Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochemistry ; 59(4): 541-551, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31841311

RESUMO

Blocking interactions between PD-1 and PD-L1 opens a new era of cancer treatment involving immunity modulation. Although most immunotherapies use monoclonal antibodies, small-molecule inhibitors offer advantages. To facilitate development of small-molecule therapeutics, we implemented a rapid approach to characterize the binding interfaces of small-molecule inhibitors with PD-L1. We determined its interaction with a synthetic macrocyclic peptide by using two mass spectrometry-based approaches, hydrogen-deuterium exchange and fast photochemical oxidation of proteins (FPOP), and corroborated the findings with our X-ray structure of the PD-L1/macrocycle complex. Although all three approaches show that the macrocycle binds directly to PD-L1 over the regions of residues 46-87 and 114-125, the two protein footprinting approaches show additional binding at the N-terminus of PD-L1, and FPOP reveals some critical binding residues. The outcomes not only show the binding regions but also demonstrate the utility of MS-based footprinting in probing protein/ligand inhibitory interactions in cancer immunotherapy.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/química , Anticorpos Monoclonais/química , Antígeno B7-H1/metabolismo , Cristalografia por Raios X/métodos , Humanos , Imunoterapia , Ligantes , Compostos Macrocíclicos/química , Compostos Macrocíclicos/farmacologia , Espectrometria de Massas , Modelos Moleculares , Oxirredução , Peptídeos/química , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Pegadas de Proteínas/métodos , Bibliotecas de Moléculas Pequenas/farmacologia
2.
Nat Struct Mol Biol ; 23(12): 1101-1110, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27775709

RESUMO

Host and virus interactions occurring at the post-transcriptional level are critical for infection but remain poorly understood. Here, we performed comprehensive transcriptome-wide analyses revealing that human cytomegalovirus (HCMV) infection results in widespread alternative splicing (AS), shortening of 3' untranslated regions (3' UTRs) and lengthening of poly(A)-tails in host gene transcripts. We found that the host RNA-binding protein CPEB1 was highly induced after infection, and ectopic expression of CPEB1 in noninfected cells recapitulated infection-related post-transcriptional changes. CPEB1 was also required for poly(A)-tail lengthening of viral RNAs important for productive infection. Strikingly, depletion of CPEB1 reversed infection-related cytopathology and post-transcriptional changes, and decreased productive HCMV titers. Host RNA processing was also altered in herpes simplex virus-2 (HSV-2)-infected cells, thereby indicating that this phenomenon might be a common occurrence during herpesvirus infections. We anticipate that our work may serve as a starting point for therapeutic targeting of host RNA-binding proteins in herpesvirus infections.


Assuntos
Infecções por Citomegalovirus/genética , Citomegalovirus/genética , RNA Mensageiro/genética , RNA Viral/genética , Fatores de Transcrição/genética , Transcriptoma , Fatores de Poliadenilação e Clivagem de mRNA/genética , Regiões 3' não Traduzidas , Processamento Alternativo , Linhagem Celular , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Poliadenilação , Fatores de Transcrição/metabolismo , Regulação para Cima , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
3.
J Virol ; 90(3): 1259-77, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26559848

