , Biomarcadores Tumorais/biossíntese , Antígeno Ki-67/genética , Neoplasias Ovarianas/genética , Timidina Quinase/biossíntese , Adenocarcinoma/patologia , Adulto , Idoso , Biomarcadores Tumorais/genética , Proliferação de Células/genética , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Antígeno Ki-67/biossíntese , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Ovarianas/patologia , Prognóstico , Modelos de Riscos Proporcionais , Timidina Quinase/genética
5.
Tumour Biol ; 39(3): 1010428317695961, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28349820

RESUMO

Recurrence of breast cancer after radiotherapy may be partly explained by the presence of radioresistant cells. Thus, it would be desirable to develop an effective therapy against radioresistant cells. In this study, we demonstrated the intense antitumor activity of cytokine-induced killer cells against MCF-7 and radioresistant MCF-7 cells, as revealed by cytokine-induced killer-mediated cytotoxicity, tumor cell proliferation, and tumor invasion. Radioresistant MCF-7 cells were more susceptible to cytokine-induced killer cell killing. The stronger cytotoxicity of cytokine-induced killer cells against radioresistant MCF-7 cells was dependent on the expression of major histocompatibility complex class I polypeptide-related sequence A/B on radioresistant MCF-7 cells after exposure of cytokine-induced killer cells to sensitized targets. In addition, we demonstrated that cytokine-induced killer cell treatment sensitized breast cancer cells to chemotherapy via the downregulation of TK1, TYMS, and MDR1. These results indicate that cytokine-induced killer cell treatment in combination with radiotherapy and/or chemotherapy may induce synergistic antitumor activities and represent a novel strategy for breast cancer.


Assuntos
Neoplasias da Mama/radioterapia , Terapia Baseada em Transplante de Células e Tecidos , Células Matadoras Induzidas por Citocinas/metabolismo , Recidiva Local de Neoplasia/radioterapia , Subfamília B de Transportador de Cassetes de Ligação de ATP/biossíntese , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Células Matadoras Induzidas por Citocinas/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos da radiação , Feminino , Humanos , Células MCF-7 , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia , Tolerância a Radiação , Timidina Quinase/biossíntese , Timidilato Sintase/biossíntese
6.
Biochim Biophys Acta Gene Regul Mech ; 1860(3): 374-382, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28093273

RESUMO

FHIT is a genome caretaker gene that is silenced in >50% of cancers. Loss of Fhit protein expression promotes accumulation of DNA damage, affects apoptosis and epithelial-mesenchymal transition, though molecular mechanisms underlying these alterations have not been fully elucidated. Initiation of genome instability directly follows Fhit loss and the associated reduced Thymidine Kinase 1 (TK1) protein expression. The effects on TK1 of Fhit knockdown and Fhit induction in the current study confirmed the role of Fhit in regulating TK1 expression. Changes in Fhit expression did not impact TK1 protein turnover or transcription from the TK1 promoter, nor steady-state levels of TK1 mRNA or turnover. Polysome profile analysis showed that up-regulated Fhit expression resulted in decreased TK1 RNA in non-translating messenger ribonucleoproteins and increased ribosome density on TK1 mRNA. Fhit does not bind RNA but its expression increased luciferase expression from a transgene bearing the TK1 5'-UTR. Fhit has been reported to act as a scavenger decapping enzyme, and a similar result with a mutant (H96) that binds but does not cleave nucleoside 5',5'-triphosphates suggests the impact on TK1 translation is due to its ability to modulate the intracellular level of cap-like molecules. Consistent with this, cells expressing Fhit mutants with reduced activity toward cap-like dinucleotides exhibit DNA damage resulting from TK1 deficiency, whereas cells expressing wild-type Fhit or the H96N mutant do not. The results have implications for the mechanism by which Fhit regulates TK1 mRNA, and more broadly, for its modulation of multiple functions as tumor suppressor/genome caretaker.


Assuntos
Hidrolases Anidrido Ácido/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Proteínas de Neoplasias/metabolismo , Regiões Promotoras Genéticas/fisiologia , Timidina Quinase/biossíntese , Hidrolases Anidrido Ácido/genética , Substituição de Aminoácidos , Linhagem Celular Tumoral , Humanos , Mutação de Sentido Incorreto , Proteínas de Neoplasias/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Timidina Quinase/genética
7.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 691-698, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27906616

RESUMO

Tomato thymidine kinase 1 (ToTK1) is a deoxyribonucleoside kinase (dNK) that has been subject to study because of its potential to phosphorylate the nucleoside analogue 3-azido-2,3-dideoxythymidine (azidothymidine, AZT) equally well as its natural substrate thymidine (dThd). The combination of ToTK1 and AZT has been tested in two animal studies for its efficiency and use in suicide gene therapy for malignant glioma. The determination of the 3D structure of ToTK1 might shed light on the structure-function relationships of nucleoside activation by this enzyme and thereby show routes toward further improvement of ToTK1 and other TK1-like dNKs for suicide gene therapy. Here we report the successful expression of both full-length ToTK1 and a C-terminal truncated ToTK1 in Spodoptera frugiperda and Trichoplusia ni insect cells using the baculovirus expression vector system. This constitutes a further step on the road to determine the 3D structure of the first TK1 of plant origin, but also an enzyme with great potential for dNK-mediated suicide gene therapy.


Assuntos
Proteínas de Plantas/genética , Solanum lycopersicum/enzimologia , Timidina Quinase/genética , Animais , Baculoviridae/genética , Expressão Gênica , Vetores Genéticos , Proteínas de Plantas/biossíntese , Células Sf9 , Spodoptera , Timidina Quinase/biossíntese
8.
Oncotarget ; 7(40): 65042-65051, 2016 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-27542255

RESUMO

OBJECTIVE: Gene therapy is a frontier in modern medicine. In the present study, we explored a new technique for the effective treatment of multidrug-resistant (MDR) breast cancer by combining fully the advantages of multidisciplinary fields, including image-guided minimally invasive interventional oncology, radiofrequency technology, and direct intratumoral gene therapy. RESULTS: Combination treatment with PHSP-TK plus RFH resulted in significantly higher TK gene transfection/expression, as well as a lower cell proliferation rate and a higher cell apoptosis index, than those of control groups. In vivo validation experiments with MRI confirmed that combination therapy resulted in a significant reduction of relative tumor volume compared with those of control animals, which was supported by the results of histologic and apoptosis analyses. MATERIALS AND METHODS: The heat shock protein promoter (PHSP) was used to precisely control the overexpression of thymidine kinase (TK) (PHSP-TK). Serial in vitro experiments were performed to confirm whether radiofrequency hyperthermia (RFH) could enhance PHSP-TK transfection and expression in a MDR breast cancer cell line (MCF7/Adr). Serial in vivo experiments were then carried out to validate the feasibility of the new technique, termed interventional RFH-enhanced direct intratumoral PHSP-TK gene therapy. The therapeutic effect of combination therapy was evaluated by MRI and confirmed by subsequent laboratory correlation. CONCLUSIONS: This study has established "proof-of-principle" of a new technique, interventional RFH-enhanced local gene therapy for MDR breast cancer, which may open new avenues for the effective management of MDR breast cancers via the simultaneous integration of interventional oncology, RF technology, and direct intratumoral gene therapy.


Assuntos
Neoplasias da Mama , Terapia Genética/métodos , Hipertermia Induzida/métodos , Timidina Quinase/genética , Transfecção/métodos , Animais , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Feminino , Genes Transgênicos Suicidas , Proteínas de Choque Térmico/genética , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Regiões Promotoras Genéticas , Timidina Quinase/administração & dosagem , Timidina Quinase/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Nucl Med ; 57(7): 1136-40, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27081170

RESUMO

UNLABELLED: The radiotracer 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) is commonly used to measure cell proliferation in vivo. As a marker of cell proliferation, (18)F-FLT is expected to be differentially taken up by arrested and actively dividing cells, but PET measures only aggregate uptake by tumor cells and therefore the single-cell distribution of (18)F-FLT is unknown. We used a novel in vitro radioluminescence microscopy technique to measure the differential distribution of (18)F-FLT radiotracer with single-cell precision. METHODS: Using radioluminescence microscopy, we imaged the absolute uptake of (18)F-FLT in live MDA-MB-231 cells grown under different serum conditions. We then compared (18)F-FLT uptake with a standard measure of cell proliferation, using fluorescence microscopy of 5-ethynyl-2'-deoxyuridine incorporation in fixed cells. RESULTS: According to 5-ethynyl-2'-deoxyuridine staining, few cells (1%) actively cycled under serum deprivation whereas most of them (71%) did under 20% serum. The distribution of (18)F-FLT reflected this dynamic. At 0% serum, uptake of (18)F-FLT was heterogeneous but relatively low. At 20% serum, a subpopulation of (18)F-FLT-avid cells, representing 61% of the total population, emerged. Uptake of (18)F-FLT in this population was 5-fold higher than in the remainder of the cells. Such a dichotomous distribution is not typically observed with other radiotracers, such as (18)F-FDG. CONCLUSION: These results suggest that increased (18)F-FLT uptake by proliferating cells is due to a greater fraction of (18)F-FLT-avid cells rather than a change in (18)F-FLT uptake by individual cells. This finding is consistent with the fact that (18)F-FLT uptake is mediated by thymidine kinase 1 expression, which is higher in actively dividing cells. Overall, these findings suggest that, within the same patient, changes in (18)F-FLT uptake reflect changes in the number of actively dividing cells, provided other parameters remain the same.


Assuntos
Didesoxinucleosídeos/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Análise de Célula Única , Linhagem Celular Tumoral , Proliferação de Células , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Microscopia/métodos , Tomografia por Emissão de Pósitrons , Timidina Quinase/biossíntese , Timidina Quinase/metabolismo
10.
Oncotarget ; 7(14): 18896-905, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26934443

RESUMO

Suicide gene therapy using herpes simplex virus-1 thymidine kinase (HSV-TK) in combination with ganciclovir (GCV) has emerged as a potential new method for treating cancer. We hypothesize that the efficacy of HSV-TK/GCV therapy is at least partially dependent on p53 status in hepatocellular carcinoma (HCC) patients. Using recombinant adenoviral vectors (rAdV), TK, p53, and ASPP2 were overexpressed individually and in combination in Hep3B (p53 null) and HepG2 (p53 wild-type) cell lines and in primary HCC tumor cells. p53 overexpression induced death in Hep3B cells, but not HepG2 cells. ASPP2 overexpression increased rAdV-TK/GCV-induced HepG2 cell death by interacting with endogenous p53. Similarly, ASPP2 reduced survival in rAdV-TK/GCV-treated primary HCC cells expressing p53 wild-type but not a p53 R249S mutant. Mutated p53 was unable to bind to ASPP2, suggesting that the increase in rAdV-TK/GCV-induced cell death resulting from ASPP2 overexpression was dependent on its interaction with p53. Additionally, γ-H2AX foci, ATM phosphorylation, Bax, and p21 expression increased in rAdV-TK/GCV-treated HepG2 cells as compared to Hep3B cells. This suggests that the combined use of HSV-TK, GCV, rAdV-p53 and rAdV-ASPP2 may improve therapeutic efficacy in HCC patients lacking functional p53.


Assuntos
Proteínas Reguladoras de Apoptose/administração & dosagem , Carcinoma Hepatocelular/terapia , Ganciclovir/farmacologia , Terapia Genética/métodos , Neoplasias Hepáticas/terapia , Proteína Supressora de Tumor p53/administração & dosagem , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Reguladoras de Apoptose/genética , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Morte Celular , Linhagem Celular Tumoral , Terapia Combinada , Genes Transgênicos Suicidas , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/biossíntese , Timidina Quinase/genética , Timidina Quinase/metabolismo , Transfecção , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
11.
Oncotarget ; 6(31): 31018-29, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26427042

RESUMO

The constant presence of the viral genome in Epstein-Barr virus (EBV)-associated gastric cancers (EBVaGCs) suggests the applicability of novel EBV-targeted therapies. The antiviral nucleoside drug, ganciclovir (GCV), is effective only in the context of the viral lytic cycle in the presence of EBV-encoded thymidine kinase (TK)/protein kinase (PK) expression. In this study, screening of the Johns Hopkins Drug Library identified gemcitabine as a candidate for combination treatment with GCV. Pharmacological induction of EBV-TK or PK in EBVaGC-originated tumor cells were used to study combination treatment with GCV in vitro and in vivo. Gemcitabine was found to be a lytic inducer via activation of the ataxia telangiectasia-mutated (ATM)/p53 genotoxic stress pathway in EBVaGC. Using an EBVaGC mouse model and a [125I] fialuridine (FIAU)-based lytic activation imaging system, we evaluated gemcitabine-induced lytic activation in an in vivo system and confirmed the efficacy of gemcitabine-GCV combination treatment. This viral enzyme-targeted anti-tumor strategy may provide a new therapeutic approach for EBVaGCs.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Antivirais/farmacologia , Carcinoma/tratamento farmacológico , Desoxicitidina/análogos & derivados , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Ganciclovir/farmacologia , Herpesvirus Humano 4/efeitos dos fármacos , Terapia de Alvo Molecular , Neoplasias Gástricas/tratamento farmacológico , Animais , Carcinoma/diagnóstico , Carcinoma/genética , Carcinoma/virologia , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Reposicionamento de Medicamentos , Indução Enzimática , Infecções por Vírus Epstein-Barr/diagnóstico , Infecções por Vírus Epstein-Barr/virologia , Feminino , Herpesvirus Humano 4/enzimologia , Herpesvirus Humano 4/patogenicidade , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Quinases/biossíntese , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética , Neoplasias Gástricas/virologia , Timidina Quinase/biossíntese , Fatores de Tempo , Transfecção , Carga Tumoral/efeitos dos fármacos , Proteínas Virais/biossíntese , Ativação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
12.
Urol Oncol ; 33(10): 426.e21-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26231311

RESUMO

OBJECTIVE: Thymidine kinases have an important role in the synthesis of DNA and exhibit high activity in rapidly proliferating cells. Thymidine kinase 1 (TK1) activity has been shown to be increased in various cancer types and proposed as a prognostic parameter. Aim of the present study was to investigate TK1 in muscle-invasive urothelial carcinoma (UC). METHODS: Corresponding UC and benign samples from paraffin embedded tissue of 111 patients treated with cystectomy for invasive UC from 1996 to 2006 were immunohistochemically (IHC) assessed for TK1. IHC expression patterns were evaluated in a semiquantitative fashion by 2 independent reviewers. Localization of staining was categorized into pure nuclear and additional cytoplasmic localization. Uni- and multivariate analyses were performed to assess differential expression in normal and UC tissue and to evaluate the diagnostic and predictive capability of TK1 by correlation to clinical data. To correlate TK1 expression with molecular subtypes of UC, analysis of TK1 RNA expression levels of the Cancer Genome Atlas UC cohort was performed. RESULTS: TK1 was significantly overexpressed in invasive UC, compared to benign urothelium (P<0.0001), and cytoplasmic expression was more often found in cancer tissue than in benign tissue (P = 0.0001). No correlations of TK1 protein expression patterns to standard histopathological determinants were detected. In univariate analysis, TK1 nuclear and cytoplasmic expression was associated with improved cancer-specific survival (P = 0.0119). However, only metastasis status and histologic grade were identified as independent predictors of cancer-specific survival in multivariate analysis. TK1 expression was merely found in the basal layers of benign urothelium. RNA overexpression of TK1 could be correlated to the biologically more aggressive basal UC subtype. CONCLUSIONS: TK1 expression is significantly different in invasive UC and benign urothelium, which underlines its potential as a diagnostic marker. Although TK1 is considered to be a marker of proliferation, and TK1 RNA overexpression is associated with an aggressive UC subtype, its capability as a predictive IHC biomarker for invasive UC remains limited.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células de Transição/enzimologia , Timidina Quinase/biossíntese , Neoplasias da Bexiga Urinária/enzimologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células de Transição/mortalidade , Carcinoma de Células de Transição/patologia , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Modelos de Riscos Proporcionais , Timidina Quinase/análise , Análise Serial de Tecidos , Neoplasias da Bexiga Urinária/mortalidade , Neoplasias da Bexiga Urinária/patologia
13.
PLoS One ; 10(6): e0128922, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26067671

RESUMO

Glioblastoma (GBM) is an infiltrative tumor that is difficult to eradicate. Treating GBM with mesenchymal stem cells (MSCs) that have been modified with the HSV-Tk suicide gene has brought significant advances mainly because MSCs are chemoattracted to GBM and kill tumor cells via a bystander effect. To use this strategy, abundantly present adipose-tissue-derived mesenchymal stem cells (AT-MSCs) were evaluated for the treatment of GBM in mice. AT-MSCs were prepared using a mechanical protocol to avoid contamination with animal protein and transduced with HSV-Tk via a lentiviral vector. The U-87 glioblastoma cells cultured with AT-MSC-HSV-Tk died in the presence of 25 or 50 µM ganciclovir (GCV). U-87 glioblastoma cells injected into the brains of nude mice generated tumors larger than 3.5 mm2 after 4 weeks, but the injection of AT-MSC-HSV-Tk cells one week after the U-87 injection, combined with GCV treatment, drastically reduced tumors to smaller than 0.5 mm2. Immunohistochemical analysis of the tumors showed the presence of AT-MSC-HSV-Tk cells only within the tumor and its vicinity, but not in other areas of the brain, showing chemoattraction between them. The abundance of AT-MSCs and the easier to obtain them mechanically are strong advantages when compared to using MSCs from other tissues.


Assuntos
Tecido Adiposo/metabolismo , Glioblastoma/metabolismo , Células-Tronco Mesenquimais/enzimologia , Simplexvirus/genética , Timidina Quinase/biossíntese , Transdução Genética , Proteínas Virais/biossíntese , Tecido Adiposo/patologia , Animais , Efeito Espectador/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Ganciclovir/farmacologia , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Nus , Simplexvirus/enzimologia , Timidina Quinase/genética , Proteínas Virais/genética
14.
BMC Cancer ; 15: 439, 2015 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-26016667

RESUMO

BACKGROUND: Gastric cancer (GC) is one of the major malignant diseases worldwide, especially in Asia, and Japan and Korea have the highest incidence in the world. Because most of the cases that are refractory to therapies die due to peritoneal dissemination (PD) of the cancer cells, controlling PD is important for patient survival. GSDMB is a member of the gasdermin gene family. Because GSDMB is expressed in many types of cancer, including GC, it is likely that the gene contains a regulatory region that is utilized for therapy of occult PD through cancer cell-specific expression of cytotoxic genes. METHODS: We performed reporter assays to identify the regulatory region for the cancer cell-specific expression. We also constructed a lentiviral therapeutic vector that expresses herpes simplex virus thymidine kinase (HSVtk) in a GC cell-specific manner, and tested it in a mouse model of PD. RESULTS: We identified the regulatory region at +496 to +989 from the GSDMB transcription start site and designated it as a GSDMB enhancer. The lentiviral therapeutic vector suppressed proliferation of a GC cell line, 60As6, in vitro in the presence of ganciclovir, and intraperitoneal administration of the vector prolonged the survival term of mice that were intraperitoneally inoculated with 60As6 one week prior to the administration. CONCLUSIONS: The GSDMB-driven HSVtk expression vector had a therapeutic effect on the occult PD model mice. This strategy can potentially be used to treat GC patients with PD.


Assuntos
Terapia Genética , Proteínas de Neoplasias/genética , Neoplasias Peritoneais/terapia , Neoplasias Gástricas/terapia , Timidina Quinase/genética , Animais , Vetores Genéticos/uso terapêutico , Humanos , Camundongos , Cavidade Peritoneal/patologia , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/patologia , Peritônio/patologia , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Sequências Reguladoras de Ácido Nucleico/genética , Simplexvirus/enzimologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Timidina Quinase/biossíntese
15.
Mol Oncol ; 9(6): 1129-39, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25769404

RESUMO

No consensus treatment regime exists beyond surgery for malignant peripheral nerve sheath tumours (MPNST), and the purpose of the present study was to find new approaches to stratify patients with good and poor prognosis and to better guide therapeutic intervention for this aggressive soft tissue cancer. From a total of 67 MPNSTs from Scandinavian patients with and without neurofibromatosis type 1, 30 MPNSTs were investigated by genome-wide RNA expression profiling and 63 MPNSTs by immunohistochemical (IHC) analysis, and selected genes were submitted to analyses of disease-specific survival. The potential drug target genes survivin (BIRC5), thymidine kinase 1 (TK1), and topoisomerase 2-alpha (TOP2A), all encoded on chromosome arm 17q, were up-regulated in MPNST as compared to benign neurofibromas. Each of them was found to be independent prognostic markers on the gene expression level, as well as on the protein level. A prognostic profile was identified by combining the nuclear expression scores of the three proteins. For patients with completely resected tumours only 15% in the high risk group were alive after two years, as compared to 78% in the low risk group. In conclusion, we found a novel protein expression profile which identifies MPNST patients with inferior prognosis even after assumed curative surgery. The tested proteins are drug targets; therefore the expression profile may provide predictive information guiding the design of future clinical trials. Importantly, as the effect is seen on the protein level using IHC, the biomarker panel can be readily implemented in routine clinical testing.


Assuntos
Antígenos de Neoplasias/biossíntese , DNA Topoisomerases Tipo II/biossíntese , Proteínas de Ligação a DNA/biossíntese , Regulação Neoplásica da Expressão Gênica , Proteínas Inibidoras de Apoptose/biossíntese , Proteínas de Neoplasias/biossíntese , Neurilemoma , Timidina Quinase/biossíntese , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Intervalo Livre de Doença , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Pessoa de Meia-Idade , Neurilemoma/metabolismo , Neurilemoma/mortalidade , Neurilemoma/patologia , Neurilemoma/cirurgia , Proteínas de Ligação a Poli-ADP-Ribose , Estudos Retrospectivos , Taxa de Sobrevida , Survivina
16.
Cancer Lett ; 359(2): 206-10, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25596375

RESUMO

PDX1 is overexpressed in pancreatic cancer, and activates the insulin promoter (IP). Adenoviral IP-thymidine kinase and ganciclovir (TK/GCV) suppresses human pancreatic ductal carcinoma (PDAC) in mice, but repeated doses carry significant toxicity. We hypothesized that multiple cycles of liposomal IP-TK/GCV ablate human PDAC in SCID mice with minimal toxicity compared to adenoviral IP-TK/GCV. SCID mice with intraperitoneal human pancreatic cancer PANC-1 tumor implants were given a single cycle of 35 µg iv L-IP-TK, or four cycles of 1, 10, 20, 30, or 35 µg iv L-IP-TK (n = 20 per group), followed by intraperitoneal GCV. Insulin and glucose levels were monitored in mice treated with four cycles of 35 µg iv L-IP-TK. We found that four cycles of 10-35 µg L-IP-TK/GCV ablated more PANC-1 tumor volume compared to a single cycle with 35 µg. Mice that received four cycles of 10 µg L-IP-TK demonstrated the longest survival (P < 0.05), with a median survival of 126 days. In comparison, mice that received a single cycle of 35 µg L-IP-TK/GCV or GCV alone survived a median of 92 days and 68.7 days, respectively. There were no significant changes in glucose or insulin levels following treatment. In conclusion, multiple cycles of liposomal IP-TK/GCV ablate human PDAC in SCID mice with minimal toxicity, suggesting non-viral vectors are superior to adenoviral vectors for IP-gene therapy.


Assuntos
Antivirais/uso terapêutico , Carcinoma Ductal Pancreático/terapia , Ganciclovir/uso terapêutico , Neoplasias Pancreáticas/terapia , Timidina Quinase/genética , Proteínas Virais/genética , Adenoviridae/enzimologia , Animais , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Terapia Genética , Humanos , Insulina/genética , Ilhotas Pancreáticas/patologia , Lipossomos , Masculino , Camundongos SCID , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , Ratos , Timidina Quinase/biossíntese , Transfecção , Carga Tumoral , Proteínas Virais/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Cancer Biother Radiopharm ; 30(1): 8-15, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25545853

RESUMO

Tumor-specific enhancer/promoter is applicable for targeting gene expression in tumors and helpful for tumor-targeting imaging and therapy. We aimed to acquire α-fetoprotein (AFP)-producing hepatocellular carcinoma (HCC) specific images using adenovirus containing HSV1-tk gene controlled by AFP enhancer/promoter and evaluate in vivo ganciclovir (GCV)-medicated therapeutic effects on AFP-targeted HSV1-tk expression with (18)F-FDG positron emission tomography (PET). Recombinant adenovirus expressing HSV1-tk under AFP enhancer/promoter was produced (AdAFP-TK) and the expression levels were evaluated by RT-PCR and (125)I-IVDU uptake. GCV-mediated HSV1-tk cytotoxicity was determined by MTT assay. After the mixture of AdAFP-fLuc and AdAFP-TK was administrated, bioluminescent images (BLIs) and (18)F-FHBG PET images were obtained in tumor-bearing mice. In vivo therapeutic effects of AdAFP-TK and GCV in the HuH-7 xenograft model were monitored by (18)F-FDG PET. When infected with AdAFP-TK, cell viability in HuH-7 was reduced, but those in HT-29 and SK-Hep-1 were not significantly decreased at any GCV concentration less than 100 µM. AFP-targeted fLuc and HSV1-tk expression were clearly visualized by BLI and (18)F-FHBG PET images in AFP-producing HCC, respectively. In vivo GCV-mediated tumor growth inhibition by AFP-targeted HSV1-tk expression was monitored by (18)F-FDG PET. Recombinant AdAFP-TK could be applied for AFP-targeted HCC gene therapy and imaging in AFP-producing HCC.


Assuntos
Carcinoma Hepatocelular/genética , Herpesvirus Humano 1/enzimologia , Neoplasias Hepáticas/metabolismo , Timidina Quinase/biossíntese , alfa-Fetoproteínas/biossíntese , Animais , Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/virologia , Linhagem Celular Tumoral , Feminino , Células HT29 , Células Hep G2 , Herpesvirus Humano 1/genética , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Tomografia por Emissão de Pósitrons , Regiões Promotoras Genéticas , Timidina Quinase/genética , Timidina Quinase/metabolismo , alfa-Fetoproteínas/genética
18.
Sci Rep ; 4: 7403, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25492703

RESUMO

Transgene insertion plays an important role in gene therapy and in biological studies. Transposon-based systems that integrate transgenes by transposase-catalyzed "cut-and-paste" mechanism have emerged as an attractive system for transgenesis. Hyperactive piggyBac transposon is particularly promising due to its ability to integrate large transgenes with high efficiency. However, prolonged expression of transposase can become a potential source of genotoxic effects due to uncontrolled transposition of the integrated transgene from one chromosomal locus to another. In this study we propose a vector design to decrease post-transposition expression of transposase and to eliminate the cells that have residual transposase expression. We design a single plasmid construct that combines the transposase and the transpositioning transgene element to share a single polyA sequence for termination. Consequently, the separation of the transposase element from the polyA sequence after transposition leads to its deactivation. We also co-express Herpes Simplex Virus thymidine kinase (HSV-tk) with the transposase. Therefore, cells having residual transposase expression can be eliminated by the administration of ganciclovir. We demonstrate the utility of this combination transposon system by integrating and expressing a model therapeutic gene, human coagulation Factor IX, in HEK293T cells.


Assuntos
Elementos de DNA Transponíveis , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Cromossomos Humanos/genética , Cromossomos Humanos/metabolismo , Fator IX/biossíntese , Fator IX/genética , Células HEK293 , Humanos , Timidina Quinase/biossíntese , Timidina Quinase/genética , Transposases/biossíntese , Transposases/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
19.
Antimicrob Agents Chemother ; 58(11): 6758-66, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25182642

RESUMO

Mitochondrial thymidine kinase 2 (TK2) and deoxyguanosine kinase (dGK) catalyze the initial phosphorylation of deoxynucleosides in the synthesis of the DNA precursors required for mitochondrial DNA (mtDNA) replication and are essential for mitochondrial function. Antiviral nucleosides are known to cause toxic mitochondrial side effects. Here, we examined the effects of 3'-azido-2',3'-dideoxythymidine (AZT) (zidovudine) on mitochondrial TK2 and dGK levels and found that AZT treatment led to downregulation of mitochondrial TK2 and dGK in U2OS cells, whereas cytosolic deoxycytidine kinase (dCK) and thymidine kinase 1 (TK1) levels were not affected. The AZT effects on mitochondrial TK2 and dGK were similar to those of oxidants (e.g., hydrogen peroxide); therefore, we examined the oxidative effects of AZT. We found a modest increase in cellular reactive oxygen species (ROS) levels in the AZT-treated cells. The addition of uridine to AZT-treated cells reduced ROS levels and protein oxidation and prevented the degradation of mitochondrial TK2 and dGK. In organello studies indicated that the degradation of mitochondrial TK2 and dGK is a mitochondrial event. These results suggest that downregulation of mitochondrial TK2 and dGK may lead to decreased mitochondrial DNA precursor pools and eventually mtDNA depletion, which has significant implications for the regulation of mitochondrial nucleotide biosynthesis and for antiviral therapy using nucleoside analogs.


Assuntos
Mitocôndrias/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Timidina Quinase/biossíntese , Zidovudina/farmacologia , Antimetabólitos/farmacologia , Antimetabólitos/toxicidade , Antivirais/toxicidade , Linhagem Celular Tumoral , Replicação do DNA/genética , DNA Mitocondrial/biossíntese , Regulação para Baixo , Humanos , Mitocôndrias/efeitos dos fármacos , Nucleosídeos/química , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Uridina/farmacologia , Zidovudina/toxicidade
20.
Eur Radiol ; 24(12): 3199-209, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25120206

RESUMO

OBJECTIVE: To examine correlations of (18)F-fluorothymidine (FLT) uptake with pathological tumour size and immunohistochemical Ki-67, and thymidine kinase 1 (TK-1) expressions in primary and metastatic node colorectal cancer foci. METHODS: Thirty primary cancers (PCs) and 37 metastatic nodes (MNs) were included. FLT uptake was assessed by visual scores (non-visible: 0-1 and visible: 2-4), standardized uptake value (SUV), and correlated with size, Ki-67, and TK-1. SUV was measured in visible lesions. FLT heterogeneity was assessed by visual scores (no heterogeneous uptake: 0 and heterogeneous uptake: 1-4). RESULTS: Forty-two lesions were visible. The visible group showed significantly higher values than the non-visible group in size, Ki-67, and TK-1 (each p < 0.05). Size correlated significantly with visual score (PC; ρ = 0.74 and MN; ρ = 0.63), SUVmax (PC; ρ = 0.49, and MN; ρ = 0.76), and SUVmean (PC; ρ = 0.40 and MN; ρ = 0.76) (each p < 0.05). Visual score correlated significantly with size (ρ = 0.86), Ki-67max (ρ = 0.35), Ki-67mean (ρ = 0.38), TK-1max (ρ = 0.35) and TK-1mean (ρ = 0.25) (each p < 0.05). No significant correlations were found between FLT uptake and Ki-67 or TK-1 in 42 visible lesions (each p > 0.05). Heterogeneous FLT uptake was noted in 73 % (22/30) of PCs. CONCLUSION: FLT uptake correlated with size. Heterogeneous FLT distribution in colorectal cancers may be one of the causes of weak or lack of FLT uptake/Ki-67 or TK-1 correlation. KEY POINTS: FLT uptake correlated well with tumour size in colorectal cancer. Weak or lack of FLT uptake/Ki-67 and TK-1 correlations were observed. Immunohistochemical Ki-67 and TK-1 expressions are not always correlated with FLT uptake.


Assuntos
Neoplasias Colorretais/diagnóstico , Didesoxinucleosídeos , Antígeno Ki-67/biossíntese , Linfonodos/metabolismo , Estadiamento de Neoplasias/métodos , Tomografia por Emissão de Pósitrons/métodos , Timidina Quinase/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/biossíntese , Biópsia , Colonoscopia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/secundário , Feminino , Radioisótopos de Flúor , Humanos , Imuno-Histoquímica , Linfonodos/diagnóstico por imagem , Linfonodos/patologia , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA