Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Cells ; 11(8)2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35455970

RESUMO

The increase in follicular atresia and the decrease in the fecundity of laying hens occur with the aging process. Therefore, the key measure for maintaining high laying performance is to alleviate follicular atresia in the aging poultry. Follicle-stimulating hormone (FSH), as an important pituitary hormone to promote follicle development and maturation, plays an important role in preventing reproductive aging in diverse animals. In this study, the physiological state of the prehierarchical small white follicles (SWFs) and atretic SWFs (ASWFs) were compared, followed by an exploration of the possible capacity of FSH to delay ASWFs' progression in the hens. The results showed that the DNA damage within follicles increased with aging, along with Golgi complex disintegration, cell cycle arrest, increased apoptosis and autophagy in the ASWFs. Subsequently, the ACNU-induced follicular atresia model was established to evaluate the enhancing capacity of FSH on increasing cell proliferation and attenuating apoptosis in ASWFs. FSH inhibited DNA damage and promoted DNA repair by regulating the CHK2/p53 pathway. Furthermore, FSH inhibited CHK2/p53, thus, suppressing the disintegration of the Golgi complex, cell cycle arrest, and increased autophagy in the atretic follicles. Moreover, these effects from FSH treatment in ACNU-induced granulosa cells were similar to the treatment by a DNA repair agent AV-153. These results indicate that FSH protects aging-resulted DNA damage in granulosa cells by inhibiting CHK2/p53 in chicken prehierarchical follicles.


Assuntos
Hormônio Foliculoestimulante , Atresia Folicular , Animais , Galinhas/metabolismo , Dano ao DNA , Feminino , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/metabolismo , Nimustina/metabolismo , Nimustina/farmacologia , Proteína Supressora de Tumor p53/metabolismo
2.
Exp Dermatol ; 27(1): 64-70, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28833504

RESUMO

Together with regulatory T cells (Tregs), tumor-associated macrophages (TAMs) play roles in maintaining the tumor microenvironment. Although cytotoxic antimelanoma drugs such as dacarbazine (DTIC), nimustine hydrochloride (ACNU) and vincristine (VCR) have been used for the treatment of malignant melanoma as adjuvant therapy in Japan, the detailed mechanisms of their immunomodulatory effects are not fully understood. As the majority of TAMs are alternatively activated M2 macrophages that favour tumor development, the aim of this study was to elucidate the immunomodulatory effects of these reagents on human monocyte-derived M2 macrophages. First, mRNA expressions and protein production of immune checkpoint molecules, PD-L1 and chemokines by CD163+ CD206+ M2 macrophages derived from peripheral blood mononuclear cells were investigated to determine the immunomodulatory effects of DTIC, ACNU, and VCR. DTIC and VCR significantly decreased PD-L1 mRNA expression, which was confirmed by flow cytometry. Moreover, the mRNA expression and production of CCL22 were significantly decreased by DTIC, which suggested that DTIC might suppress the recruitment of Tregs in the tumor site. Furthermore, the decreased expression of PD-L1 and production of CCL22 were validated in vivo, using the B16F10 mouse melanoma model, leading to abrogation of the suppressive function of T-cell proliferation. The present report suggests one of the possible antimelanoma mechanisms of DAV combination chemotherapy for melanoma patients.


Assuntos
Antineoplásicos/farmacologia , Macrófagos/efeitos dos fármacos , Melanoma/tratamento farmacológico , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Antígeno B7-H1/metabolismo , Dacarbazina/farmacologia , Feminino , Japão , Lectinas Tipo C/metabolismo , Leucócitos Mononucleares/metabolismo , Macrófagos/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Melanoma Experimental , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Nimustina/farmacologia , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Vincristina/farmacologia , Melanoma Maligno Cutâneo
3.
Clin Cancer Res ; 23(22): 7020-7033, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28821557

RESUMO

Purpose: We aimed to characterize and target drug-tolerant BRCA1-deficient tumor cells that cause residual disease and subsequent tumor relapse.Experimental Design: We studied responses to various mono- and bifunctional alkylating agents in a genetically engineered mouse model for BRCA1/p53-mutant breast cancer. Because of the large intragenic deletion of the Brca1 gene, no restoration of BRCA1 function is possible, and therefore, no BRCA1-dependent acquired resistance occurs. To characterize the cell-cycle stage from which Brca1-/-;p53-/- mammary tumors arise after cisplatin treatment, we introduced the fluorescent ubiquitination-based cell-cycle indicator (FUCCI) construct into the tumor cells.Results: Despite repeated sensitivity to the MTD of platinum drugs, the Brca1-mutated mammary tumors are not eradicated, not even by a frequent dosing schedule. We show that relapse comes from single-nucleated cells delaying entry into the S-phase. Such slowly cycling cells, which are present within the drug-naïve tumors, are enriched in tumor remnants. Using the FUCCI construct, we identified nonfluorescent G0-like cells as the population most tolerant to platinum drugs. Intriguingly, these cells are more sensitive to the DNA-crosslinking agent nimustine, resulting in an increased number of multinucleated cells that lack clonogenicity. This is consistent with our in vivo finding that the nimustine MTD, among several alkylating agents, is the most effective in eradicating Brca1-mutated mouse mammary tumors.Conclusions: Our data show that targeting G0-like cells is crucial for the eradication of BRCA1/p53-deficient tumor cells. This can be achieved with selected alkylating agents such as nimustine. Clin Cancer Res; 23(22); 7020-33. ©2017 AACR.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos/genética , Genes BRCA1 , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/genética , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Cisplatino/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Genes p53 , Humanos , Camundongos , Camundongos Knockout , Nimustina/farmacologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-27599192

RESUMO

A new method to evaluate the anticancer activity at the molecular level has been developed. In our assay, the interaction between alkylating anticancer drugs-Fe3O4/CA with DNA has been investigated for the Resonance Light Scattering (RLS) signal enhancement. Water-based nano-Fe3O4, as a probe, has the ability of good solubility, biodegradability and low bulk resistivity etc. The experimental results show that, the activity order of three kinds of drugs is Nimustine (ACNU)>Semustine (Me-CCNU)>Chlormethine (HN2), which is satisfied with the results of the cell apoptosis experiment and the IC50 by MTT method. This assay is simple, sensitive and high efficient. And the theoretical basics for the development of new anticancer drugs as well as the assessments of their efficacy to cure breast and hepatic cancer have been provided.


Assuntos
Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacologia , Portadores de Fármacos/química , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Nanopartículas Metálicas/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ácido Cítrico/química , DNA/química , DNA/metabolismo , Portadores de Fármacos/farmacologia , Feminino , Compostos Férricos/química , Humanos , Concentração Inibidora 50 , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Células MCF-7 , Nimustina/farmacologia , Semustina/farmacologia , Espectrofotometria Infravermelho , Viscosidade
5.
J Pathol ; 241(4): 511-521, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27943283

RESUMO

Women with heterozygous germline mutations in the BRCA1 tumour suppressor gene are strongly predisposed to developing early-onset breast cancer through loss of the remaining wild-type BRCA1 allele and inactivation of TP53. Although tumour prevention strategies in BRCA1-mutation carriers are still limited to prophylactic surgery, several therapeutic strategies have been developed to target the DNA repair defects (also known as 'BRCAness') of BRCA1-deficient tumours. In particular, DNA-damaging agents such as platinum drugs and poly(ADP-ribose) polymerase (PARP) inhibitors show strong activity against BRCA1-mutated tumours. However, it is unclear whether drugs that target BRCAness can also be used to prevent tumour formation in BRCA1-mutation carriers, especially as loss of wild-type BRCA1 may not be the first event in BRCA1-associated tumourigenesis. We performed prophylactic treatments in a genetically engineered mouse model in which de novo development of BRCA1-deficient mammary tumours is induced by stochastic loss of BRCA1 and p53. We found that prophylactic window therapy with nimustine, cisplatin or olaparib reduced the amount and size of mammary gland lesions, and significantly increased the median tumour latency. Similar results were obtained with intermittent prophylactic treatment with olaparib. Importantly, prophylactic window therapy with nimustine and cisplatin resulted in an increased fraction of BRCA1-proficient mammary tumours, suggesting selective survival and malignant transformation of BRCA1-proficient lesions upon prophylactic treatment with DNA-damaging agents. Prophylactic therapy with olaparib significantly prolonged mammary tumour-free survival without any significant increase in the fraction of BRCA1-proficient tumours, warranting the evaluation of this PARP inhibitor in prophylactic trials in BRCA1-mutation carriers. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Mamárias Animais/tratamento farmacológico , Ftalazinas/farmacologia , Piperazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Supressoras de Tumor/genética , Animais , Antineoplásicos/uso terapêutico , Proteína BRCA1 , Cisplatino/farmacologia , Reparo do DNA , Modelos Animais de Doenças , Feminino , Mutação em Linhagem Germinativa , Humanos , Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/prevenção & controle , Camundongos , Nimustina/farmacologia , Ftalazinas/uso terapêutico , Piperazinas/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Supressoras de Tumor/metabolismo
6.
Cancer Med ; 5(11): 3147-3155, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27734611

RESUMO

Nimustine (ACNU) has antitumor activities in patients with malignant glioma. Hyperbaric oxygen (HBO) may enhance the efficacy of certain therapies that are hampered by the hypoxic microenvironment. We examined the combined effects of ACNU and HBO in a GFP transgenic nude mice bearing human glioma model. Mice inoculated with human glioma cells SU3 were randomly divided into the four groups: (A) the control group, (B) the HBOT (HBO therapy) group, (C) the ACNU group, and (D) the HBOT+ACNU group. Tumor size was measured at the indicated time intervals with a caliper; mice were sacrificed 28 days after treatment, and immunohistochemistry staining and western blot analysis were carried out. By the end of the trial, the tumor weights of groups A, B, C, and D were (P < 0.05), 6.03 ± 1.47, 4.13 ± 1.82 (P < 0.05), 2.39 ± 0.25 (P < 0.05), and 1.43 ± 0.38 (P < 0.01), respectively. The expressions of TNF-α, MMP9, HIF-α, VEGF, NF-κB, and IL-1ß were associated with the infiltration of inflammatory cells and the inhibition rate of tumor cells. Hyperbaric oxygen therapy (HBOT) could inhibit glioma cell proliferation and inflammatory cell infiltration, and exert a sensitizing effect on ACNU therapy partially through enhancing oxygen pressure (PO2 ) in tumor tissues and lower expression levels of HIF-1α, TNF-α, IL-1ß, VEGF, MMP9, and NF-κB.


Assuntos
Antineoplásicos/farmacologia , Glioma/metabolismo , Glioma/patologia , Oxigenoterapia Hiperbárica , Nimustina/farmacologia , Animais , Biomarcadores , Peso Corporal , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glioma/mortalidade , Glioma/terapia , Humanos , Oxigenoterapia Hiperbárica/métodos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Transgênicos , Modelos Biológicos , NF-kappa B/metabolismo , Transdução de Sinais , Carga Tumoral , Fator de Necrose Tumoral alfa/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncotarget ; 6(32): 33755-68, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26418950

RESUMO

The outcome of cancer therapy strongly depends on the complex network of cell signaling pathways, including transcription factor activation following drug exposure. Here we assessed whether and how the MAP kinase (MAPK) cascade and its downstream target, the transcription factor AP-1, influence the sensitivity of malignant glioma cells to the anticancer drugs temozolomide (TMZ) and nimustine (ACNU). Both drugs induce apoptosis in glioma cells at late times following treatment. Activation of the MAPK cascade precedes apoptosis, as shown by phosphorylation of Jun kinase (JNK) and c-Jun, a main component of AP-1. Pharmacological inhibition and siRNA mediated knockdown of JNK and c-Jun reduced the level of apoptosis in LN-229 glioma cells treated with TMZ or ACNU. Analyzing the underlying molecular mechanism, we identified the pro-apoptotic gene BIM as a critical target of AP-1, which is upregulated following TMZ and ACNU. Importantly, shRNA mediated downregulation of BIM in the malignant glioma cell lines LN-229 and U87MG led to an attenuated cleavage of caspase-9 and, consequently, reduced the level of apoptosis following TMZ and ACNU treatment. Overall, we identified JNK/c-Jun activation and BIM induction as a late pro-apoptotic response of glioma cells treated with alkylating anticancer drugs.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Dacarbazina/análogos & derivados , Glioblastoma/patologia , MAP Quinase Quinase 4/metabolismo , Proteínas de Membrana/metabolismo , Nimustina/farmacologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Antineoplásicos Alquilantes/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína 11 Semelhante a Bcl-2 , Linhagem Celular Tumoral/efeitos dos fármacos , Núcleo Celular/metabolismo , Ensaio Cometa , Dacarbazina/farmacologia , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Fosforilação , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Temozolomida , Regulação para Cima
8.
J Vet Med Sci ; 77(6): 677-84, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25715778

RESUMO

Canine histiocytic sarcoma (HS) is an aggressive tumor type originating from histiocytic cell lineages. This disease is characterized by poor response to chemotherapy and short survival time. Therefore, it is of critical importance to identify and develop effective antitumor drugs against HS. The objectives of this study were to examine the drug sensitivities of 10 antitumor drugs. Using a real-time RT-PCR system, the mRNA expression levels of 16 genes related to drug resistance in 4 canine HS cell lines established from dogs with disseminated HS were determined and compared to 2 canine lymphoma cell lines (B-cell and T-cell). These 4 canine HS cell lines showed sensitivities toward microtubule inhibitors (vincristine, vinblastine and paclitaxel), comparable to those in the canine B-cell lymphoma cell line. Moreover, it was shown that P-gp in the HS cell lines used in this study did not have enough function to efflux its substrate. Sensitivities to melphalan, nimustine, methotrexate, cytarabine, doxorubicin and etoposide were lower in the 4 HS cell lines than in the 2 canine lymphoma cell lines. The data obtained in this study using cultured cell lines could prove helpful in the developing of advanced and effective chemotherapies for treating dogs that are suffering from HS.


Assuntos
Antineoplásicos/farmacologia , Doenças do Cão/tratamento farmacológico , Doenças do Cão/genética , Avaliação Pré-Clínica de Medicamentos/veterinária , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Seleção de Medicamentos Antitumorais/veterinária , Sarcoma Histiocítico/veterinária , Animais , Linhagem Celular Tumoral , Citarabina/farmacologia , Cães , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Etoposídeo/farmacologia , Sarcoma Histiocítico/tratamento farmacológico , Sarcoma Histiocítico/genética , Melfalan/farmacologia , Metotrexato/farmacologia , Nimustina/farmacologia , Paclitaxel/farmacologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Vimblastina/farmacologia , Vincristina/farmacologia
9.
Rapid Commun Mass Spectrom ; 28(5): 439-47, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24497281

RESUMO

RATIONALE: Chloroethylnitrosoureas (CENUs) are important alkylating agents employed for the clinical treatment of cancer. The cellular toxicity of CENUs is primarily due to induction of DNA interstrand crosslinks (ICLs), which has been characterized as l-(3-deoxycytidyl), 2-(l-deoxyguanosinyl)ethane (dG-dC). However, the formation of dG-dC crosslinks can be prevented by O(6) -alkylguanine-DNA alkyltransferase (AGT), which removes the O(6) -chloroethyl group from O(6) -chloroethylguanine (O(6) -ClEt-Gua), and ultimately its increased expression can result in drug resistance. Differing levels of AGT expression can lead to varying amounts of dG-dC crosslinking, which influences the sensitivity of cells to CENUs. METHODS: In this work, a sensitive method for the quantitation of dG-dC crosslinks in cellular DNA has been established using high-performance liquid chromatography/electrospray ionization tandem mass spectrometry (HPLC/ESI-MS/MS). RESULTS: The limit of detection (LOD) and limit of quantitation (LOQ) of the method were determined to be 2 fmol and 8 fmol on-column, respectively, and the recovery ranged from 96% to 105% with the relative standard deviation (RSD) below 5%. Using this method, the levels of dG-dC crosslink induced by 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea hydrochloride (ACNU) were determined in NIH/3T3 fibroblasts cells (high level of expression of AGT) and L1210 leukemia cells (low level of expression of AGT). The time-course profile indicated that the levels of dG-dC crosslink uniformly increased in the early incubation period and reached the maximum at 12 h. Subsequently, the amount of dG-dC crosslinking decreased to very low levels presumably owing to the repair of O(6) -ClEt-Gua by AGT. The crosslinking levels in L1210 cells were significantly higher than those in NIH/3T3 cells at each time point. This provides strong evidence that high express of AGT in CENU-resistant cells inhibits the formation of dG-dC crosslinks. CONCLUSIONS: This work will contribute to the further understanding of the drug resistance of CENUs, and will provide a means to evaluate the anticancer activity of new bifunctional anticancer agents.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Reagentes de Ligações Cruzadas/farmacologia , DNA , Nimustina/farmacologia , Espectrometria de Massas em Tandem/métodos , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , DNA/análise , DNA/química , DNA/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Limite de Detecção , Camundongos , Células NIH 3T3 , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização por Electrospray
10.
Cancer Gene Ther ; 19(11): 802-10, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23037811

RESUMO

Chemoprotection of haematopoietic stem cells (HSCs) by gene therapeutic transfer of drug-resistance genes represents the encouraging approach to prevent myelosuppression, which is one of the most severe side effects in tumor therapy. Thus, we cloned and evaluated six different bicistronic lentiviral SIN vectors encoding two transgenes, MGMT(P140K) (an O(6)-benzylguanine-resistant mutant of methylguanine-DNA methyltransferase) and MDR1 (multidrug resistance 1), using various linker sequences (IRESEMCV, IRESFMDV and 2A-element of FMDV (F2A)). Expression of both transgenes in HL-60 and in K562 cells was assayed by quantitative real-time PCR. Combination therapy with ACNU plus paclitaxel in HL-60 cells and with carmustin (BCNU) plus doxorubicin in K562 cells resulted in the most significant survival advantage of cells transduced with the lentiviral vector HR'SIN-MGMT(P140K)-F2A-MDR1 compared with untransduced cells. In human HSCs, overexpression of both transgenes by this vector also caused significantly increased survival and enrichment of transduced cells after treatment with BCNU plus doxorubicin or temozolomide plus paclitaxel. In summary, we could show significant chemoprotection by overexpression of MDR1 and MGMT(P140K) with a lentiviral vector using the F2A linker element in two different haematopoietic cell lines and in human primary HSCs with various combination regimens. Consequently, we are convinced that these in vitro investigations will help to improve combination chemotherapy regimens by reducing myelotoxic side effects and increasing the therapeutic efficiency.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Vetores Genéticos/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Lentivirus/metabolismo , Peptídeos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Carmustina/farmacologia , Sobrevivência Celular , Clonagem Molecular , Citoproteção , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Doxorrubicina/farmacologia , Vetores Genéticos/genética , Células HL-60 , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células K562 , Lentivirus/genética , Nimustina/farmacologia , Paclitaxel/farmacologia , Peptídeos/genética , Reação em Cadeia da Polimerase em Tempo Real , Temozolomida , Transgenes , Proteínas Supressoras de Tumor/genética
11.
J Dermatol Sci ; 68(1): 19-24, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22902076

RESUMO

BACKGROUND: The treatment of melanoma, an aggressive, chemo-resistant skin cancer characterized by rapid metastasis and a poor prognosis, requires the development of innovative therapies with improved efficacy. The p53R2 gene that encodes the ribonucleotide reductase small subunit 2 homologue is induced by several stress signals including DNA-damaging agents that activate p53. The p53R2 gene product increases the deoxynucleotide triphosphate pool in the nucleus; this facilitates DNA repair and synthesis. OBJECTIVE: We examined the expression of p53R2 in melanoma and evaluated whether p53R2 is involved in the growth and proliferation of melanoma cells. Methods We examined the clinicopathological significance of p53R2 in melanoma. To investigate the role of p53R2 in melanoma we used KHm5 and KHm6 melanoma cells that express p53R2, and p53R2-targeting small interfering (si) RNA. RESULTS: p53R2 expression was detected immunohistochemically in 56 of 78 patients (71.8%). The expression of p53R2 was significantly correlated with the depth of invasion and the tumor stage. p53R2-targeting siRNA successfully knocked down p53R2 and significantly inhibited the growth of KHm5 and 6 cells. Moreover, The degree of KHm5 and 6 cell growth inhibition was greater in the presence of both p53R2-targeting siRNA and nimustine (ACNU) than with ACNU alone, suggesting that p53R2 silencing enhanced the chemosensitivity of KHm5 and 6 cells to ACNU. CONCLUSIONS: We propose p53R2 as a therapeutic target to enhance the effectiveness of chemotherapy in patients with p53R2-positive melanoma.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Melanoma/enzimologia , Ribonucleotídeo Redutases/metabolismo , Neoplasias Cutâneas/enzimologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Criança , Cisplatino/farmacologia , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dacarbazina/farmacologia , Relação Dose-Resposta a Droga , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Melanoma/genética , Melanoma/patologia , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Nimustina/farmacologia , Prognóstico , Interferência de RNA , Ribonucleotídeo Redutases/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Transfecção , Vincristina/farmacologia , Adulto Jovem
12.
Biochim Biophys Acta ; 1823(7): 1199-207, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22609303

RESUMO

FRA1 belongs, together with c-Fos and FosB, to the family of Fos proteins that form with members of the ATF and Jun family the transcription factor AP-1 (activator protein 1). Previously we showed that c-Fos protects mouse embryonic fibroblasts against the cytotoxic effects of ultraviolet (UV) light by induction of the endonuclease XPF, leading to enhanced nucleotide excision repair (NER) activity. Here, we analyzed the regulation of FRA1 in glioma cells treated with the anticancer drug nimustine (ACNU) and its role in ACNU-induced toxicity. We show that FRA1 is upregulated in glioblastoma cells following ACNU on mRNA and protein levels. Knockdown of FRA1 by either siRNA or shRNA clearly sensitized glioma cells towards ACNU-induced cell death. Despite decreased AP-1 binding activity upon FRA1 knockdown, this effect is independent on regulation of the AP-1 target genes fasL, ercc1 and xpf. In addition, FRA1 knockdown does not affect DNA repair capacity. However, lack of FRA1 attenuated the ACNU-induced phosphorylation of CHK1 and led to a reduced arrest of cells in G2/M and, thereby, presumably leads to enhanced cell death in the subsequent cell cycle.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioma/tratamento farmacológico , Nimustina/uso terapêutico , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Reparo do DNA/genética , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Seleção de Medicamentos Antitumorais , Proteína Ligante Fas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Inativação Gênica/efeitos dos fármacos , Glioma/genética , Glioma/patologia , Humanos , Camundongos , Nimustina/farmacologia , Fator de Transcrição AP-1/metabolismo
13.
Neurotox Res ; 22(1): 43-58, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22203610

RESUMO

Effects of chemotherapeutics on glioma cell lines and spheroids are usually investigated without evaluating the effects of chemotherapeutics on normal brain tissue. To perform such investigations, the aim of this study was to establish a panel of markers for detection of general cell death and more specific neuronal and glial degeneration induced by chemotherapeutics in organotypic rat corticostriatal slice cultures. The slice cultures were exposed to the alkylating agents temozolomide (TMZ) and nimustine (ACNU), the tyrosine kinase inhibitor imatinib mesylate (IM) and the microtubule-destabilizing agent vincristine (VCR). Densitometric measurements of uptake of the fluorescent dye propidium iodide (PI) were used for quantifying cellular degeneration. Moreover, paraffin sections were hematoxylin eosine stained and immunostained for the neuronal marker microtubule-associated protein 2 (MAP2), the astroglial marker glial fibrillary acidic protein (GFAP), and the oligodendroglial marker p25α. The results showed that the supposed clinically relevant drug concentrations were non-toxic. However, a time dependent increase in PI uptake was observed for high drug concentrations, except for TMZ, where no toxicity was observed. Corresponding immunostaining showed loss of MAP2 and increased expression of GFAP and p25α for cultures exposed to 1,000 nM VCR. Cultures exposed to high concentrations of ACNU and IM disintegrated, leaving no tissue for histology. In conclusion, corticostriatal slice cultures and the established panel of markers represent an excellent tool for detecting toxicity induced by chemotherapeutics. Toxicity was not detected at clinical concentrations, but high concentrations with toxic effects were identified suggesting that some of the earlier identified anti-cancer effects are general cytotoxic effects and not specific anti-cancer effects.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Transporte/análise , Proteína Glial Fibrilar Ácida/análise , Proteínas Associadas aos Microtúbulos/análise , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Benzamidas , Biomarcadores/análise , Morte Celular/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Mesilato de Imatinib , Imuno-Histoquímica , Indicadores e Reagentes , Nimustina/farmacologia , Piperazinas/farmacologia , Propídio , Pirimidinas/farmacologia , Ratos , Temozolomida , Vincristina/farmacologia
14.
Int Immunopharmacol ; 11(11): 1844-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21820086

RESUMO

Transferrin receptor (TfR) has been used as a target for antibody-based therapy of cancer. Anti-TfR antibody together with chemotherapeutic drugs has potential for cancer therapy. In this study, we investigated the in vitro anti-tumor effects of the anti-TfR monoclonal antibody (mAb), 7579, alone or in combination with Nimustine, a chemotherapeutic drug, on the gliomas cell lines U251 and U87MG. Our results indicated that 7579 alone dramatically down-regulated surface expression of TfR on tumor cells and induced S phase accumulation and apoptosis of tumor cells. Compared with 7579 or Nimustine used alone, the combination of 7579 with Nimustine demonstrated enhanced growth inhibitory effect on tumor cells. PI (Propidium iodide)/Annexin V staining analyzed by FCM (flow cytometry) demonstrated that 7579 enhanced the cytotoxic effects of chemotherapeutic drug on tumor cells, indicating the therapeutic effect of 7579 was mediated mainly by promoting tumor cell necrosis. Using the median-effect/combination-index isobologram method, we further evaluated the nature of 7579/chemotherapeutic drug interactions. Synergistic interaction was observed for combination of 7579 with Nimustine. Our study provides additional evidence to develop combination therapies of anti-TfR mAbs-plus chemoimmunotherapy for gliomas.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Nimustina/farmacologia , Receptores da Transferrina/antagonistas & inibidores , Anticorpos Monoclonais Murinos/administração & dosagem , Antineoplásicos/administração & dosagem , Western Blotting , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Glioma/patologia , Humanos , Imuno-Histoquímica , Nimustina/administração & dosagem , Receptores da Transferrina/imunologia
15.
PLoS One ; 6(5): e19659, 2011 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-21573016

RESUMO

Nimustine (ACNU) and temozolomide (TMZ) are DNA alkylating agents which are commonly used in chemotherapy for glioblastomas. ACNU is a DNA cross-linking agent and TMZ is a methylating agent. The therapeutic efficacy of these agents is limited by the development of resistance. In this work, the role of the Fanconi anemia (FA) repair pathway for DNA damage induced by ACNU or TMZ was examined. Cultured mouse embryonic fibroblasts were used: FANCA(-/-), FANCC(-/-), FANCA(-/-)C(-/-), FANCD2(-/-) cells and their parental cells, and Chinese hamster ovary and lung fibroblast cells were used: FANCD1/BRCA2mt, FANCG(-/-) and their parental cells. Cell survival was examined after a 3 h ACNU or TMZ treatment by using colony formation assays. All FA repair pathways were involved in ACNU-induced DNA damage. However, FANCG and FANCD1/BRCA2 played notably important roles in the repair of TMZ-induced DNA damage. The most effective molecular target correlating with cellular sensitivity to both ACNU and TMZ was FANCD1/BRCA2. In addition, it was found that FANCD1/BRCA2 small interference RNA efficiently enhanced cellular sensitivity toward ACNU and TMZ in human glioblastoma A172 cells. These findings suggest that the down-regulation of FANCD1/BRCA2 might be an effective strategy to increase cellular chemo-sensitization towards ACNU and TMZ.


Assuntos
Proteína BRCA2/metabolismo , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Dacarbazina/análogos & derivados , Nimustina/farmacologia , Animais , Linhagem Celular , Dano ao DNA/genética , Reparo do DNA/genética , Dacarbazina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Anemia de Fanconi/genética , Inativação Gênica/efeitos dos fármacos , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Modelos Biológicos , RNA Interferente Pequeno/metabolismo , Rad51 Recombinase/metabolismo , Recombinação Genética/efeitos dos fármacos , Recombinação Genética/genética , Temozolomida
16.
Anticancer Res ; 30(9): 3509-14, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20944130

RESUMO

BACKGROUND: Malignant glioma is one of the most intractable diseases in the human body. Rho-kinase (ROCK) is overexpressed and has been proposed as the main cause for the refractoriness of the disease. Since efficacious treatment is required, this study investigated the effect of inhibition of ROCK isoforms. MATERIALS AND METHODS: The short hairpin RNA transcription vector was transfected into the RT2 rat glioma cell line and the characteristics of the cells were investigated. The effect of nimustine hydrochloride (ACNU) anti-neoplastic agent on cells was also measured. RESULTS: Inhibition of ROCK isoforms did not alter cell growth. Cell cycle analysis revealed that ROCK1 down-regulation reduced the G(0) phase population and ROCK2 down-regulation reduced the G(2)/M phase population. When ROCK1-down-regulated cells were exposed to ACNU, they demonstrated susceptibility to the agent. CONCLUSION: The roles of ROCK1 and ROCK2 may be different in glioma cells. Furthermore, the combination of ROCK1 down-regulation and an anti-neoplastic agent may be useful for the therapy of malignant glioma.


Assuntos
Glioma/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glioma/genética , Humanos , Immunoblotting , Isoenzimas , Nimustina/farmacologia , RNA Interferente Pequeno , Ratos , Transfecção , Quinases Associadas a rho/genética
17.
Cancer Sci ; 101(8): 1881-5, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20487264

RESUMO

Nimustine (ACNU) is a chloroethylating agent which was the most active chemotherapy agent used for patients with high-grade gliomas until the introduction of temozolomide, which became the standard of care for patients with newly diagnosed glioblastomas in Japan. Since temozolomide was established as the standard first-line therapy for glioblastoma multiforme (GBM), ACNU has been employed as a salvage chemotherapy agent for recurrent GBM in combination with other drugs. The acting molecular mechanism in ACNU has yet to be elucidated. ACNU is a cross-linking agent which induces DNA double-strand breaks (DSBs). The work described here was intended to clarify details in repair pathways which are active in the repair of DNA DSBs induced by ACNU. DSBs are repaired through the homologous recombination (HR) and non-homologous end-joining (NHEJ) pathways. Cultured mouse embryonic fibroblasts were used which have deficiencies in DNA DSB repair genes which are involved in HR repair (X-ray repair cross-complementing group 2 [XRCC2] and radiation sensitive mutant 54 [Rad54]), and in NHEJ repair (DNA ligase IV [Lig4]). Cellular sensitivity to ACNU treatment was evaluated with colony forming assays. The most effective molecular target which correlated with ACNU cell sensitivity was Lig4. In addition, it was found that Lig4 small-interference RNA (siRNA) efficiently enhanced cell lethality which was induced by ACNU in human glioblastoma A172 cells. These findings suggest that the down-regulation of Lig4 might provide a useful tool which can be used to increase cell sensitivity in response to ACNU chemotherapy.


Assuntos
Antineoplásicos/farmacologia , DNA Ligases/antagonistas & inibidores , Nimustina/farmacologia , Animais , Linhagem Celular , Quebras de DNA de Cadeia Dupla , DNA Helicases , DNA Ligase Dependente de ATP , DNA Ligases/fisiologia , Reparo do DNA , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Camundongos , Proteínas Nucleares/fisiologia
18.
Ai Zheng ; 28(6): 575-80, 2009 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-19635193

RESUMO

BACKGROUND AND OBJECTIVE: Epigenetic silencing of the DNA repair gene, O6-methylguanine-DNA methyltransferase (MGMT), is associated with the therapeutic response to methylating agents. This study was to assess the value of detecting the promoter methylation of MGMT gene in chemotherapy for glioma. METHODS: Methylation-specific PCR (MSP) was employed to detect MGMT promoter CpG island methylation in 39 samples of glioma taken from surgery. Western blot and immunohistochemistry were used to detect protein expression. MTT were employed to detect the sensitivity of two glioma cell lines to alkylating agents, ACNU and TMZ. The Kaplan-Meier curve was adopted to estimate the overall survival according to the methylation status of the MGMT promoter. RESULTS: Methylation of MGMT promoter CpG island was detectable in 46.2% of glioma tissues, but not in any normal tissues. The expression rate of MGMT protein was 61.5%. The status of MGMT methylation status was association with the protein level of MGMT (P<0.05). The MGMT gene was demethylated in glioma cell line SHG-44 following 5-Aza-CdR treatment; the expression of MGMT protein was restored and the resistance of SHG44 cells to alkylating agents was reversed. The overall survival was higher in patients with methylated MGMT promoter than in those with unmethylated MGMT promoter (P<0.05). CONCLUSIONS: The status of MGMT promoter CpG island methylation is closely correlated to MGMT protein expression and sensitivity of cells to alkylating agents in glioma. Detection of the methylated sequences of MGMT may be used as a predictive factor for the treatment of glioma.


Assuntos
Neoplasias Encefálicas , Metilação de DNA , Glioma , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Adolescente , Adulto , Idoso , Antimetabólitos Antineoplásicos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Ilhas de CpG/genética , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Decitabina , Resistencia a Medicamentos Antineoplásicos , Feminino , Seguimentos , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Nimustina/farmacologia , Regiões Promotoras Genéticas , Taxa de Sobrevida , Temozolomida , Adulto Jovem
19.
Anticancer Res ; 29(7): 2453-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19596913

RESUMO

Flap endonuclease 1 (FEN1) is a DNA replication/repair protein involved in Okazaki fragment processing, long-patch base excision repair, DNA double-strand break repair and stalled replication fork restart. FEN1 is also important for preservation of telomere stability and thus a key node in maintaining genomic stability. The aim of the present study was to elucidate the level of expression of FEN1 protein in cancer of testis, lung and brain. FEN1 protein expression was studied by Western blot analysis in specimens of tumor tissues compared with the normal tissue from the same patient or normal brain extract. In addition, FEN1 was transiently down-regulated in the glioblastoma cell line LN308 by transfection with siRNA. The transfected cells were treated with cisplatin, temozolomide, nimustine and methyl methanesulfonate (MMS). Induced apoptosis (subG1 fraction) was analysed by flow cytometry. Our data show a clear overexpression of FEN1 in 19/25 samples from testicular tumors (mostly seminomas) and 4/4 samples from lung tumors (non-small cell lung cancer). For brain tumors, 9/11 glioblastoma multiforme and 5/8 astrocytomas expressed FEN1 protein at a higher level than did normal brain tissue. Overall, the data demonstrate that FEN1 overexpression is common in testis, lung and brain tumors. Low-level expression of FEN1 by siRNA down-regulation increased sensitivity to methylating agents (temozolomide, MMS) and cisplatin in LN308 glioma cells, which indicates that altered FEN1 expression might impact the therapeutic response.


Assuntos
Neoplasias Encefálicas/metabolismo , Endonucleases Flap/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Testiculares/metabolismo , Antineoplásicos/farmacologia , Apoptose , Sequência de Bases , Neoplasias Encefálicas/patologia , Cisplatino/farmacologia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Endonucleases Flap/genética , Citometria de Fluxo , Humanos , Neoplasias Pulmonares/patologia , Masculino , Metanossulfonato de Metila/farmacologia , Nimustina/farmacologia , RNA Mensageiro/genética , RNA Interferente Pequeno , Temozolomida , Neoplasias Testiculares/patologia
20.
Biochem Pharmacol ; 76(3): 303-11, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18573489

RESUMO

O6-Methylguanine and O6-chloroethylguanine are primary DNA lesions produced by two types of antineoplastic drugs, 8-carbamoyl-3-methylimidazo[5,1-d]-1,2,3,5-tetrazin-4(3H)-one (temozolomide, TMZ) and 1-(4-amino-2-methyl-5-pyrimidinyl) methyl-3-(2-chloroethyl)-3-nitrosourea (ACNU), respectively. They can be repaired by O6-methylguanine-DNA methyltransferase, coded by the Mgmt gene. Otherwise, these two types of lesions induce apoptosis in different ways. O6-Chloroethylguanine blocks DNA replication thereby inducing apoptosis. On the other hand, O6-methylguanine does not block DNA replication and the resulting O6-methylguanine-thymine mispair is recognized by mismatch repair-related proteins, including MLH1, thereby inducing apoptosis. Reflecting this, mouse cells lacking both MGMT and MLH1 are resistant to TMZ, but not to ACNU. The translocation of phosphatidylserine in cell membrane as well as a change of mitochondrial transmembrane potentials occurred in an MLH1-dependent manner after treatment with TMZ, but no such MLH1 dependency was observed in the case of ACNU treatment. By using cell lines defective in both APAF-1 and MGMT, it was revealed that the APAF-1 function is required for execution of apoptosis induced by either TMZ or ACNU. There is almost 12h delay in occurrence of apoptosis-related mitochondrial depolarization in TMZ-treated cells in comparison to those of ACNU-treated cells, reflecting the fact that at least one cycle of DNA replication is required to trigger apoptosis in the former case, but not in the latter.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Dano ao DNA , Reparo do DNA , Dacarbazina/análogos & derivados , Fibroblastos/efeitos dos fármacos , Nimustina/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Antineoplásicos/química , Apoptose/genética , Linhagem Celular , Dacarbazina/química , Dacarbazina/farmacologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteína 1 Homóloga a MutL , Nimustina/química , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , O(6)-Metilguanina-DNA Metiltransferase/genética , O(6)-Metilguanina-DNA Metiltransferase/fisiologia , Fosfatidilserinas/metabolismo , Relação Estrutura-Atividade , Temozolomida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...