Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Nucleic Acids ; 13: 514-533, 2018 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-30388625

RESUMO

Conventional HIV gene therapy approaches are based on engineering HIV target cells that are non-permissive to viral replication. However, expansion of gene-modified HIV target cells has been limited in patients. Alternative genetic strategies focus on generating gene-modified producer cells that secrete antiviral proteins (AVPs). The secreted AVPs interfere with HIV entry, and, therefore, they extend the protection against infection to unmodified HIV target cells. Since any cell type can potentially secrete AVPs, hematopoietic and non-hematopoietic cell lineages can function as producer cells. Secretion of AVPs from non-hematopoietic cells opens the possibility of using a genetic approach for HIV prevention. Another strategy aims at modifying cytotoxic T cells to selectively target and eliminate infected cells. This review provides an overview of the different genetic approaches for HIV treatment and prevention.

2.
Mol Ther Nucleic Acids ; 13: 347-364, 2018 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-30340139

RESUMO

HIV entry is a highly specific and time-sensitive process that can be divided into receptor binding, coreceptor binding, and membrane fusion. Bifunctional antiviral proteins (bAVPs) exploit the multi-step nature of the HIV entry process by binding to two different extracellular targets. They are generated by expressing a fusion protein containing two entry inhibitors with a flexible linker. The resulting fusion proteins exhibit exceptional neutralization potency and broad cross-clade inhibition. In this review, we summarize the HIV entry process and provide an overview of the design, antiviral potency, and methods of delivery of bAVPs. Additionally, we discuss the advantages and limitations of bAVPs for HIV prevention and treatment.

3.
Mol Ther Nucleic Acids ; 9: 132-144, 2017 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-29246292

RESUMO

HIV entry inhibitors are highly effective in controlling virus replication. We have developed a lentiviral vector that expresses a secreted entry inhibitor, soluble CD4 (sCD4), which binds to the HIV envelope glycoproteins and inactivates the virus. We have shown that sCD4 was secreted from gene-modified CD4+ T cells, as well as from human umbilical cord blood-derived CD34+ hematopoietic stem/progenitor cells (HSPCs), and protected unmodified HIV target cells from infection in vitro. To investigate the in vivo application of our approach, we injected gene-modified HSPCs into NOD/SCID/γcnull (NSG) mice. NSG hosts supported multi-lineage differentiation of human gene-modified HSPCs. Upon challenge with HIV, humanized mice capable of secreting sCD4 demonstrated a reduction of viral load over time compared to control humanized mice. In contrast to gene therapy approaches that render only gene-modified HIV target cells resistant to infection, our approach also showed protection of unmodified CD4+ T cells in the peripheral blood and tissues. Our findings provide support for the continuous delivery of secreted entry inhibitors via gene therapy as an alternative to oral administration of antiretroviral drugs or injection of antiretroviral proteins, including antibodies.

4.
Mol Ther Nucleic Acids ; 7: 387-395, 2017 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-28624214

RESUMO

HIV entry into target cells is a highly sequential and time-sensitive process. In recent years, potent HIV Env-targeting antibodies, such as VRC01, have been identified. However, antibodies bind only to a single epitope, and mutations that confer resistance to antibody-mediated inhibition of HIV entry have been detected. In contrast, HIV cannot escape from binding to soluble CD4 (sCD4) without a fitness disadvantage. sCD4 has the unique ability to induce conformational changes within the HIV envelope glycoproteins (Env) that allow fusion inhibitors to bind. We have previously linked sCD4 to the fusion inhibitor FIT45 (sCD4-FIT45) and examined delivery of the bifunctional entry inhibitor via gene therapy. Here, we extend our studies and analyze the ability of sCD4-FIT45 to inhibit HIV Env-mediated cell fusion and HIV entry of several primary isolates. sCD4-FIT45 inhibited both cell fusion and HIV entry with remarkable antiviral activity. The mean 50% inhibitory concentrations (IC50) for sCD4-FIT45 were <0.2 µg/mL in both assays. Importantly, inhibition by sCD4-FIT45 was more potent than by VRC01, sCD4, or the previously described bifunctional protein sCD4-scFv17b. In contrast to sCD4, sCD4-FIT45 as well as VRC01 and sCD4-scFv17b did not mediate cell fusion between HIV Env+ and CD4-CCR5+ cells. The results presented here provide further evidence for the testing of sCD4-FIT45 and development of bifunctional proteins based on the sCD4-fusion inhibitor architecture.

5.
BMC Biotechnol ; 16(1): 66, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27590008

RESUMO

BACKGROUND: Recombinant proteins of therapeutic use are ideally produced in human cells to ensure appropriate co- and post-translational modifications. However, purification of secreted proteins from the culture media is impeded by low expression from transfected cell lines and the presence of serum proteins. Here we describe a simple and cost-effective approach based on lentiviral vector-mediated gene delivery and expression of a secreted His-tagged protein from human embryonic kidney 293 T cells and direct affinity chromatography purification from the cell culture media. RESULTS: Using a protein-based HIV entry inhibitor, soluble CD4 (sCD4), we demonstrated that 293 T cells transduced with a lentiviral vector mediated over 10-fold higher secretion of sCD4 in comparison to 293 T cells transfected with the corresponding plasmid. Secretion of sCD4 increased with the dose of the lentiviral vector up to a multiplicity of infection of 50. Exchanging the native signal peptide of sCD4 with the signal peptide of human alpha-1 antitrypsin increased expression by 50 %. There was no difference in expression from a monocistronic or bicistronic lentiviral vector. Reduction of the serum concentration in the culture media had no significant effect on the secretion of sCD4. Small-scale purification from 50 ml of culture media with reduced serum content yielded up to 1 mg of pure sCD4. Purified sCD4 bound to recombinant HIV envelope glycoprotein 120 (Env gp120) and inhibited HIV entry at concentrations comparable to published results. CONCLUSION: The procedure outlined in this study can be performed without the need for specialized reagents or equipment and could easily be adapted by any laboratory. Furthermore, the method could be used to produce sCD4 fusion proteins or other His-tagged proteins.


Assuntos
Vetores Genéticos/genética , Histidina/genética , Lentivirus/genética , Engenharia de Proteínas , Proteínas Recombinantes/metabolismo , Cromatografia de Afinidade , Células HEK293 , Humanos , Proteínas Recombinantes/genética
6.
Curr Gene Ther ; 9(1): 20-5, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19275568

RESUMO

AIDS is the result of infection by a lentivirus, HIV-1, which primarily infects CD4+ T cells and macrophages. There is presently no vaccine and none will be available in the foreseeable future. Highly active antiretroviral drug therapy has led to a dramatic reduction of viral load in many infected individuals, and has decreased mortality in the developing world. However, besides long-term drug toxicity and eventual emergence of drug-resistant strains, withdrawal from the therapy (even after effective and continuous treatment) results in re-emergence of the virus since cells harbouring the latent viral reservoirs persist. These issues highlight the need for alternative therapies, e.g. gene therapy. This review summarizes various gene therapy strategies that target early stages of HIV-1 life cycle. We will cover strategies that allow interference at the level of the released virion RNA, reverse transcriptase, pre-integration complex, integrase, dsDNA and provirus DNA in gene-modified cells.


Assuntos
DNA Viral/genética , Terapia Genética , Infecções por HIV/terapia , HIV-1/genética , Provírus/genética , Integração Viral/fisiologia , Infecções por HIV/genética , Infecções por HIV/virologia , Humanos
7.
J Gen Virol ; 89(Pt 10): 2605-2610, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18796730

RESUMO

This study examined whether insertion of a mobile group II intron into infectious human immunodeficiency virus type 1 (HIV-1) provirus DNA could inhibit virus replication. Introns targeted against two sites within the integrase-coding region were used. The intron-inserted HIV-1 provirus DNA clones were isolated and tested for virus replication. Similar amounts of HIV-1 RNA, Gag protein and progeny virus were produced from HIV-1 provirus DNA and intron-inserted HIV-1 provirus DNA. However, when the progeny virus was tested for its infectivity, although the group II intron-inserted HIV-1 RNA was packaged and reverse-transcribed, the dsDNA failed to integrate, as expected in the absence of a functional integrase, and virus replication was aborted. These results demonstrate that group II introns can confer 'complete' inhibition of HIV-1 replication at the intended step and should be further exploited for HIV-1 gene therapy and other targeted genetic repairs.


Assuntos
HIV-1/genética , HIV-1/fisiologia , Íntrons/genética , Inativação de Vírus , Replicação Viral , Linhagem Celular , Replicação do DNA , DNA Viral/genética , DNA Viral/metabolismo , Terapia Genética/métodos , HIV-1/patogenicidade , Humanos , Provírus/genética , Transfecção
8.
J Gen Virol ; 89(Pt 9): 2252-2261, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18753234

RESUMO

Rz1-7 is a multimeric hammerhead ribozyme targeting seven unique sites within the human CCR5 mRNA that is active in vitro. Mouse stem cell virus-based MGIN and human immunodeficiency virus (HIV)-1-based HEG1 vectors were used to express Rz1-7 in a human CD4+ T lymphoid cell line. Stable transductants expressed Rz1-7, which was further shown to be active, since CCR5 mRNA and surface CCR5 protein expression levels decreased. High levels of progeny virus were produced when the transduced cells were challenged with an X4-tropic HIV-1 (NL4-3) strain, suggesting that Rz1-7 expression does not affect X4-tropic virus replication. When the transduced cells expressing Rz1-7 were challenged with the R5-tropic HIV-1 (BaL) strain, 99-100% inhibition of progeny virus production was observed for the duration of the experiment (approximately 2 months). When the cells were precultured for 2-3 months prior to HIV-1 infection, inhibition was more prominent in cells transduced with MGIN-Rz1-7 than with HEG1-Rz1-7. Inhibition occurred at the level of viral entry, as no HIV-1 DNA could be detected. These results demonstrate that Rz1-7 confers excellent inhibition of R5-tropic HIV-1 replication at the level of entry. Therefore, we anticipate that this multimeric ribozyme will be beneficial for HIV-1 gene therapy.


Assuntos
HIV-1/genética , HIV-1/fisiologia , RNA Catalítico/genética , RNA Catalítico/metabolismo , Receptores CCR5/genética , Animais , Sequência de Bases , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , DNA Viral/genética , DNA Viral/metabolismo , Expressão Gênica , Genes Virais , Vetores Genéticos , Humanos , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Viral/genética , Transdução Genética , Internalização do Vírus
9.
Curr Gene Ther ; 8(4): 264-72, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18691022

RESUMO

Acquired immune deficiency syndrome (AIDS) is caused by a lentivirus, human immunodeficiency virus type-1 (HIV-1). Viral entry is mediated by specific interaction of the viral envelope (Env) glycoprotein with a cell surface molecule CD4 which serves as the primary receptor and a chemokine (C-C or C-X-C motif) receptor CCR5 or CXCR4 which serves as a co-receptor. The viral Env, the cellular CD4 receptor, or the CCR5/CXCR4 co-receptors may be the targets of therapeutic interventions. Compared to the high variability of the viral Env protein, lack of variability in the CD4 receptor and the CCR5 or CXCR4 co-receptor makes them better targets to prevent viral entry. Downregulation of CD4 or CXCR4 is likely to have harmful consequences for the immune function or cellular maturation and homing. In contrast, individuals who lack functional CCR5 have no apparent immune defects, and show decreased susceptibility to HIV-1 infection and delayed progression to AIDS. CCR5 is essential for HIV-1 infection through all routes of transmission. Therefore, its downregulation may not only prevent disease progression, but also the spread of HIV-1 transmission. To block CCR5 function, a number of molecules were developed, including low molecular weight compounds, chemokines, N-terminally-modified chemokine analogues, chemokine-derived molecules, chemokine-based synthetic peptides, and anti-CCR5 monoclonal antibodies. Gene therapy strategies were developed using intrakines and intrabodies to prevent cell surface expression of CCR5 and zinc finger-nucleases, or using small interfering RNAs, antisense RNAs, or ribozymes to decrease co-receptor synthesis. This review describes the importance of targeting CCR5 and summarizes the status of various anti-CCR5 gene therapy strategies.


Assuntos
Antagonistas dos Receptores CCR5 , Terapia Genética , Infecções por HIV/terapia , HIV-1/fisiologia , Animais , Infecções por HIV/genética , Humanos
10.
Biopolymers ; 83(2): 122-34, 2006 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-16683262

RESUMO

Artificial proteins potentially barrier-free in the folding kinetics are approached computationally under the guidance of protein-folding theories. The smallest and fastest folding globular protein triple-helix-bundle (THB) is so modified as to minimize or eliminate its presumed barriers in folding speed. As the barriers may reside in the ordering of either secondary or tertiary structure, the elements of both secondary and tertiary structure in the protein are targeted for prenucleation with suitable stereochemically constrained amino acid residues. The required elements of topology and sequence for the THB are optimized independently; first the topology is optimized with simulated annealing in polypeptides of highly simplified alphabet; next, the sequence in side chains is optimized using the standard inverse design methods. The resultant three best-adapted THBs, variable in topology and distinctive in sequences, are assessed by comparing them with a few benchmark proteins. The results of mainly molecular dynamics (MD) comparisons, undertaken in explicit water at different temperatures, show that the designed sequences are favorably placed against the chosen benchmarks as THB proteins potentially thermostable in the native folds. Folding simulation experiments with MD establish that the designed sequences are rapid in the folding of individual helices, but not in the evolution of tertiary structure; energetic cum topological frustrations remain but could be the artifacts of the starting conformations that were chosen in the THBs in the folding simulations. Overall, a practical high-throughput approach for de novo protein design has been developed that may have fruitful application for any type of tertiary structure.


Assuntos
Simulação por Computador , Dobramento de Proteína , Proteínas/química , Sequência de Aminoácidos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Estereoisomerismo , Termodinâmica
11.
Front Biosci ; 11: 2940-8, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16720366

RESUMO

A combination gene therapy strategy using an ASPsi-gag antisense RNA (targeted against the packaging signal and the gag-coding region) and a multimeric hammerhead ribozyme Rz1-9 (targeted against nine sites within the env-coding region) or Rz1-14 (targeted against 14 sites within the 5' leader and the pro-, pol-, vif- and env-coding regions) was assessed for inhibiting HIV-1 replication. A murine stem cell virus (MSCV)-based MGIN vector was used to express Rz1-9, Rz1-14, ASPsi-gag, Rz1-9ASPsi-gag, or Rz1-14ASPsi-gag RNA in a CD4+ T lymphoid cell line. Stable transductants were shown to express similar levels of interfering RNA. HIV-1 replication was inhibited in cells expressing Rz1-9 and Rz1-14. Little inhibition of HIV-1 replication was observed in cells expressing ASPsi-gag RNA. Thus, the multimeric hammerhead ribozymes inhibit HIV-1 replication better than the antisense RNA. Inhibition of HIV-1 replication in cells expressing Rz1-9ASPsi-gag or Rz1-14ASPsi-gag RNA was worse than that obtained with the multimeric ribozymes alone. This result suggests that co-expression of antisense RNA decreases the anti-HIV potential of ribozymes. The multimeric ribozymes and the antisense RNA were designed to target different sites within the HIV-1 RNA. They are not expected to interact with each other. Neither are they expected to compete with each other for binding to the HIV-1 RNA. Instead, the antisense RNA binding to its (1553 nt-long) target site may have resulted in a decreased ribozyme turn over. Furthermore, since the antisense RNA/HIV-1 RNA hybrids are degraded by the cells, the co-expressed antisense RNA may have led to ribozyme degradation.


Assuntos
Terapia Genética , HIV-1/genética , HIV-1/fisiologia , RNA Antissenso/metabolismo , RNA Catalítico/genética , RNA Catalítico/metabolismo , Linfócitos T CD4-Positivos/virologia , Vetores Genéticos , Infecções por HIV/terapia , Vírus da Leucemia Murina de Moloney/genética , RNA Antissenso/genética , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Replicação Viral
12.
Front Biosci ; 7: a15-28, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11815282

RESUMO

Oncoretroviral vectors were engineered to allow constitutive expression of an antisense RNA and the trans-activator of transcription (Tat)-inducible expression of a mRNA containing the trans-activation response (TAR) element, the Rev response element (RRE), and the efficient packaging signal (Psi(e) of human immunodeficiency virus-1 (HIV-1) RNA. Nuclear export of this mRNA by the regulator of expression of virion proteins (Rev) would allow its translation into wild type (WT) (MoTN-Ti-GE-Ri- Ter) or trans-dominant negative mutant (TDM) (MoTN-Ti-GmEm-Ri-Ter) Gag and Env proteins. Thus, the antisense RNA produced in a constitutive manner would ensure that even if there is leaky expression, no WT/TDM Gag or Env protein would be produced in the uninfected cells. If cells become infected by HIV-1, the antisense RNA would inhibit HIV-1 replication. Failure on the part of antisense RNA to inhibit virus replication would allow GE/GmEm mRNA production. The GE/GmEm mRNA would cause partial inhibition of HIV-1 replication as it contains the TAR, RRE, and Psi(e) signal sequences. Translation of GmEm mRNA would give rise to TDM Gag and Env proteins, which would further decrease progeny virus infectivity. Tat- and Rev-inducibility was demonstrated in transfected HeLa and HeLa-Tev cells. Full-length WT/TDM Gag production was confirmed by Western blot analysis. Amphotropic vector particles were used to transduce a human CD4+ T-lymphoid cell line, and the stable transductants were challenged with HIV-1. Virus replication was better inhibited by the MoTN-Ti-GE-Ri-Ter vector than by the MoTN-Ti-GmEm-Ri-Ter vector. Inhibition of HIV-1 replication was also demonstrated in transduced CD4+ human peripheral blood T lymphocytes (PBLs). Moreover, our results suggest that cloning in the reverse transcriptional orientation must be avoided to prevent antisense RNA-mediated inhibition of transgene and endogenous gene expression.


Assuntos
Produtos do Gene env/genética , Produtos do Gene gag/genética , Terapia Genética/métodos , HIV-1 , RNA Antissenso/genética , Linhagem Celular , Produtos do Gene env/metabolismo , Produtos do Gene gag/metabolismo , Produtos do Gene rev/farmacologia , Produtos do Gene tat/farmacologia , Vetores Genéticos , Infecções por HIV/terapia , HIV-1/genética , Células HeLa , Humanos , Vírus da Leucemia Murina de Moloney/genética , Mutação , Fases de Leitura Aberta , RNA Antissenso/metabolismo , RNA Viral/análise , Linfócitos T/virologia , Transcrição Gênica , Transdução Genética , Replicação Viral , Produtos do Gene rev do Vírus da Imunodeficiência Humana , Produtos do Gene tat do Vírus da Imunodeficiência Humana
13.
Front Biosci ; 7: a29-36, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11815297

RESUMO

Rz1-9 is a multimeric hammerhead ribozyme targeted against nine highly conserved sites within the env coding region of human immunodeficiency virus type-1 (HIV-1) RNA. This ribozyme was shown to inhibit HIV-1 replication (1, 2). However, Rz1-9 target sites are only conserved within the clade B of HIV-1. In this study, we have designed another multimeric ribozyme, Rz10-14. This multimeric ribozyme is targeted against five sites that are highly conserved amongst all major clades of HIV-1. A third multimeric ribozyme, Rz1-14, was obtained by combining both Rz1-9 and Rz10-14. A mouse stem cell virus-based MGIN vector (3) was used to express these ribozymes in a human CD4+ T lymphoid cell line. Stable transductants expressed vector RNA containing ribozymes which were shown to be active. In HIV-1 challenge experiments, very little or no virus production could be detected in the pools of stable MT4 transductants expressing Rz1-14 for 60 days tested. Inhibition of virus replication was most prominent with Rz1-14, followed by Rz1-9, and then Rz10-14. Thus, the combined multimeric ribozyme, Rz1-14, is more effective than Rz1-9 or Rz10-14. As Rz1-14 is targeted against all major clades of HIV-1, it will be further pursued for use in HIV-1 gene therapy.


Assuntos
Terapia Genética/métodos , HIV-1 , RNA Catalítico/genética , Retroviridae/genética , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Marcação de Genes , Vetores Genéticos , Infecções por HIV/terapia , HIV-1/genética , HIV-1/crescimento & desenvolvimento , Humanos , RNA Catalítico/metabolismo , Especificidade da Espécie , Transdução Genética , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...