RESUMO

UNLABELLED: Human cytomegalovirus (HCMV) is the major viral cause of birth defects and a serious problem in immunocompromised individuals and has been associated with atherosclerosis. Previous studies have shown that the induction of autophagy can inhibit the replication of several different types of DNA and RNA viruses. The goal of the work presented here was to determine whether constitutive activation of autophagy would also block replication of HCMV. Most prior studies have used agents that induce autophagy via inhibition of the mTOR pathway. However, since HCMV infection alters the sensitivity of mTOR kinase-containing complexes to inhibitors, we sought an alternative method of inducing autophagy. We chose to use trehalose, a nontoxic naturally occurring disaccharide that is found in plants, insects, microorganisms, and invertebrates but not in mammals and that induces autophagy by an mTOR-independent mechanism. Given the many different cell targets of HCMV, we proceeded to determine whether trehalose would inhibit HCMV infection in human fibroblasts, aortic artery endothelial cells, and neural cells derived from human embryonic stem cells. We found that in all of these cell types, trehalose induces autophagy and inhibits HCMV gene expression and production of cell-free virus. Treatment of HCMV-infected neural cells with trehalose also inhibited production of cell-associated virus and partially blocked the reduction in neurite growth and cytomegaly. These results suggest that activation of autophagy by the natural sugar trehalose or other safe mTOR-independent agents might provide a novel therapeutic approach for treating HCMV disease. IMPORTANCE: HCMV infects multiple cell types in vivo, establishes lifelong persistence in the host, and can cause serious health problems for fetuses and immunocompromised individuals. HCMV, like all other persistent pathogens, has to finely tune its interplay with the host cellular machinery to replicate efficiently and evade detection by the immune system. In this study, we investigated whether modulation of autophagy, a host pathway necessary for the recycling of nutrients and removal of protein aggregates, misfolded proteins, and pathogens, could be used to target HCMV. We found that autophagy could be significantly increased by treatment with the nontoxic, natural disaccharide trehalose. Importantly, trehalose had a profound inhibitory effect on viral gene expression and strongly impaired viral spread. These data constitute a proof-of-concept for the use of natural products targeting host pathways rather than the virus itself, thus reducing the risk of the development of resistance to treatment.


Assuntos
Autofagia/efeitos dos fármacos , Citomegalovirus/fisiologia , Trealose/metabolismo , Replicação Viral , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Humanos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/virologia
4.
PLoS One ; 9(2): e89195, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24558487

RESUMO

HIV viral protein R (Vpr) induces a cell cycle arrest at the G2/M phase by activating the ATR DNA damage/replication stress signalling pathway through engagement of the DDB1-CUL4A-DCAF1 E3 ubiquitin ligase via a direct binding to the substrate specificity receptor DCAF1. Since no high resolution structures of the DDB1-DCAF1-Vpr substrate recognition module currently exist, we used a mutagenesis approach to better define motifs in DCAF1 that are crucial for Vpr and DDB1 binding. Herein, we show that the minimal domain of DCAF1 that retained the ability to bind Vpr and DDB1 was mapped to residues 1041 to 1393 (DCAF1 WD). Mutagenic analyses identified an α-helical H-box motif and F/YxxF/Y motifs located in the N-terminal domain of DCAF1 WD that are involved in exclusive binding to DDB1. While we could not identify elements specifically involved in Vpr binding, overall, the mutagenesis data suggest that the predicted ß-propeller conformation of DCAF1 is likely to be critical for Vpr association. Importantly, we provide evidence that binding of Vpr to DCAF1 appears to modulate the formation of a DDB1/DCAF1 complex. Lastly, we show that expression of DCAF1 WD in the absence of endogenous DCAF1 was not sufficient to enable Vpr-mediated G2 arrest activity. Overall, our results reveal that Vpr and DDB1 binding on DCAF1 can be genetically separated and further suggest that DCAF1 contains determinants in addition to the Vpr and DDB1 minimal binding domain, which are required for Vpr to enable the induction of a G2 arrest.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/fisiologia , Infecções por HIV/metabolismo , Complexos Multiproteicos/metabolismo , Transdução de Sinais/fisiologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Sequência de Aminoácidos , Western Blotting , Proteínas de Transporte/genética , Proteínas Culina/metabolismo , Proteínas de Ligação a DNA/genética , Células HEK293 , Células HeLa , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Plasmídeos/genética , Ligação Proteica , Proteínas Serina-Treonina Quinases , Ubiquitina-Proteína Ligases/metabolismo
5.
J Virol ; 88(8): 4021-39, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24453373

RESUMO

UNLABELLED: Congenital human cytomegalovirus (HCMV) infection is a major cause of central nervous system structural anomalies and sensory impairments. It is likely that the stage of fetal development, as well as the state of differentiation of susceptible cells at the time of infection, affects the severity of the disease. We used human embryonic stem (ES) cell-derived primitive prerosette neural stem cells (pNSCs) and neural progenitor cells (NPCs) maintained in chemically defined conditions to study HCMV replication in cells at the early stages of neural development. In contrast to what was observed previously using fetus-derived NPCs, infection of ES cell-derived pNSCs with HCMV was nonprogressive. At a low multiplicity of infection, we observed only a small percentage of cells expressing immediate-early genes (IE) and early genes. IE expression was found to be restricted to cells negative for the anterior marker FORSE-1, and treatment of pNSCs with retinoic acid restored IE expression. Differentiation of pNSCs into NPCs restored IE expression but not the transactivation of early genes. Virions produced in NPCs and pNSCs were exclusively cell associated and were mostly non-neural tropic. Finally, we found that viral genomes could persist in pNSC cultures for up to a month after infection despite the absence of detectable IE expression by immunofluorescence, and infectious virus could be produced upon differentiation of pNSCs to neurons. In conclusion, our results highlight the complex array of hurdles that HCMV must overcome in order to infect primitive neural stem cells and suggest that these cells might act as a reservoir for the virus. IMPORTANCE: Human cytomegalovirus (HCMV) is a betaherpesvirus that is highly prevalent in the population. HCMV infection is usually asymptomatic but can lead to severe consequences in immunosuppressed individuals. HCMV is also the most important infectious cause of congenital developmental birth defects. Manifestations of fetal HCMV disease range from deafness and learning disabilities to more severe symptoms such as microcephaly. In this study, we have used embryonic stem cells to generate primitive neural stem cells and have used these to model HCMV infection of the fetal central nervous system (CNS) in vitro. Our results reveal that these cells, which are similar to those present in the developing neural tube, do not support viral replication but instead likely constitute a viral reservoir. Future work will define the effect of viral persistence on cellular functions as well as the exogenous signals leading to the reactivation of viral replication in the CNS.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , DNA Viral/metabolismo , Células-Tronco Embrionárias/virologia , Células-Tronco Neurais/virologia , Diferenciação Celular , Citomegalovirus/genética , Infecções por Citomegalovirus/fisiopatologia , DNA Viral/genética , Células-Tronco Embrionárias/citologia , Feminino , Humanos , Células-Tronco Neurais/citologia , Replicação Viral
6.
PLoS Pathog ; 6(9): e1001080, 2010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20824083

RESUMO

HIV-1 Viral protein R (Vpr) induces a cell cycle arrest at the G2/M phase by activating the ATR DNA damage/stress checkpoint. Recently, we and several other groups showed that Vpr performs this activity by recruiting the DDB1-CUL4A (VPRBP) E3 ubiquitin ligase. While recruitment of this E3 ubiquitin ligase complex has been shown to be required for G2 arrest, the subcellular compartment where this complex forms and functionally acts is unknown. Herein, using immunofluorescence and confocal microscopy, we show that Vpr forms nuclear foci in several cell types including HeLa cells and primary CD4+ T-lymphocytes. These nuclear foci contain VPRBP and partially overlap with DNA repair foci components such as gamma-H2AX, 53BP1 and RPA32. While treatment with the non-specific ATR inhibitor caffeine or depletion of VPRBP by siRNA did not inhibit formation of Vpr nuclear foci, mutations in the C-terminal domain of Vpr and cytoplasmic sequestration of Vpr by overexpression of Gag-Pol resulted in impaired formation of these nuclear structures and defective G2 arrest. Consistently, we observed that G2 arrest-competent sooty mangabey Vpr could form these foci but not its G2 arrest-defective paralog Vpx, suggesting that formation of Vpr nuclear foci represents a critical early event in the induction of G2 arrest. Indeed, we found that Vpr could associate to chromatin via its C-terminal domain and that it could form a complex with VPRBP on chromatin. Finally, analysis of Vpr nuclear foci by time-lapse microscopy showed that they were highly mobile and stable structures. Overall, our results suggest that Vpr recruits the DDB1-CUL4A (VPRBP) E3 ligase to these nuclear foci and uses these mobile structures to target a chromatin-bound cellular substrate for ubiquitination in order to induce DNA damage/replication stress, ultimately leading to ATR activation and G2 cell cycle arrest.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/fisiologia , Cromatina/metabolismo , Fase G2/fisiologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Adulto , Proteínas Mutadas de Ataxia Telangiectasia , Western Blotting , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Cromatina/genética , Reparo do DNA , Células HeLa , Humanos , Imunoprecipitação , Proteínas Serina-Treonina Quinases/genética , Ubiquitina-Proteína Ligases , Ubiquitinação , Replicação Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
7.
J Virol ; 84(7): 3320-30, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20089662

RESUMO

HIV-1 viral protein R (Vpr) induces cell cycle arrest at the G(2)/M phase by a mechanism involving the activation of the DNA damage sensor ATR. We and others recently showed that Vpr performs this function by subverting the activity of the DDB1-CUL4A (VPRBP) E3 ubiquitin ligase. Vpr could thus act as a connector between the E3 ligase and an unknown cellular factor whose ubiquitination would induce G(2) arrest. While attractive, this model is based solely on the indirect observation that some mutants of Vpr retain their interaction with the E3 ligase but fail to induce G(2) arrest. Using a tandem affinity purification approach, we observed that Vpr interacts with ubiquitinated cellular proteins and that this association requires the recruitment of an active E3 ligase given that the depletion of VPRBP by RNA interference or the overexpression of a dominant negative mutant of CUL4A decreased this association. Importantly, G(2)-arrest-defective mutants of Vpr in the C-terminal putative substrate-interacting domain displayed a decreased association with ubiquitinated proteins. We also found that the inhibition of proteasomal activity increased this association and that the ubiquitin chains were at least in part constituted of classical K48 linkages. Interestingly, the inhibition of K48 polyubiquitination specifically impaired the Vpr-induced phosphorylation of H2AX, an early target of ATR, but did not affect UV-induced H2AX phosphorylation. Overall, our results provide direct evidence that the association of Vpr with the DDB1-CUL4A (VPRBP) E3 ubiquitin ligase induces the K48-linked polyubiquitination of as-yet-unknown cellular proteins, resulting in their proteasomal degradation and ultimately leading to the activation of ATR and G(2) arrest.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Fase G2 , Poliubiquitina/química , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Ubiquitina/química , Ubiquitinação , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/fisiologia , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Transporte/metabolismo , Linhagem Celular , Proteínas Culina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Humanos , Fosforilação , Ubiquitina-Proteína Ligases/fisiologia
8.
Blood ; 115(7): 1354-63, 2010 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-20008788

RESUMO

HIV up-regulates cell-surface expression of specific ligands for the activating NKG2D receptor, including ULBP-1, -2, and -3, but not MICA or MICB, in infected cells both in vitro and in vivo. However, the viral factor(s) involved in NKG2D ligand expression still remains undefined. HIV-1 Vpr activates the DNA damage/stress-sensing ATR kinase and promotes G(2) cell-cycle arrest, conditions known to up-regulate NKG2D ligands. We report here that HIV-1 selectively induces cell-surface expression of ULBP-2 in primary CD4(+) T lymphocytes by a process that is Vpr dependent. Importantly, Vpr enhanced the susceptibility of HIV-1-infected cells to NK cell-mediated killing. Strikingly, Vpr alone was sufficient to up-regulate expression of all NKG2D ligands and thus promoted efficient NKG2D-dependent NK cell-mediated killing. Delivery of virion-associated Vpr via defective HIV-1 particles induced analogous biologic effects in noninfected target cells, suggesting that Vpr may act similarly beyond infected cells. All these activities relied on Vpr ability to activate the ATR-mediated DNA damage/stress checkpoint. Overall, these results indicate that Vpr is a key determinant responsible for HIV-1-induced up-regulation of NKG2D ligands and further suggest an immunomodulatory role for Vpr that may not only contribute to HIV-1-induced CD4(+) T-lymphocyte depletion but may also take part in HIV-1-induced NK-cell dysfunction.


Assuntos
Infecções por HIV/imunologia , HIV-1/imunologia , Células Matadoras Naturais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/imunologia , Proteínas Mutadas de Ataxia Telangiectasia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Proteínas de Ciclo Celular/imunologia , Proteínas de Ciclo Celular/metabolismo , Morte Celular/imunologia , Proteínas Ligadas por GPI , Infecções por HIV/patologia , Infecções por HIV/virologia , HIV-1/genética , Células HeLa , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Rim/citologia , Células Matadoras Naturais/citologia , Lentivirus/genética , Ligantes , Mutagênese Sítio-Dirigida , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Plasmídeos , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Estresse Fisiológico/imunologia , Regulação para Cima/imunologia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo
9.
PLoS Pathog ; 3(7): e85, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17630831

RESUMO

Human immunodeficiency virus type 1 (HIV-1) viral protein R (Vpr) has been shown to cause G2 cell cycle arrest in human cells by inducing ATR-mediated inactivation of p34cdc2, but factors directly engaged in this process remain unknown. We used tandem affinity purification to isolate native Vpr complexes. We found that damaged DNA binding protein 1 (DDB1), viral protein R binding protein (VPRBP), and cullin 4A (CUL4A)--components of a CUL4A E3 ubiquitin ligase complex, DDB1-CUL4A(VPRBP)--were able to associate with Vpr. Depletion of VPRBP by small interfering RNA impaired Vpr-mediated induction of G2 arrest. Importantly, VPRBP knockdown alone did not affect normal cell cycle progression or activation of ATR checkpoints, suggesting that the involvement of VPRBP in G2 arrest was specific to Vpr. Moreover, leucine/isoleucine-rich domain Vpr mutants impaired in their ability to interact with VPRBP and DDB1 also produced strongly attenuated G2 arrest. In contrast, G2 arrest-defective C-terminal Vpr mutants were found to maintain their ability to associate with these proteins, suggesting that the interaction of Vpr with the DDB1-VPRBP complex is necessary but not sufficient to block cell cycle progression. Overall, these results point toward a model in which Vpr could act as a connector between the DDB1-CUL4A(VPRBP) E3 ubiquitin ligase complex and an unknown cellular factor whose proteolysis or modulation of activity through ubiquitination would activate ATR-mediated checkpoint signaling and induce G2 arrest.


Assuntos
Proteínas Culina/metabolismo , Proteínas de Ligação a DNA/metabolismo , HIV-1/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Animais , Células Cultivadas , Fase G2/fisiologia , Regulação Viral da Expressão Gênica , Inativação Gênica , Cabras , Humanos , Camundongos , Interferência de RNA , RNA Interferente Pequeno/genética , Coelhos , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Replicação Viral , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética
10.
Curr Gene Ther ; 6(1): 111-23, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16475949

RESUMO

Enhanced DNA repair in many cancer cells can be correlated to the resistance to cancer treatment, and thus contributes to a poor prognosis. Ionizing radiation and many anti-cancer drugs induce DNA double-strand breaks (DSBs), which are usually regarded as the most toxic types of DNA damages. Repair of DNA DSBs is vital for maintaining genomic stability and hence crucial for survival and propagation of all cellular organisms. Therefore, reducing the capacity of cancer cells to repair DSBs could sensitize tumors to radio/chemotherapy. Many investigators have used gene therapy strategies to down-regulate or inactivate proteins involved in the repair of DSBs in order to reduce the survival of cancer cells. Herein, are reviewed several protein candidates that have been targeted by different gene therapy approaches. Results obtained from in vitro and in vivo experiments are presented and discussed in the perspective of potential gene therapy clinical trials.


Assuntos
Enzimas Reparadoras do DNA/genética , Reparo do DNA , Proteínas de Ligação a DNA/genética , Marcação de Genes , Terapia Genética/tendências , Neoplasias/terapia , Animais , Terapia Genética/métodos , Humanos , Neoplasias/genética
11.
Mol Cancer Ther ; 3(12): 1525-32, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15634645

RESUMO

Recent studies suggest that erythropoietin plays an important role in the process of neoplastic transformation and malignant phenotype progression observed in malignancy. To study the role of erythropoietin and its receptor (EPOR) on the response of cancer cells in vitro, we used two solid tumor cell lines, namely the human malignant glioma cell line U87 and the primary cervical cancer cell line HT100. All experiments were done with heat-inactivated fetal bovine serum in order to inactivate any endogenous bovine erythropoietin. The expression of the EPOR in these cells was confirmed with immunoblot techniques. The addition of exogenous recombinant human erythropoietin (rhEPO) induces the cancer cells to become more resistant to ionizing radiation and to cisplatin. Furthermore, this rhEPO-induced resistance to ionizing radiation and to cisplatin was reversed by the addition of tyrphostin (AG490), an inhibitor of JAK2. Our findings indicate that rhEPO result in a significant, JAK2-dependent, in vitro resistance to ionizing radiation and to cisplatin in the human cancer cells lines studied in this report.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Eritropoetina/uso terapêutico , Glioma/patologia , Tolerância a Radiação , Neoplasias do Colo do Útero/patologia , Animais , Bovinos , Eritropoetina/metabolismo , Feminino , Glioma/tratamento farmacológico , Glioma/radioterapia , Humanos , Janus Quinase 2 , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Radiação Ionizante , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos da radiação , Tirfostinas/farmacologia , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/radioterapia
12.
Cancer Gene Ther ; 10(8): 637-46, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12872145

RESUMO

We have constructed a retroviral bicistronic vector, MFG/GID, that transduces the expression of both the A3 isoform of the rat glutathione S-transferase (GST A3), and the tyr-22 variant of the human dihydrofolate reductase (DHFR(L22Y)). Transduction of murine 3T3 fibroblasts with this vector increased their in vitro resistance to chlorambucil (1.8-fold) and trimetrexate (TMTX) (748-fold). TMTX selection of a mixed population of 20% GID-transduced NIH 3T3 cells and 80% control cells resulted in a marked increase in the GST peroxidase activity associated with the GST A3 isoform (17.7-fold). MFG/GID-transduced primary clonogenic murine hematopoietic progenitor cells were likewise more resistant to TMTX and chlorambucil than control beta-gal-transduced cells. Selecting GID-transduced hematopoietic cells with a combination of TMTX and a nucleoside transport inhibitor resulted in a marked increase in resistance upon re-exposure to TMTX (99% survival). Similarly, GID-transduced hematopoietic cells selected with TMTX were more resistant to chlorambucil, with 40% survival at a drug concentration that killed practically all control cells. These results suggest that antifolate-mediated selection of MFG/GID-transduced hematopoietic cells could be used as a mean to enrich the population of transduced cells prior to or following transplantation, thus potentially conferring in vivo chemoprotection to nitrogen mustards and antifolates.


Assuntos
Resistência a Medicamentos , Antagonistas do Ácido Fólico/farmacologia , Glutationa Transferase/genética , Compostos de Mostarda Nitrogenada/farmacologia , Retroviridae/genética , Tetra-Hidrofolato Desidrogenase/genética , Animais , Antineoplásicos Alquilantes/farmacologia , Fibroblastos/efeitos dos fármacos , Antagonistas do Ácido Fólico/efeitos adversos , Técnicas de Transferência de Genes , Vetores Genéticos , Glutationa Transferase/metabolismo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Camundongos , Células NIH 3T3 , Compostos de Mostarda Nitrogenada/efeitos adversos , Isoformas de Proteínas/química , Ratos , Tetra-Hidrofolato Desidrogenase/metabolismo , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...