Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Host Microbe ; 23(5): 628-635.e7, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29746834

RESUMO

BK polyomavirus (BKV) frequently causes nephropathy (BKVN) in kidney transplant recipients (KTRs). BKV has also been implicated in the etiology of bladder and kidney cancers. We characterized BKV variants from two KTRs who developed BKVN followed by renal carcinoma. Both patients showed a swarm of BKV sequence variants encoding non-silent mutations in surface loops of the viral major capsid protein. The temporal appearance and disappearance of these mutations highlights the intra-patient evolution of BKV. Some of the observed mutations conferred resistance to antibody-mediated neutralization. The mutations also modified the spectrum of receptor glycans engaged by BKV during host cell entry. Intriguingly, all observed mutations were consistent with DNA damage caused by antiviral APOBEC3 cytosine deaminases. Moreover, APOBEC3 expression was evident upon immunohistochemical analysis of renal biopsies from KTRs. These results provide a snapshot of in-host BKV evolution and suggest that APOBEC3 may drive BKV mutagenesis in vivo.


Assuntos
Vírus BK/genética , Citosina Desaminase/fisiologia , Transplante de Rim , Infecções por Polyomavirus/virologia , Infecções Tumorais por Vírus/virologia , Desaminases APOBEC , Adulto , Substituição de Aminoácidos , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Vírus BK/imunologia , Proteínas do Capsídeo/genética , Linhagem Celular , Mapeamento Cromossômico , Citidina Desaminase , Dano ao DNA , DNA Viral/análise , DNA Viral/genética , Feminino , Células HEK293 , Humanos , Itália , Nefropatias/patologia , Nefropatias/virologia , Masculino , Pessoa de Meia-Idade , Mutação , Infecções por Polyomavirus/sangue , Infecções por Polyomavirus/imunologia , Infecções por Polyomavirus/patologia , Infecções Tumorais por Vírus/sangue , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia
2.
J Leukoc Biol ; 94(2): 337-41, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23744645

RESUMO

The RIG-I signaling pathway is critical in the activation of the type I IFN-dependent antiviral innate-immune response. We thus examined whether RIG-I activation can inhibit HIV replication in macrophages. We showed that the stimulation of monocyte-derived macrophages with 5'ppp-dsRNA, a synthetic ligand for RIG-I, induced the expression of RIG-I, IFN-α/ß, and several IRFs, key regulators of the IFN signaling pathway. In addition, RIG-I activation induced the expression of multiple intracellular HIV-restriction factors, including ISGs, several members of the APOBEC3 family, tetherin and CC chemokines, the ligands for HIV entry coreceptor (CCR5). The inductions of these factors were associated with the inhibition of HIV replication in macrophages stimulated by 5'ppp-dsRNA. These observations highlight the importance of RIG-I signaling in macrophage innate immunity against HIV, which can be beneficial for the treatment of HIV disease, where intracellular immune defense is compromised by the virus.


Assuntos
RNA Helicases DEAD-box/fisiologia , HIV-1/fisiologia , Macrófagos/virologia , Replicação Viral , Desaminases APOBEC , Antígenos CD/fisiologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Células Cultivadas/virologia , Quimiocinas/biossíntese , Quimiocinas/genética , Citidina Desaminase , Citosina Desaminase/fisiologia , Proteína DEAD-box 58 , Proteínas Ligadas por GPI/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Transcriptase Reversa do HIV/análise , HIV-1/enzimologia , Humanos , Imunidade Inata , Interferon-alfa/biossíntese , Interferon-alfa/genética , Interferon beta/biossíntese , Interferon beta/genética , Ligantes , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Monócitos/citologia , RNA de Cadeia Dupla/imunologia , RNA Viral/imunologia , Receptores CCR5/fisiologia , Receptores Imunológicos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Replicação Viral/efeitos dos fármacos
3.
Mol Cell ; 49(4): 632-44, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23333304

RESUMO

The HIV-1 accessory protein Vif hijacks a cellular Cullin-RING ubiquitin ligase, CRL5, to promote degradation of the APOBEC3 (A3) family of restriction factors. Recently, the cellular transcription cofactor CBFß was shown to form a complex with CRL5-Vif and to be essential for A3 degradation and viral infectivity. We now demonstrate that CBFß is required for assembling a well-ordered CRL5-Vif complex by inhibiting Vif oligomerization and by activating CRL5-Vif via direct interaction. The CRL5-Vif-CBFß holoenzyme forms a well-defined heterohexamer, indicating that Vif simultaneously hijacks CRL5 and CBFß. Heterodimers of CBFß and RUNX transcription factors contribute toward the regulation of genes, including those with immune system functions. We show that binding of Vif to CBFß is mutually exclusive with RUNX heterodimerization and impacts the expression of genes whose regulatory domains are associated with RUNX1. Our results provide a mechanism by which a pathogen with limited coding capacity uses one factor to hijack multiple host pathways.


Assuntos
Fator de Ligação a CCAAT/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Citosina Desaminase/metabolismo , Regulação da Expressão Gênica , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Desaminases APOBEC , Sequência de Aminoácidos , Sequência de Bases , Fator de Ligação a CCAAT/química , Fator de Ligação a CCAAT/fisiologia , Sequência Consenso , Subunidade alfa 2 de Fator de Ligação ao Core/química , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Citidina Desaminase , Citosina Desaminase/química , Citosina Desaminase/fisiologia , Expressão Gênica , Genes Reporter , Células HEK293 , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Estrutura Quaternária de Proteína , Linfócitos T/metabolismo , Linfócitos T/virologia , Ubiquitinação , Produtos do Gene vif do Vírus da Imunodeficiência Humana/química , Produtos do Gene vif do Vírus da Imunodeficiência Humana/fisiologia
4.
Uirusu ; 62(1): 27-38, 2012 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-23189822

RESUMO

Recent in vivo findings clearly indicate that mammalian cytidine deaminase APOBEC3 can function as a physiological restriction factor to retrotransposons and infectious retroviruses. However, some retroviruses, including primate lentiviruses, have evolved to counter their natural host's APOBEC3. To survive this arms race, primates seem to have acquired multiple copies of APOBEC3 genes. Surprisingly, however, during the process of the diversification of rodent species, as well as the human race, some ancestral individuals acquired genetic variants that reduced the protein levels of APOBEC3 expression, and these variants currently show unexpectedly wide geographic distributions. These data suggest that in the absence of a heavy burden of infectious retroviruses, high-level expression of APOBEC3 cytidine deaminase might be costly to the integrity of the host genome.


Assuntos
Citidina Desaminase/fisiologia , Citosina Desaminase/fisiologia , Evolução Molecular , Retroviridae/fisiologia , Replicação Viral , Desaminases APOBEC , Animais , Citidina Desaminase/genética , Citosina Desaminase/genética , Duplicação Gênica , Genoma/genética , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Polimorfismo Genético , Retroviridae/patogenicidade
5.
Uirusu ; 61(1): 67-72, 2011 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-21972557

RESUMO

Human cells developed the defense systems against retrovirus infections during the evolutions. These systems include retroviral restrictions by DNA cytidine deaminases of APOBEC3 family (A, B, C, DE, F, G, and H), which are potent factors to block the viral replication by blocking reverse transcription and/or integration and by hypermutating viral cDNA. In case of HIV-1, the viral protein, Vif abrogates the APOBEC3F/G function through specific machinery of ubiquitination and proteasomal degradation. Without Vif, APOBEC3F/G are incorporated into virus particles and block reverse transcription and/or integration in a newly infected cell. Recent advances in our understanding about biochemical and structure-biological characteristics of the enzymes provide new insights to reveal more detailed molecular mechanisms for anti-retroviral activity by APOBEC3 family. Here I briefly review how APOBEC3 proteins block retrovirus replications, focusing on APOBEC3G.


Assuntos
Antirretrovirais , Citosina Desaminase/farmacologia , Citosina Desaminase/fisiologia , Retroviridae/genética , Retroviridae/fisiologia , Replicação Viral/genética , Desaminases APOBEC , Desaminase APOBEC-3G , Citidina Desaminase , Citosina Desaminase/química , DNA Complementar/genética , DNA Viral/genética , HIV-1 , Humanos , Família Multigênica , Mutação , Transcrição Reversa , Ubiquitinação , Integração Viral , Produtos do Gene vif do Vírus da Imunodeficiência Humana/fisiologia
6.
Cell Host Microbe ; 8(6): 534-43, 2010 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-21147467

RESUMO

Viruses, including retroviruses like human immunodeficiency virus (HIV) and mouse mammary tumor virus (MMTV), are transmitted from mother to infants through milk. Lymphoid cells and antibodies are thought to provide mammary gland and milk-borne immunity. In contrast, little is known about the role of mammary epithelial cells (MECs). The APOBEC3 family of retroviral restriction factors is highly expressed in macrophages and lymphoid and dendritic cells. We now show that APOBEC3 proteins are also expressed in mouse and human MECs. Lymphoid cell-expressed APOBEC3 restricts in vivo spread of MMTV to lymphoid and mammary tissue. In contrast, mammary gland-expressed APOBEC3 is packaged into MMTV virions and decreases the infectivity of milk-borne viruses. Moreover, APOBEC3G and other APOBEC3 genes are expressed in human mammary cells and have the potential to restrict viruses produced in this cell type. These data point to a role for APOBEC3 proteins in limiting infectivity of milk-transmitted viruses.


Assuntos
Citidina Desaminase/fisiologia , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/metabolismo , Vírus do Tumor Mamário do Camundongo/fisiologia , Leite/virologia , Infecções por Retroviridae/metabolismo , Desaminases APOBEC , Animais , Células Cultivadas , Citidina Desaminase/biossíntese , Citosina Desaminase/biossíntese , Citosina Desaminase/fisiologia , Células Epiteliais/virologia , Feminino , HIV-1/patogenicidade , Humanos , Transmissão Vertical de Doenças Infecciosas , Linfócitos/metabolismo , Glândulas Mamárias Animais/virologia , Glândulas Mamárias Humanas/virologia , Vírus do Tumor Mamário do Camundongo/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/virologia , Infecções por Retroviridae/transmissão , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/transmissão , Infecções Tumorais por Vírus/virologia , Vírion/fisiologia , Montagem de Vírus , Produtos do Gene vif do Vírus da Imunodeficiência Humana/fisiologia
7.
Mol Ther ; 18(5): 921-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20179680

RESUMO

To target chemotherapy to tumor vascular endothelial cells (TVECs), we created the AdTie2RprCDFib(knob-RGD+) vector by inserting into an AdEasy adenoviral vector (Ad) backbone: (i) the cytosine deaminase (CD) gene driven by the Tie2 receptor promoter (Tie2Rpr) into the E1 region of Ad; (ii) mutations that reduce binding of the fiber knob to the Coxsackie adenovirus receptor (CAR); and (iii) the RGD peptide into the H1 loop of fiber for binding to the alpha(V)beta(3) integrin receptors on TVECs. To reduce uptake of the AdTie2RprCDFib(knob-RGD+) by reticuloendothelial (RE) and liver cells, we intravenously (i.v.) injected Hetastarch and low-dose Ad (one million vector particles (VPs)) prior to i.v. injection of a therapeutic dose (one billion VPs) of the AdTie2RprCDFib(knob-RGD+) vector. This treatment induced regressions of N202 breast cancer and B16 melanoma without toxicity to normal tissues. We showed that the tumor regression was induced by infection of the TVECs and not by the infection of tumor cells by the AdTie2RprCDFib(knob-RGD+) vector.


Assuntos
Neoplasias da Mama/terapia , Células Endoteliais/metabolismo , Melanoma/terapia , Adenoviridae/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Citosina Desaminase/genética , Citosina Desaminase/fisiologia , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Melanoma Experimental/terapia , Camundongos , Camundongos Nus , Microscopia Confocal , Regiões Promotoras Genéticas/genética , Receptor TIE-2/genética
8.
Mol Ther ; 18(1): 223-31, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19844197

RESUMO

The ability of human adipose tissue-derived mesenchymal stem cells (AT-MSCs), engineered to express the suicide gene cytosine deaminase::uracil phosphoribosyltransferase (CD::UPRT), to convert the relatively nontoxic 5-fluorocytosine (5-FC) into the highly toxic antitumor 5-fluorouracil (5-FU) together with their ability to track and engraft into tumors and micrometastases makes these cells an attractive tool to activate prodrugs directly within the tumor mass. In this study, we tested the feasibility and efficacy of these therapeutic cells to function as cellular vehicles of prodrug-activating enzymes in prostate cancer (PC) therapy. In in vitro migration experiments we have shown that therapeutic AT-MSCs migrated to all the prostate cell lines tested. In a pilot preclinical study, we observed that coinjections of human bone metastatic PC cells along with the transduced AT-MSCs into nude mice treated with 5-FC induced a complete tumor regression in a dose dependent manner or did not even allow the establishment of the tumor. More importantly, we also demonstrated that the therapeutic cells were effective in significantly inhibiting PC tumor growth after intravenous administration that is a key requisite for any clinical application of gene-directed enzyme prodrug therapies.


Assuntos
Citosina Desaminase/fisiologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Pentosiltransferases/fisiologia , Neoplasias da Próstata/terapia , Animais , Linhagem Celular Tumoral , Citosina Desaminase/genética , Flucitosina/farmacologia , Fluoruracila/farmacologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Pentosiltransferases/genética , Neoplasias da Próstata/induzido quimicamente
9.
Vet Immunol Immunopathol ; 134(1-2): 14-24, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19896724

RESUMO

The interplay between viral and cellular factors determines the outcome of an initial contact between a given virus and its natural host or upon encounter of a novel host. Thus, the potential of inducing disease as well as crossing host species barriers are the consequences of the molecular interactions between the parasite and its susceptible, tolerant or resistant host. Cellular restriction factors, for instance APOBEC3 and TRIM5 proteins, targeting defined pathogens or groups of pathogens as well as viral genes counter-acting these cellular defense systems are of prime importance in this respect and may even represent novel targets for prevention and therapy of virus infections. Due to the importance of host-encoded antiviral restriction and viral counter-defense for pathogenicity and host tropism, the responsible molecular factors and mechanisms are currently under intense investigation. In this review we will introduce host restriction and retroviral counter-defense systems with a special emphasis on the cat and its naturally occurring exogenous retroviruses which is a valid model for human disease, a model that will contribute to increase our basic understanding and potential applications of these important aspects of host-virus interaction.


Assuntos
Proteínas de Transporte/fisiologia , Doenças do Gato/virologia , Citosina Desaminase/fisiologia , Infecções por Retroviridae/veterinária , Retroviridae/fisiologia , Animais , Gatos/virologia , Interações Hospedeiro-Patógeno/fisiologia , Vírus da Imunodeficiência Felina/fisiologia , Lentivirus Felinos/fisiologia , Vírus da Leucemia Felina/fisiologia , Infecções por Retroviridae/virologia , Integração Viral/fisiologia , Replicação Viral/fisiologia
10.
Cancer Gene Ther ; 17(5): 299-306, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19893595

RESUMO

Previously, we have shown that the genetically modified human neural stem cells (NSCs) show remarkable migratory and tumor-tropic capability to track down brain tumor cells and deliver therapeutic agents with significant therapeutic benefit. Human NSCs that were retrovirally transduced with cytosine deaminase (CD) gene showed remarkable 'bystander killer effect' on the glioma cells after application of the prodrug, 5-fluorocytosine (5-FC). Interferon-beta (IFN-beta) is known for its antiproliferative effects in a variety of cancers. In our pilot clinical trial in glioma, the IFN-beta gene has shown potent antitumor activity in patients with malignant glioma. In the present study, we sought to examine whether human NSCs genetically modified to express both CD and IFN-beta genes intensified antitumor effect on experimental glioma. In vitro studies showed that CD/IFN-beta-expressing NSCs exerted a remarkable bystander effect on human glioma cells after the application of 5-FC, as compared with parental NSCs and CD-expressing NSCs. In animal models with human glioma orthotopic xenograft, intravenously infused CD/IFN-beta-expressing NSCs produced striking antitumor effect after administration of the prodrug 5-FC. Furthermore, the same gene therapy regimen prolonged survival periods significantly in the experimental animals. The results of the present study indicate that the multimodal NSC-based treatment strategy might have therapeutic potential against gliomas.


Assuntos
Citosina Desaminase/fisiologia , Terapia Genética/métodos , Glioma/tratamento farmacológico , Glioma/terapia , Interferon beta/fisiologia , Animais , Efeito Espectador , Linhagem Celular Tumoral , Citosina Desaminase/genética , Modelos Animais de Doenças , Feminino , Flucitosina/uso terapêutico , Humanos , Interferon beta/genética , Camundongos , Camundongos Nus
11.
Molecules ; 14(11): 4517-45, 2009 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-19924084

RESUMO

Gene directed enzyme prodrug therapy (GDEPT) of cancer aims to improve the selectivity of chemotherapy by gene transfer, thus enabling target cells to convert nontoxic prodrugs to cytotoxic drugs. A zone of cell kill around gene-modified cells due to transfer of toxic metabolites, known as the bystander effect, leads to tumour regression. Here we discuss the implications of either striving for a strong bystander effect to overcome poor gene transfer, or avoiding the bystander effect to reduce potential systemic effects, with the aid of three successful GDEPT systems. This review concentrates on bystander effects and drug development with regard to these enzyme prodrug combinations, namely herpes simplex virus thymidine kinase (HSV-TK) with ganciclovir (GCV), cytosine deaminase (CD) from bacteria or yeast with 5-fluorocytodine (5-FC), and bacterial nitroreductase (NfsB) with 5-(azaridin-1-yl)-2,4-dinitrobenzamide (CB1954), and their respective derivatives.


Assuntos
Terapia Genética/métodos , Pró-Fármacos/uso terapêutico , Animais , Aziridinas/uso terapêutico , Citosina Desaminase/genética , Citosina Desaminase/fisiologia , Flucitosina/uso terapêutico , Ganciclovir/uso terapêutico , Humanos , Nitrorredutases/genética , Nitrorredutases/fisiologia , Timidina Quinase/genética
12.
Arch Virol ; 154(10): 1579-88, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19669862

RESUMO

Apolipoprotein B mRNA-editing enzyme-catalytic polypeptide-like (APOBEC) proteins are members of a protein family sharing the common characteristic of cytidine deaminase activity. The antiviral activity of APOBEC3G and APOBEC3F has been studied more extensively than that of the other members of this family. The antiviral activity of APOBEC3B and APOBEC3DE has also been described. Studies of other APOBEC proteins have not revealed any antiviral activities against HIV-1; however, further investigation is required. In the absence of human immunodeficiency virus type 1 (HIV-1) virion infectivity factor (Vif), APOBEC3G and APOBEC3F are incorporated into HIV-1 virions and hypermutate the viral genomic DNA by their cytidine deaminase activity. HIV-1 Vif protein suppresses the antiviral role of APOBEC proteins by several mechanisms that lead to inhibition of incorporation of APOBEC3G/3F into HIV-1 virions. The detailed mechanisms involved in the suppression of APOBEC proteins by Vif are still being elucidated. Novel studies in which as yet undefined aspects of the suppression of APOBEC proteins are investigated could reveal important and potentially exploitable information for addressing HIV-1 infection in humans.


Assuntos
Citidina Desaminase/fisiologia , Citosina Desaminase/fisiologia , HIV-1/fisiologia , Produtos do Gene vif do Vírus da Imunodeficiência Humana/fisiologia , Desaminase APOBEC-3G , Citidina Desaminase/antagonistas & inibidores , Citosina Desaminase/antagonistas & inibidores , DNA Viral/metabolismo , Regulação Viral da Expressão Gênica/fisiologia , Humanos , Ubiquitinação , Vírion/metabolismo
13.
Chin Med J (Engl) ; 122(11): 1267-72, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19567135

RESUMO

BACKGROUND: Cytosine deaminase (CD) converts 5-fluorocytosine (5-FC) to 5-fluorouracil (5-FU) in CD/5-FC gene therapy, 5-FU will be mostly converted into nontoxic beta-alanine without uracil phosphoribosyltransferase (UPRT). UPRT catalyzes the conversion of 5-FU to 5-fluorouridine monophosphate, which directly kills CD::UPRT-expressing cells and surrounding cells via the bystander effect. But the pharmacokinetics and the bystander effect of CD::UPRT/5-FC has not been verified in vivo and in vitro. Before the CD::UPRT/5-FC bi-gene therapy system is used in clinical trial, it is essential to monitor the transgene expression and function in vivo. Thus, we developed a preclinical tumor model to investigate the feasibility of using (19)F-magnetic resonance spectroscopy ((19)F-MRS) and optical imaging to measure non-invasive CD and UPRT expression and its bystander effect. METHODS: C6 and C6-CD::UPRT cells were cultured with 5-FC. The medium, cells and their mixture were analyzed by (19)F-MRS. Rats with intracranial xenografted encephalic C6-CD::UPRT glioma were injected intraperitoneally with 5-FC and their (19)F-MRS spectra recorded. Then the pharmacokinetics of 5-FC was proved. Mixtures of C6 and C6-CD::UPRT cells at different ratios were cultured with 5-FC and the cytotoxic efficacy and survival rate of cells recorded. To determine the mechanism of the bystander effect, the culture media from cell comprising 25% and 75% C6-CD::UPRT cells were examined by (19)F-MRS. A comparative study of mean was performed using analysis of variance (ANOVA). RESULTS: (19)F-MRS on samples from C6-CD::UPRT cells cultured with 5-FC showed three broad resonance signals corresponding to 5-FC, 5-FU and fluorinated nucleotides (F-Nuctd). For the C6 mixture, only the 5-FC peak was detected. In vivo serial (19)F-MRS spectra showed a strong 5-FC peak and a weak 5-FU peak at 20 minutes after 5-FC injection. The 5-FU concentration reached a maximum at about 50 minutes. The F-Nuctd signal appeared after about 1 hour, reached a maximum at around 160 minutes, and was detectable for several hours. At a 10% ratio of C6-CD::UPRT cells, the survival rate was (79.55 +/- 0.88)% (P < 0.01). As the C6-CD::UPRT ratio increased, the survival rate of the cells decreased. (19)F-MRS showed that the signals for 5-FU and F-Nuctd in the culture medium increased as the ratio of C6-CD::UPRT in the mixture increased. CONCLUSIONS: (19)F-MRS studies indicated that C6-CD::UPRT cells could effectively express CD and UPRT enzymes. The CD::UPRT/5-FC system showed an obvious bystander effect. This study demonstrated that CD::UPRT/5-FC gene therapy is suitable for 5-FC to F-Nuctd metabolism; and (19)F-MRS can monitor transferred CD::UPRT gene expression and catalysis of substrates noninvasively, dynamically and quantitatively.


Assuntos
Citosina Desaminase/fisiologia , Flucitosina/farmacocinética , Flucitosina/uso terapêutico , Terapia Genética/métodos , Glioma/terapia , Pentosiltransferases/fisiologia , Animais , Antimetabólitos/farmacocinética , Antimetabólitos/uso terapêutico , Linhagem Celular , Citosina Desaminase/genética , Glioma/tratamento farmacológico , Humanos , Imageamento por Ressonância Magnética , Masculino , Pentosiltransferases/genética , Ratos , Ratos Sprague-Dawley
14.
Cancer Res ; 69(11): 4791-9, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19487291

RESUMO

Cytosine deaminase is used in combination with 5-fluorocytosine as an enzyme-prodrug combination for targeted genetic cancer treatment. This approach is limited by inefficient gene delivery and poor prodrug conversion activities. Previously, we reported individual point mutations within the substrate binding pocket of bacterial cytosine deaminase (bCD) that result in marginal improvements in the ability to sensitize cells to 5-fluorocytosine (5FC). Here, we describe an expanded random mutagenesis and selection experiment that yielded enzyme variants, which provide significant improvement in prodrug sensitization. Three of these mutants were evaluated using enzyme kinetic analyses and then assayed in three cancer cell lines for 5FC sensitization, bystander effects, and formation of 5-fluorouracil metabolites. All variants displayed 18- to 19-fold shifts in substrate preference toward 5FC, a significant reduction in IC(50) values and improved bystander effect compared with wild-type bCD. In a xenograft tumor model, the best enzyme mutant was shown to prevent tumor growth at much lower doses of 5FC than is observed when tumor cells express wild-type bCD. Crystallographic analyses of this construct show the basis for improved activity toward 5FC, and also how two different mutagenesis strategies yield closely related but mutually exclusive mutations that each result in a significant alteration of enzyme specificity.


Assuntos
Apoptose , Citosina Desaminase/uso terapêutico , Proteínas de Escherichia coli/uso terapêutico , Flucitosina/uso terapêutico , Neoplasias/terapia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Terapia Combinada , Citosina Desaminase/genética , Citosina Desaminase/fisiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Feminino , Flucitosina/farmacologia , Terapia Genética/métodos , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Moleculares , Proteínas Mutantes/fisiologia , Proteínas Mutantes/uso terapêutico , Neoplasias/genética , Neoplasias/patologia , Pró-Fármacos/uso terapêutico , Engenharia de Proteínas , Ratos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Biomaterials ; 30(23-24): 4014-20, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19427690

RESUMO

Combined treatment using nonviral agent-mediated enzyme/prodrug therapy and immunotherapy had been proposed as a powerful alternative method of cancer therapy. The present study was aimed to evaluate the cytotoxicity in vitro and the therapeutic efficacy in vivo when the cytosine deaminase/5-fluorocytosine (CD/5-FC) and TNF-related apoptosis-inducing ligand (TRAIL) genes were jointly used against rat C6 glioma cells. The potency of the FA-PEG-PEI used as a nonviral vector was tested in the FR-expressed C6 glioma cells and Wistar rats. The C6 glioma cells and animal model were treated by the combined application of FA-PEG-PEI/pCD/5-FC and FA-PEG-PEI/pTRAIL. The antitumor effect was evaluated by survival assays and tumor volume. This study revealed a significant increase of cytotoxicity in vitro following the combined application of FA-PEG-PEI/pCD/5-FC and FA-PEG-PEI/pTRAIL treatments in C6 glioma cells. Animal studies showed a significant growth inhibition of the C6 glioma xenografts using the combined treatment. These results demonstrated that the combined treatment generated additive cytotoxic effect in C6 glioma cells in both in vitro and in vivo conditions, and indicated that such treatment method using both enzyme/prodrug therapy and TRAIL immunotherapy might be a promising therapeutic strategy in treating glioma.


Assuntos
Ácido Fólico/química , Terapia Genética/métodos , Vetores Genéticos/química , Glioma/terapia , Polietilenoglicóis/química , Polietilenoimina/química , Animais , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Citosina Desaminase/genética , Citosina Desaminase/fisiologia , Imuno-Histoquímica , Masculino , Ratos , Ratos Wistar , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia
16.
Mol Ther ; 17(8): 1373-80, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19471249

RESUMO

A substantial fraction of sporadic and inherited colorectal and endometrial cancers in humans is deficient in DNA mismatch repair (MMR). These cancers are characterized by length alterations in ubiquitous simple sequence repeats, a phenotype called microsatellite instability. Here we have exploited this phenotype by developing a novel approach for the highly selective gene therapy of MMR-deficient tumors. To achieve this selectivity, we mutated the VP22FCU1 suicide gene by inserting an out-of-frame microsatellite within its coding region. We show that in a significant fraction of microsatellite-instable (MSI) cells carrying the mutated suicide gene, full-length protein becomes expressed within a few cell doublings, presumably resulting from a reverting frameshift within the inserted microsatellite. Treatment of these cells with the innocuous prodrug 5-fluorocytosine (5-FC) induces strong cytotoxicity and we demonstrate that this owes to multiple bystander effects conferred by the suicide gene/prodrug combination. In a mouse model, MMR-deficient tumors that contained the out-of-frame VP22FCU1 gene displayed strong remission after treatment with 5-FC, without any obvious adverse systemic effects to the mouse. By virtue of its high selectivity and potency, this conditional enzyme/prodrug combination may hold promise for the treatment or prevention of MMR-deficient cancer in humans.


Assuntos
Antimetabólitos/farmacologia , Flucitosina/farmacologia , Genes Transgênicos Suicidas/fisiologia , Instabilidade de Microssatélites/efeitos dos fármacos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Citosina Desaminase/genética , Citosina Desaminase/fisiologia , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Mutação da Fase de Leitura/genética , Genes Transgênicos Suicidas/genética , Humanos , Camundongos , Neoplasias/genética , Neoplasias/terapia , Pentosiltransferases/genética , Pentosiltransferases/fisiologia , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/fisiologia
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 40(6): 983-7, 2009 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-20067103

RESUMO

OBJECTIVE: To construct the targeting double suicide gene therapy vector pcDNA3.1 (-) Cp-CD-TK driven by carcino-embryonic antigen promoter (CEA promoter, Cp), and to investigate whether the vector could control the expression of CD-TK gene specificity and impact on proliferation of CEA positive colon cancer cells. METHODS: Three kinds of target gene, Cp, CD, and TK were obtained by PCR, the products were double digested and inserted into pcDNA3.1 (-), then the targeting vector pcDNA3.1 (-) Cp-CD-TK was transfected into CEA positive human colon cancer SW480 cells and CEA negative Hela cells, the expression of CD-TK was examined by RT-PCR. The sensitivities of SW480 cells transfected with pcDNA3.1 (-) Cp-CD-TK to pro-drug 5-Fluorocytosine (5-Fc) and Ganciclovir (GCV) were detected by MTT analysis. RESULTS: Recombinants with Cp, CD and TK insert were obtained. Constructed targeting gene therapy vector pcDNA3.1 (-) Cp-CD-TK was confirmed by gel electrophoresis and sequencing. RT-PCR analysis further confirmed CD-TK gene was expressed in SW480 cells and was not expressed in CEA negative Hela cells. MTT assay demonstrated that SW480 cells transfected with targeting vector pcDNA3.1 (-) Cp-CD-TK were sensitive to pro-drug 5-Fc and GCV. CONCLUSION: Targeting double suicide gene therapy vector pcDNA3.1 (-) Cp-CD-TK was constructed correctly. The vector could make CD-TK gene express specifically in CEA positive cells for the purpose of targeting killing colon cancer.


Assuntos
Antígeno Carcinoembrionário/genética , Neoplasias Colorretais/genética , Genes Transgênicos Suicidas/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/terapia , Citosina Desaminase/genética , Citosina Desaminase/fisiologia , Humanos , Regiões Promotoras Genéticas/genética , Timidina Quinase/genética , Timidina Quinase/fisiologia
18.
Proc Natl Acad Sci U S A ; 105(14): 5501-6, 2008 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-18391217

RESUMO

The HIV-1 Vif protein is essential for overcoming the antiviral activity of DNA-editing apolipoprotein B mRNA editing enzyme, catalytic polypeptide 3 (APOBEC3) cytidine deaminases. We show that naturally occurring HIV-1 Vif point mutants with suboptimal anti-APOBEC3G activity induce the appearance of proviruses with lamivudine (3TC) drug resistance-associated mutations before any drug exposure. These mutations, ensuing from cytidine deamination events, were detected in >40% of proviruses with partially defective Vif mutants. Transfer of drug resistance from hypermutated proviruses via recombination allowed for 3TC escape under culture conditions prohibitive for any WT viral growth. These results demonstrate that defective hypermutated genomes can shape the phenotype of the circulating viral population. Partially active Vif alleles resulting in incomplete neutralization of cytoplasmic APOBEC3 molecules are directly responsible for the generation of a highly diverse, yet G-to-A biased, proviral reservoir, which can be exploited by HIV-1 to generate viable and drug-resistant progenies.


Assuntos
Citidina/metabolismo , Citosina Desaminase/fisiologia , Farmacorresistência Viral , Produtos do Gene vif/genética , HIV-1 , Desaminases APOBEC , Citidina Desaminase , Desaminação , Produtos do Gene vif/fisiologia , Humanos , Lamivudina , Mutação , Provírus
19.
Virus Res ; 134(1-2): 74-85, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18262674

RESUMO

The ability of members of the APOBEC3 (A3) family of proteins to confer intrinsic immunity to retroviral infection was recognized in several studies. More specifically, A3 proteins are cytidine deaminases (CDAs) that cause hypermutations of nascent retroviral genomes by deamination of cytidine residues. Although A3 proteins can restrict the replication of HIV, this inhibition is overcome by the viral infectivity factor (Vif). Inhibitory effects of APOBEC proteins are not limited to HIV but extend to other viruses and endogenous mobile genetic elements that share a reverse transcription process analogous to that of exogenous retroviruses. In sharp contrast, another conundrum of A3 proteins is that they inhibit viral replication even in the absence of CDA activity and recent advances have defined the inhibition of reverse transcriptase (RT) catalyzed DNA elongation reactions by A3 proteins. Together, these proteins provide strong and immediate intracellular immunity against incoming pathogens and restrict the movement of mobile genetic elements protecting the genome.


Assuntos
Citosina Desaminase/fisiologia , Infecções por HIV/imunologia , HIV/genética , Transcrição Reversa , Desaminases APOBEC , Sequência de Aminoácidos , Citidina Desaminase , Citosina Desaminase/química , Citosina Desaminase/genética , Citosina Desaminase/imunologia , HIV/fisiologia , Infecções por HIV/virologia , Humanos , Família Multigênica , Estrutura Terciária de Proteína , Retroelementos , Replicação Viral
20.
J Neurooncol ; 86(1): 3-11, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17594053

RESUMO

The multiple drug resistance protein (MDR1) is frequently overexpressed in human glioma. The aim of this study is to clone the MDR1 promoter from C6/ADR, construct the double suicide genes expressive vector controlled by MDR1 promoter, and explore its targeted expression in C6/ADR cells. MDR1 promoter from C6/ADR genomic DNA, which was linked with T vector, was amplified by using Polymerase chain reaction (PCR). After cut by NdeI and HindIII, MDR1 promoter was cloned into pcDNA3-TK (thymidine kinase) plasmid. The cytosine deaminase (CD) gene from pcDNA3-CD-TK plasmid was directly cloned into the above vector to construct pcDNA3-MDR1-promoter-CD-TK vector. Then this vector was transfected into C6 and C6/ADR cells respectively by liposome. After selection by G418, the tumor cell lines were stably established. Then these cell lines were examined through PCR and RT-PCR to respectively detect the integration and expression of TK and CD genes. The results showed the length and sequence of MDR1 promoter amplified by PCR were confirmed by DNA sequencing. The pcDNA3-MDR1-promoter-CD-TK expression vectors were constructed successfully. PCR indicated the double suicide genes were integrated into C6 and C6/ADR cells. RT-PCR revealed that CD and TK genes expressed in C6/ADR/CD-TK cells, whereas not in C6/CD-TK cells. In conclusions, construction of expressive vector containing double suicide genes controlled by MDR1 promoter with targeted expression in C6/ADR will provide a sound basis for targeted gene therapy for multidrug resistance (MDR) glioma.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Transgênicos Suicidas/genética , Glioma/terapia , Regiões Promotoras Genéticas/fisiologia , Animais , Linhagem Celular Tumoral , Citosina Desaminase/genética , Citosina Desaminase/fisiologia , Citometria de Fluxo , Terapia Genética , Vetores Genéticos/fisiologia , Ratos , Timidina Quinase/genética , Timidina Quinase/fisiologia , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA