Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cancer Cell ; 41(5): 853-870.e13, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-37084735

RESUMO

We uncover a tumor-suppressive process in urothelium called transcriptional-translational conflict caused by deregulation of the central chromatin remodeling component ARID1A. Loss of Arid1a triggers an increase in a nexus of pro-proliferation transcripts, but a simultaneous inhibition of the eukaryotic elongation factor 2 (eEF2), which results in tumor suppression. Resolution of this conflict through enhancing translation elongation speed enables the efficient and precise synthesis of a network of poised mRNAs resulting in uncontrolled proliferation, clonogenic growth, and bladder cancer progression. We observe a similar phenomenon in patients with ARID1A-low tumors, which also exhibit increased translation elongation activity through eEF2. These findings have important clinical implications because ARID1A-deficient, but not ARID1A-proficient, tumors are sensitive to pharmacologic inhibition of protein synthesis. These discoveries reveal an oncogenic stress created by transcriptional-translational conflict and provide a unified gene expression model that unveils the importance of the crosstalk between transcription and translation in promoting cancer.


Assuntos
Cromatina , Neoplasias da Bexiga Urinária , Humanos , Neoplasias da Bexiga Urinária/genética
2.
Oncogene ; 39(45): 6920-6934, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32989257

RESUMO

Oncogenic mutations in RAS genes, like KRASG12D or NRASG12D, trap Ras in the active state and cause myeloproliferative disorder and T cell leukemia (T-ALL) when induced in the bone marrow via Mx1CRE. The RAS exchange factor RASGRP1 is frequently overexpressed in T-ALL patients. In T-ALL cell lines overexpression of RASGRP1 increases flux through the RASGTP/RasGDP cycle. Here we expanded RASGRP1 expression surveys in pediatric T-ALL and generated a RoLoRiG mouse model crossed to Mx1CRE to determine the consequences of induced RASGRP1 overexpression in primary hematopoietic cells. RASGRP1-overexpressing, GFP-positive cells outcompeted wild type cells and dominated the peripheral blood compartment over time. RASGRP1 overexpression bestows gain-of-function colony formation properties to bone marrow progenitors in medium containing limited growth factors. RASGRP1 overexpression enhances baseline mTOR-S6 signaling in the bone marrow, but not in vitro cytokine-induced signals. In agreement with these mechanistic findings, hRASGRP1-ires-EGFP enhances fitness of stem- and progenitor- cells, but only in the context of native hematopoiesis. RASGRP1 overexpression is distinct from KRASG12D or NRASG12D, does not cause acute leukemia on its own, and leukemia virus insertion frequencies predict that RASGRP1 overexpression can effectively cooperate with lesions in many other genes to cause acute T-ALL.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/patologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Animais , Medula Óssea/patologia , Células Cultivadas , Criança , Ensaio de Unidades Formadoras de Colônias , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Feminino , Regulação Leucêmica da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/genética , Transplante de Células-Tronco Hematopoéticas , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Cultura Primária de Células , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo , Quimeras de Transplante
3.
JCI Insight ; 52019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31237864

RESUMO

Colorectal cancer (CRC) is the third most frequent neoplastic disorder and is a main cause of tumor-related mortality as many patients progress to stage IV metastatic CRC. Standard care consists of combination chemotherapy (FOLFIRI or FOLFOX). Patients with WT KRAS typing are eligible to receive anti-EGFR therapy combined with chemotherapy. Unfortunately, predicting efficacy of CRC anti-EGFR therapy has remained challenging. Here we uncover that the EGFR-pathway component RasGRP1 acts as CRC tumor suppressor in the context of aberrant Wnt signaling. We find that RasGRP1 suppresses EGF-driven proliferation of colonic epithelial organoids. Having established that RasGRP1 dosage levels impacts biology, we focused on CRC patients next. Mining five different data platforms, we establish that RasGRP1 expression levels decrease with CRC progression and predict poor clinical outcome of patients. Lastly, deletion of one or two Rasgrp1 alleles makes CRC spheroids more susceptible to EGFR inhibition. Retrospective analysis of the CALGB80203 clinical trial shows that addition of anti-EGFR therapy to chemotherapy significantly improves outcome for CRC patients when tumors express low RasGRP1 suppressor levels. In sum, RasGRP1 is a unique biomarker positioned in the EGFR pathway and of potential relevance to anti-EGFR therapy for CRC patients.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Proteínas de Ligação a DNA/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Antineoplásicos Imunológicos/farmacologia , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Proliferação de Células/efeitos dos fármacos , Cetuximab/farmacologia , Cetuximab/uso terapêutico , Ensaios Clínicos como Assunto , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Biologia Computacional , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Conjuntos de Dados como Assunto , Modelos Animais de Doenças , Progressão da Doença , Intervalo Livre de Doença , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Fatores de Troca do Nucleotídeo Guanina/análise , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Knockout , Cultura Primária de Células , Prognóstico , Transdução de Sinais/efeitos dos fármacos , Esferoides Celulares , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/análise , Proteínas Supressoras de Tumor/genética
4.
Cancer Cell ; 31(5): 685-696.e6, 2017 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-28486107

RESUMO

Constitutive activation of Gαq signaling by mutations in GNAQ or GNA11 occurs in over 80% of uveal melanomas (UMs) and activates MAPK. Protein kinase C (PKC) has been implicated as a link, but the mechanistic details remained unclear. We identified PKC δ and ɛ as required and sufficient to activate MAPK in GNAQ mutant melanomas. MAPK activation depends on Ras and is caused by RasGRP3, which is significantly and selectively overexpressed in response to GNAQ/11 mutation in UM. RasGRP3 activation occurs via PKC δ- and ɛ-dependent phosphorylation and PKC-independent, DAG-mediated membrane recruitment, possibly explaining the limited effect of PKC inhibitors to durably suppress MAPK in UM. The findings nominate RasGRP3 as a therapeutic target for cancers driven by oncogenic GNAQ/11.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Sistema de Sinalização das MAP Quinases , Melanoma/enzimologia , Mutação , Neoplasias Uveais/enzimologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Diglicerídeos/metabolismo , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/patologia , Camundongos Nus , Fosforilação , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/genética , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Fatores de Tempo , Transfecção , Carga Tumoral , Neoplasias Uveais/tratamento farmacológico , Neoplasias Uveais/genética , Neoplasias Uveais/patologia , Fatores ras de Troca de Nucleotídeo Guanina
7.
Nat Cell Biol ; 17(6): 804-15, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26005835

RESUMO

The character of EGFR signals can influence cell fate but mechanistic insights into intestinal EGFR-Ras signalling are limited. Here we show that two distinct Ras nucleotide exchange factors, RasGRP1 and SOS1, lie downstream of EGFR but act in functional opposition. RasGRP1 is expressed in intestinal crypts where it restricts epithelial growth. High RasGRP1 expression in colorectal cancer (CRC) patient samples correlates with a better clinical outcome. Biochemically, we find that RasGRP1 creates a negative feedback loop that limits proliferative EGFR-SOS1-Ras signals in CRC cells. Genetic Rasgrp1 depletion from mice with either an activating mutation in KRas or with aberrant Wnt signalling due to a mutation in Apc resulted in both cases in exacerbated Ras-ERK signalling and cell proliferation. The unexpected opposing cell biological effects of EGFR-RasGRP1 and EGFR-SOS1 signals in the same cell shed light on the intricacy of EGFR-Ras signalling in normal epithelium and carcinoma.


Assuntos
Receptores ErbB/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Mucosa Intestinal/metabolismo , Proteína SOS1/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/metabolismo , Células Epiteliais/metabolismo , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Humanos , Mucosa Intestinal/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transplante de Neoplasias , Proteínas Proto-Oncogênicas p21(ras)/genética , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Transplante Heterólogo , Proteínas Wnt/genética , Via de Sinalização Wnt/genética
8.
Sci Signal ; 6(304): ra105, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24300897

RESUMO

Oncogenic K-Ras proteins, such as K-Ras(G12D), accumulate in the active, guanosine triphosphate (GTP)-bound conformation and stimulate signaling through effector kinases. The presence of the K-Ras(G12D) oncoprotein at a similar abundance to that of endogenous wild-type K-Ras results in only minimal phosphorylation and activation of the canonical Raf-mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) signaling cascades in primary hematopoietic cells, and these pathways remain dependent on growth factors for efficient activation. We showed that phospholipase C-γ (PLC-γ), PI3K, and their generated second messengers link activated cytokine receptors to Ras and ERK signaling in differentiated bone marrow cells and in a cell population enriched for leukemia stem cells. Cells expressing endogenous oncogenic K-Ras(G12D) remained dependent on the second messenger diacylglycerol for the efficient activation of Ras-ERK signaling. These data raise the unexpected possibility of therapeutically targeting proteins that function upstream of oncogenic Ras in cancer.


Assuntos
Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Substituição de Aminoácidos , Animais , Células Cultivadas , Citocinas/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Células-Tronco Hematopoéticas/patologia , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Mutação de Sentido Incorreto , Células-Tronco Neoplásicas/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfolipase C gama/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Sistemas do Segundo Mensageiro/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
9.
Cancer Res ; 71(5): 1573-83, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21363918

RESUMO

The mammalian target of rapamycin (mTOR) pathway is implicated widely in cancer pathophysiology. Dual inhibition of the mTOR kinase complexes mTORC1 and mTORC2 decreases tumor xenograft growth in vivo and VEGF secretion in vitro, but the relationship between these two effects are unclear. In this study, we examined the effects of mTORC1/2 dual inhibition on VEGF production, tumor angiogenesis, vascular regression, and vascular regrowth, and we compared the effects of dual inhibition to mTORC1 inhibition alone. ATP-competitive inhibitors OSI-027 and OXA-01 targeted both mTORC1 and mTORC2 signaling in vitro and in vivo, unlike rapamycin that only inhibited mTORC1 signaling. OXA-01 reduced VEGF production in tumors in a manner associated with decreased vessel sprouting but little vascular regression. In contrast, rapamycin exerted less effect on tumoral production of VEGF. Treatment with the selective VEGFR inhibitor OSI-930 reduced vessel sprouting and caused substantial vascular regression in tumors. However, following discontinuation of OSI-930 administration tumor regrowth could be slowed by OXA-01 treatment. Combining dual inhibitors of mTORC1 and mTORC2 with a VEGFR2 inhibitor decreased tumor growth more than either inhibitor alone. Together, these results indicate that dual inhibition of mTORC1/2 exerts antiangiogenic and antitumoral effects that are even more efficacious when combined with a VEGFR antagonist.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Proteínas/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Linhagem Celular Tumoral , Humanos , Imuno-Histoquímica , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Nus , Microscopia Confocal , Complexos Multiproteicos , Neoplasias Experimentais/metabolismo , Neovascularização Patológica/metabolismo , Quinolinas/farmacologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Tiofenos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Clin Cancer Res ; 13(19): 5926-34, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17908989

RESUMO

PURPOSE: In this study, we tested the hypothesis that inhibition of mitogen-activated protein kinase kinases (MKK) inhibits tumor growth by acting on angiogenic signaling and by extension may form the basis of an effective strategy for treatment of Kaposi's sarcoma. EXPERIMENTAL DESIGN: Murine endothelial cells expressing the human herpes virus 8 G protein-coupled receptor (vGPCR-SVEC) were treated with anthrax lethal toxin (LeTx). LeTx is a binary toxin ordinarily secreted by Bacillus anthracis and is composed of two proteins: protective antigen (the binding moiety) and lethal factor (the active moiety). Lethal factor is a protease that cleaves and inactivates MKKs. RESULTS: In vitro, treatment of vGPCR-SVEC with LeTx inhibited MKK signaling, moderately inhibited cell proliferation, and blocked the ability of these cells to form colonies in soft agar. Treatment with LeTx also blocked the ability of these cells to release several angioproliferative cytokines, notably vascular endothelial growth factor (VEGF). In contrast, inhibition of mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 with U0126 caused a substantial inhibition of proliferation but only modestly inhibited VEGF release. In xenograft models, i.v. injection of LeTx caused reduced tumor growth characterized immunohistochemically by inhibition of MKK signaling, decreased rates of proliferation, and reduced levels of VEGF and VEGF receptor 2, with a corresponding decrease in vascular density. CONCLUSIONS: These data support a role for MKK signaling in tumor growth and vascularization and are consistent with the hypothesis that inhibition of MKK signaling by LeTx or a similar agent may be an effective strategy for the treatment of Kaposi's sarcoma as well as other vascular tumors.


Assuntos
Antígenos de Bactérias/farmacologia , Toxinas Bacterianas/farmacologia , Células Endoteliais/metabolismo , Receptores de Quimiocinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Camundongos , Microcirculação , Células NIH 3T3 , Transplante de Neoplasias , Neovascularização Patológica , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/terapia , Transdução de Sinais , Fatores de Tempo
11.
Toxicol Appl Pharmacol ; 211(2): 97-105, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16039682

RESUMO

Alkylating agents play an important role in the chemotherapy of malignant melanomas. The activity of alkylating agents depends on their capacity to form alkyl adducts with DNA, in some cases causing cross-linking of DNA strands. However, the use of these agents is limited by cellular resistance induced by the DNA repair enzyme O(6)-methylguanine DNA-methyltransferase (MGMT) which removes alkyl groups from alkylated DNA strands. To determine to what extent the expression of MGMT in melanoma cells induces resistance to alkylating agents, the human cell line CAL77 Mer- (i.e., MGMT deficient) were transfected with pcMGMT vector containing human MGMT cDNA. Several clones expressing MGMT at a high level were selected to determine their sensitivity to chemotherapeutic drugs. Melanoma-transfected cells were found to be significantly less sensitive to nitrosoureas (carmustine, fotemustine, streptozotocin) and temozolomide with an increase of IC(50) values between 3 and 14 when compared to parent cells. No difference in cell survival rates between MGMT-proficient and -deficient cells was observed for melphalan, chlorambucil, busulphan, thiotepa and cisplatin which preferentially induce N(7) guanine lesions. Surprisingly, MGMT overexpression increased the sensitivity of CAL77 cells to mitomycin C by approximately 10-fold. Treatment of clonal cell lines with buthionine-[S,R]-sulfoximine (BSO), an inhibitor of gamma-glutamylcysteine synthetase which depletes cellular glutathione, completely reversed this unexpected increase in sensitivity to mitomycin C. This observation suggests that glutathione is involved in the sensitivity of MGMT-transfected cells to mitomycin C and may act synergistically with MGMT via an unknown mechanism.


Assuntos
Dacarbazina/análogos & derivados , Mitomicina/toxicidade , Compostos de Nitrosoureia/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/genética , Antineoplásicos/toxicidade , Butionina Sulfoximina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Dacarbazina/toxicidade , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glutationa/metabolismo , Humanos , Concentração Inibidora 50 , Melanoma/enzimologia , Melanoma/genética , Melanoma/patologia , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Temozolomida , Transfecção
12.
Mol Pharmacol ; 65(4): 897-905, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15044619

RESUMO

Previous studies have shown that glutathione S-transferases (GSTs) can operate in synergy with efflux transporters, multi-drug resistance proteins (MRPs), to confer resistance to several carcinogens, mutagens and anticancer drugs. To address the poorly documented role of the GSTM1 in cancer chemoresistance, we used CAL1 human melanoma cells expressing no endogenous GSTM1 and a high level of MRP1. Cells were transfected with an expression vector containing the GSTM1 cDNA, and different clones were selected expressing different levels of GSTM1 (RT-PCR, Western blot, and enzyme activity). Cells overexpressing GSTM1 displayed a 3- to 4-fold increase in resistance to anticancer drugs vincristine (VCR) and chlorambucil (CHB) in proliferation, cytotoxic, and clonogenic survival assays. Inhibitors of MRP1 (sulfinpyrazone, verapamil) and GST (dicumarol, curcumin) completely reversed the GSTM1-associated resistance to VCR, indicating that a MRP efflux function is necessary to potentiate GSTM1-mediated resistance to VCR. Conversely, MRP1 inhibitors had no effect on the sensitivity to CHB. Using immunofluorescence assay, GSTM1 was also shown to protect microtubule network integrity from VCR-induced inhibition of microtubule polymerization. In conclusion, these results show that GSTM1 alone is involved in melanoma resistance to CHB, whereas it can act in synergy with MRP1 to protect cells from toxic effects of VCR.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Antineoplásicos Fitogênicos/farmacologia , Glutationa Transferase/fisiologia , Vincristina/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Clorambucila/farmacologia , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Glutationa Transferase/metabolismo , Humanos , Melanoma/patologia , Microtúbulos/efeitos dos fármacos , Células Tumorais Cultivadas
13.
J Pharmacol Exp Ther ; 307(2): 816-23, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12970393

RESUMO

Fotemustine is a third generation chloroethylnitrosourea that has demonstrated significant antitumoral effects in malignant melanoma. However, its use is somewhat limited by its toxic side effects and chemoresistance caused by direct repair of O6-alkyl groups by the enzyme O6-methylguanine DNA-methyltransferase (MGMT). The aim of this work was to determine to what extent the expression of MGMT influences cytotoxicity, DNA damage, and apoptosis induced by new nitrososulfamide analogs of fotemustine (compounds 4 and 8), which have previously demonstrated interesting antiproliferative properties. We carried out complementary strategies that consisted of MGMT cDNA transfection in CAL77 Mer- melanoma cells and of MGMT inhibition with O6-benzylguanine (BG) in A375 Mer+ melanoma cells. MGMT-transfected cells were 7 to 9 times less sensitive to fotemustine than parent cells, whereas no difference between the transfected and parent cells was observed for nitrososulfamide analogs. The cytotoxicity of these analogs vis à vis a MGMT-proficient A375 melanoma cell line was approximately 3 times greater than that of fotemustine. Coincubation of these cells with O6-benzylguanine significantly increased the cytotoxicity of fotemustine and compound 8, whereas BG had little effect on the cytotoxicity of compound 4. Furthermore, DNA fragmentation determined by a comet assay was greater with nitrososulfamide analogs than with fotemustine. O6-benzylguanine increased DNA fragmentation for fotemustine and compound 8, but not for compound 4, which induced comets with a typical apoptotic appearance. The ability of this compound to induce apoptosis in the absence of BG was confirmed by a specific enzyme-linked immunosorbent assay apoptotic assay using a single-stranded DNA monoclonal antibody.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Dano ao DNA/efeitos dos fármacos , Guanina/análogos & derivados , Compostos de Nitrosoureia/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/biossíntese , Compostos Organofosforados/farmacologia , Antineoplásicos/química , Carmustina/farmacologia , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Guanina/farmacologia , Humanos , Melanoma/patologia , Compostos de Nitrosoureia/química , O(6)-Metilguanina-DNA Metiltransferase/genética , Compostos Organofosforados/química , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA