Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 20(7): 1410-6, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22565846

RESUMO

Liver gene transfer for hemophilia B has shown very promising results in recent clinical studies. A potential complication of gene-based treatments for hemophilia and other inherited disorders, however, is the development of neutralizing antibodies (NAb) against the therapeutic transgene. The risk of developing NAb to the coagulation factor IX (F.IX) transgene product following adeno-associated virus (AAV)-mediated hepatic gene transfer for hemophilia is small but not absent, as formation of inhibitory antibodies to F.IX is observed in experimental animals following liver gene transfer. Thus, strategies to modulate antitransgene NAb responses are needed. Here, we used the anti-B cell monoclonal antibody rituximab (rtx) in combination with cyclosporine A (CsA) to eradicate anti-human F.IX NAb in rhesus macaques previously injected intravenously with AAV8 vectors expressing human F.IX. A short course of immunosuppression (IS) resulted in eradication of anti-F.IX NAb with restoration of plasma F.IX transgene product detection. In one animal, following IS anti-AAV6 antibodies also dropped below detection, allowing for successful AAV vector readministration and resulting in high levels (60% or normal) of F.IX transgene product in plasma. Though the number of animals is small, this study supports for the safety and efficacy of B cell-targeting therapies to eradicate NAb developed following AAV-mediated gene transfer.


Assuntos
Fator IX/imunologia , Técnicas de Transferência de Genes , Terapia Genética , Hemofilia B/terapia , Imunidade Humoral/efeitos dos fármacos , Animais , Anticorpos Monoclonais Murinos/farmacologia , Anticorpos Neutralizantes/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Ciclosporinas/farmacologia , Dependovirus/genética , Fator IX/genética , Hemofilia B/genética , Fatores Imunológicos/farmacologia , Imunossupressores/farmacologia , Macaca mulatta , Rituximab , Transgenes
2.
Nature ; 475(7355): 217-21, 2011 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-21706032

RESUMO

Editing of the human genome to correct disease-causing mutations is a promising approach for the treatment of genetic disorders. Genome editing improves on simple gene-replacement strategies by effecting in situ correction of a mutant gene, thus restoring normal gene function under the control of endogenous regulatory elements and reducing risks associated with random insertion into the genome. Gene-specific targeting has historically been limited to mouse embryonic stem cells. The development of zinc finger nucleases (ZFNs) has permitted efficient genome editing in transformed and primary cells that were previously thought to be intractable to such genetic manipulation. In vitro, ZFNs have been shown to promote efficient genome editing via homology-directed repair by inducing a site-specific double-strand break (DSB) at a target locus, but it is unclear whether ZFNs can induce DSBs and stimulate genome editing at a clinically meaningful level in vivo. Here we show that ZFNs are able to induce DSBs efficiently when delivered directly to mouse liver and that, when co-delivered with an appropriately designed gene-targeting vector, they can stimulate gene replacement through both homology-directed and homology-independent targeted gene insertion at the ZFN-specified locus. The level of gene targeting achieved was sufficient to correct the prolonged clotting times in a mouse model of haemophilia B, and remained persistent after induced liver regeneration. Thus, ZFN-driven gene correction can be achieved in vivo, raising the possibility of genome editing as a viable strategy for the treatment of genetic disease.


Assuntos
Reparo do DNA/genética , Modelos Animais de Doenças , Marcação de Genes/métodos , Terapia Genética/métodos , Genoma/genética , Hemofilia B/genética , Hemostasia , Animais , Sequência de Bases , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Endonucleases/química , Endonucleases/genética , Endonucleases/metabolismo , Éxons/genética , Fator IX/análise , Fator IX/genética , Vetores Genéticos/genética , Células HEK293 , Hemofilia B/fisiopatologia , Humanos , Íntrons/genética , Fígado/metabolismo , Regeneração Hepática , Camundongos , Camundongos Endogâmicos C57BL , Mutação/genética , Fenótipo , Homologia de Sequência , Dedos de Zinco
3.
Exp Hematol ; 38(12): 1157-66, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20817073

RESUMO

OBJECTIVE: The thymus serves as a critical site of T-lymphocyte ontogeny and selection. Thymic infection by HIV-1 is known to disrupt thymocyte maturation by both direct and indirect means; however, the mechanism behind these effects remains poorly defined. Macrophages represent one of the most important peripheral targets of HIV-1 infection, are resident in the thymic stroma, and play a central role in thymocyte maturation. MATERIALS AND METHODS: Studies presented here define three primary features and outcomes of thymic macrophages (TM) and HIV-1 infection: (1) The distinctive TM phenotype (surface markers and cytokine production measured by immunofluorescence, fluorescence-activated cell sorting, and reverse transcriptase polymerase chain reaction) relative to macrophages from other sources (blood [monocyte-derived macrophages] and bone marrow); (2) infection of TM by different HIV-1 subtypes (X4, R5, and X4/R5) measured by enzyme-linked immunosorbent assay and polymerase chain reaction; and (3) consequences of HIV-1 infection on cytokine production by TM measured by reverse transcriptase polymerase chain reaction. RESULTS: The results demonstrate that TM display a distinctive phenotype of HIV-1 receptors (CD4(lo), CXCR4(lo), CCR5(med), CCR3(hi)), chemokine production (macrophage inflammatory protein-1α(+); regulated on activation, normal T expressed and secreted(+); macrophage inflammatory protein-1b(-); stromal cell-derived factor -1(-)); and cytokine production (tumor necrosis factor-α(+), interleukin-8(+), macrophage colony-stimulating factor(+), interleukin-6(-)) relative to either monocyte-derived macrophages or bone marrow. TM were infected in vitro with R5 and X4/R5-tropic HIV-1 subtypes, and developed syncytia formation during long-term X4/R5 culture. In contrast, TM supported only transient replication of X4-tropic HIV-1. Lastly, infection of TM with HIV-1 abolished the production of all cytokines tested in long-term in vitro cultures. CONCLUSIONS: Taken together, these results indicate that TM are a potential direct target of in situ HIV-1 infection, and that this infection may result in the disruption of macrophage functions that govern normal thymocyte maturation.


Assuntos
Citocinas/biossíntese , HIV-1/patogenicidade , Macrófagos/imunologia , Macrófagos/virologia , Timo/imunologia , Humanos , Lactente , Recém-Nascido
4.
J Clin Invest ; 119(6): 1688-95, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19436115

RESUMO

Adeno-associated virus (AAV) vectors are effective gene delivery vehicles mediating long-lasting transgene expression. Data from a clinical trial of AAV2-mediated hepatic transfer of the Factor IX gene (F9) into hemophilia B subjects suggests that CTL responses against AAV capsid can eliminate transduced hepatocytes and prevent long-term F9 expression. However, the capacity of hepatocytes to present AAV capsid-derived antigens has not been formally demonstrated, nor whether transduction by AAV sensitizes hepatocytes for CTL-mediated destruction. To investigate the fate of capsids after transduction, we engineered a soluble TCR for the detection of capsid-derived peptide:MHC I (pMHC) complexes. TCR multimers exhibited antigen and HLA specificity and possessed high binding affinity for cognate pMHC complexes. With this reagent, capsid pMHC complexes were detectable by confocal microscopy following AAV-mediated transduction of human hepatocytes. Although antigen presentation was modest, it was sufficient to flag transduced cells for CTL-mediated lysis in an in vitro killing assay. Destruction of hepatocytes was inhibited by soluble TCR, demonstrating a possible application for this reagent in blocking undesirable CTL responses. Together, these studies provide a mechanism for the loss of transgene expression and transient elevations in aminotransferases following AAV-mediated hepatic gene transfer in humans and a potential therapeutic intervention to abrogate these limitations imposed by the host T cell response.


Assuntos
Apresentação de Antígeno/imunologia , Proteínas do Capsídeo/imunologia , Citotoxicidade Imunológica/imunologia , Dependovirus/genética , Vetores Genéticos/genética , Hepatócitos/imunologia , Hepatócitos/metabolismo , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Hepatócitos/citologia , Antígenos de Histocompatibilidade/imunologia , Humanos , Multimerização Proteica , Receptores de Antígenos de Linfócitos T/imunologia , Solubilidade , Especificidade por Substrato , Linfócitos T Citotóxicos/imunologia
5.
J Med Virol ; 81(1): 65-74, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19031458

RESUMO

Humoral immune responses occur following exposure to Adeno-associated virus (AAV) or AAV vectors. Many studies characterized antibody responses to AAV, but human IgG subclass responses to AAV have not been previously described. In this study, IgG subclass responses were examined in serum samples of normal human subjects exposed to wild-type AAV, subjects injected intramuscularly with AAV vectors and subjects injected intravascularly with AAV vectors. A diversity of IgG subclass responses to AAV capsid were found in different subjects. IgG1 was found to be the dominant response. IgG2, IgG3, and IgG4 responses were also observed in most normal human subjects; IgG2 and IgG3 each represented the major fraction of total anti-AAV capsid IgG in a subset of normal donors. Subjects exposed to AAV vectors showed IgG responses to AAV capsid of all four IgG subclasses. IgG responses to AAV capsid in clinical trial subjects were inversely proportional to the level of pre-existing anti-AAV antibody and independent of the vector dose. The high levels of anti-AAV capsid IgG1 can mask differences in IgG2, IgG3, and IgG4 responses that were observed in this study. Analysis of IgG subclass distribution of anti-AAV capsid antibodies indicates a complex, non-uniform pattern of responses to this viral antigen. J. Med. Virol. 81:65-74, 2009. (c) 2008 Wiley-Liss, Inc.


Assuntos
Anticorpos Antivirais/sangue , Dependovirus/imunologia , Vetores Genéticos/imunologia , Imunoglobulina G/sangue , Infecções por Parvoviridae/imunologia , Adolescente , Adulto , Idoso , Capsídeo/imunologia , Humanos , Imunoglobulina M/sangue , Pessoa de Meia-Idade , Testes de Neutralização
6.
Mol Ther ; 17(1): 144-52, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18941440

RESUMO

In a gene therapy clinical trial for hemophilia B, adeno-associated virus 2 (AAV2) capsid-specific CD8(+) T cells were previously implicated in the elimination of vector-transduced hepatocytes, resulting in loss of human factor IX (hFIX) transgene expression. To test the hypothesis that expression of AAV2 cap DNA impurities in the AAV2-hFIX vector was the source of epitopes presented on transduced cells, transcription of cap was assessed by quantitative reverse transcription-PCR (Q-RT-PCR) following transduction of target cells with the vector used in the clinical trial. Transcriptional profiling was also performed for residual Amp(R), and adenovirus E2A and E4. Although trace amounts of DNA impurities were present in the clinical vector, transcription of these sequences was not detected after transduction of human hepatocytes, nor in mice administered a dose 26-fold above the highest dose administered in the clinical study. Two methods used to minimize encapsidated DNA impurities in the clinical vector were: (i) a vector (cis) production plasmid with a backbone exceeding the packaging limit of AAV; and (ii) a vector purification step that achieved separation of the vector from vector-related impurities (e.g., empty capsids). In conclusion, residual cap expression was undetectable following transduction with AAV2-hFIX clinical vectors. Preformed capsid protein is implicated as the source of epitopes recognized by CD8(+) T cells that eliminated vector-transduced cells in the clinical study.


Assuntos
Dependovirus/genética , Dependovirus/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Transcrição Gênica/genética , Transcrição Gênica/imunologia , Animais , Capsídeo/imunologia , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase
7.
Mol Ther ; 16(12): 1960-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18827805

RESUMO

We set out to analyze the fundamental biological differences between AAV2 and AAV8 that may contribute to their different performances in vivo. High-throughput protein interaction screens were used to identify binding partners for each serotype. Of the >8,000 proteins probed, 115 and 134 proteins were identified that interact with AAV2 and AAV8, respectively. Notably, 76 of these protein interactions were shared between the two serotypes. CDK2/cyclinA kinase was identified as a binding partner for both serotypes in the screen. Subsequent analysis confirmed direct binding of CDK2/cyclinA by AAV2 and AAV8. Inhibition of CDK2/cyclinA resulted in increased levels of vector transduction. Biophysical study of vector particle stability and genome uncoating demonstrated slightly greater thermostability for AAV8 than for AAV2. Heat-induced genome uncoating occurred at the same temperature as particle degradation, suggesting that these two processes may be intrinsically related for adeno-associated virus (AAV). Together, these analyses provide insight into commonalities and divergences in the biology of functionally distinct hepatotropic AAV serotypes.


Assuntos
Biofísica , Dependovirus/genética , Fígado/metabolismo , Anticorpos Monoclonais/imunologia , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/metabolismo , Ciclina A/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Dependovirus/imunologia , Dependovirus/metabolismo , Vetores Genéticos/genética , Genoma Viral/genética , Cinética , Ligação Proteica
9.
Mol Ther ; 16(1): 138-45, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17955024

RESUMO

Adeno-associated virus (AAV) vectors demonstrate highly efficient gene transfer to hepatocytes in vivo. One of the remaining obstacles to the treatment of hemophilia B patients with AAV vectors is the sensitivity of these vectors to antibody-mediated neutralization following systemic delivery. Testing and implementation of strategies to circumvent pre-existing antibodies requires knowledge of the clearance kinetics of AAV from circulation. In this study, AAV clearance kinetics were established for serotypes 2 and 8 in cell culture and in mice. Administration of pooled neutralizing serum subsequent to administration of the vector was used to define the time period in which the vector is susceptible to antibody-mediated neutralization. These experiments defined the in vivo clearance rates for both AAV2 and AAV8 vectors to be between 2 and 4 hours. In mice, portal vein and tail vein administration of each vector was tested with similar results. Cell culture studies in W162 cells established that cellular attachment and internalization both contribute to the clearance kinetics of AAV vectors. These studies characterize the in vivo clearance rates of AAV vectors for the first time and guide the development of future strategies for the avoidance of antibody-mediated AAV vector neutralization.


Assuntos
Anticorpos Antivirais/fisiologia , Dependovirus/imunologia , Vetores Genéticos/imunologia , Fígado/imunologia , Transdução Genética , Animais , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/sangue , Linhagem Celular , Vetores Genéticos/antagonistas & inibidores , Vetores Genéticos/metabolismo , Humanos , Soros Imunes/administração & dosagem , Soros Imunes/sangue , Soros Imunes/fisiologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Fatores de Tempo
10.
J Virol ; 81(19): 10777-85, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17634219

RESUMO

Infection with murine leukemia virus (MLV) TR1.3 or the related molecular construct W102G causes severe neuropathology in vivo. Infection is causally linked to the development of extensive syncytia in brain capillary endothelial cells (BCEC). These viruses also induce cell fusion of murine cell lines, such as SC-1 and NIH 3T3, which are otherwise resistant to MLV-induced syncytium formation. Although the virulence of these viruses maps within the env gene, the mechanism of fusion enhancement is not fully determined. To this end, we examined the capacity of the syncytium-inducing (SI) TR1.3 and W102G MLVs to overcome the fusion inhibitory activity inherent in the full-length Env cytoplasmic tail. These studies showed that the TR1.3 and W102G Envs did not induce premature cleavage of p2E, nor did they override p2E fusion inhibition. Indeed, in the presence of mutations that disrupt p2E function, the TR1.3 and W102G Envs significantly increased the extent of cell fusion compared to that with the non-syncytium-inducing MLV FB29. Surprisingly, we also observed that TR1.3 and W102G Envs failed to elicit syncytium formation in these in vitro assays. Coexpression of gag-pol with env restored syncytium formation, and accordingly, mutations within gag-pol were used to examine the minimal functional requirements for the SI phenotype. The results indicate that both gag-dependent particle budding and cleavage of p2E are required to activate the SI phenotype of TR1.3 and W102G viruses. Collectively, these data suggest that the TR1.3 and W102G viruses induce cell fusion by the fusion-from-without pathway.


Assuntos
Produtos do Gene env/metabolismo , Produtos do Gene gag/metabolismo , Células Gigantes/virologia , Vírus da Leucemia Murina/patogenicidade , Fusão de Membrana , Animais , Células Cultivadas , Proteínas de Fusão gag-pol/metabolismo , Produtos do Gene env/genética , Produtos do Gene gag/genética , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/metabolismo , Camundongos , Mutação
11.
Nat Med ; 13(4): 419-22, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17369837

RESUMO

Hepatic adeno-associated virus (AAV)-serotype 2 mediated gene transfer results in transgene product expression that is sustained in experimental animals but not in human subjects. We hypothesize that this is caused by rejection of transduced hepatocytes by AAV capsid-specific memory CD8(+) T cells reactivated by AAV vectors. Here we show that healthy subjects carry AAV capsid-specific CD8(+) T cells and that AAV-mediated gene transfer results in their expansion. No such expansion occurs in mice after AAV-mediated gene transfer. In addition, we show that AAV-2 induced human T cells proliferate upon exposure to alternate AAV serotypes, indicating that other serotypes are unlikely to evade capsid-specific immune responses.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Capsídeo/imunologia , Dependovirus/imunologia , Ativação Linfocitária/imunologia , Infecções por Parvoviridae/imunologia , Animais , Técnicas de Transferência de Genes , Hepatócitos/imunologia , Humanos , Camundongos , Peptídeos/imunologia
12.
Mol Ther ; 15(4): 792-800, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17245353

RESUMO

The goal of these studies was to test whether adeno-associated virus (AAV) capsid-specific CD8(+) T cells cause loss of hepatic AAV-mediated gene expression in experimental animals. Mice immunized with adenoviral vectors expressing AAV capsid or with AAV vectors developed CD8(+) T cells in blood, lymphatic tissues, and liver to epitopes shared between AAV2 and AAV8, and serotype-specific neutralizing antibodies. At the height of the T cells' effector phase, mice were infused with a heterologous AAV vector expressing human factor IX under a hepatocyte-specific promoter. Despite the presence of lytic CD8(+) T cells in the liver, hepatic Factor IX expression was sustained and comparable in AAV-preimmune and naïve animals. These results suggest that, in mice, pre-existing CD8(+) T cells to AAV capsid do not affect the longevity of AAV-mediated hepatic gene transfer. These results are in contrast to the outcome of a recent gene therapy trial of hemophilia B patients who were treated by hepatic gene transfer of AAV2 vectors expressing Factor IX. The loss of Factor IX expression, accompanied by a rise in liver enzymes and detectable frequencies of circulating AAV capsid-specific T cells, suggested T-cell-mediated destruction of transduced hepatocytes following reactivation of AAV-specific T cells upon AAV transfer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Capsídeo/imunologia , Dependovirus/genética , Dependovirus/imunologia , Hepatócitos/imunologia , Hepatócitos/virologia , Transdução Genética , Transferência Adotiva , Animais , Fator IX/genética , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos/imunologia , Hemofilia B/genética , Hemofilia B/imunologia , Hemofilia B/terapia , Humanos , Depleção Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID
13.
J Virol ; 80(9): 4601-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16611920

RESUMO

TR1.3 is a Friend murine leukemia virus (MLV) that induces selective syncytium induction (SI) of brain capillary endothelial cells (BCEC), intracerebral hemorrhage, and death. Syncytium induction by TR1.3 has been mapped to a single tryptophan-to-glycine conversion at position 102 of the envelope glycoprotein (Env102). The mechanism of SI by TR1.3 was examined here in comparison to the non-syncytium-inducing, nonpathogenic MLV FB29, which displays an identical BCEC tropism. Envelope protein expression and stability on both infected cells and viral particles were not statistically different for TR1.3 and FB29. However, affinity measurements derived using purified envelope receptor binding domain (RBD) revealed a reduction of >1 log in the K(D) of TR1.3 RBD relative to FB29 RBD. Whole-virus particles pseudotyped with TR1.3 Env similarly displayed a markedly reduced binding avidity compared to FB29-pseudotyped viral particles. Lastly, decreased receptor affinity of TR1.3 Env correlated with the failure to block superinfection following acute and chronic infection by TR1.3. These results definitively show that acquisition of a SI phenotype can be directly linked to amino acid changes in retroviral Env that decrease receptor affinity, thereby emphasizing the importance of events downstream of receptor binding in the cell fusion process and pathology.


Assuntos
Vírus da Leucemia Murina/metabolismo , Vírus da Leucemia Murina/patogenicidade , Receptores Virais/metabolismo , Superinfecção/virologia , Proteínas do Envelope Viral/metabolismo , Animais , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Linhagem Celular , Regulação Viral da Expressão Gênica , Humanos , Vírus da Leucemia Murina/classificação , Fusão de Membrana , Camundongos , Vírion/metabolismo
14.
J Virol Methods ; 128(1-2): 135-42, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15941597

RESUMO

Many viruses enter cells via an interaction of the viral envelope glycoprotein (Env) with receptor inducing fusion of viral and cellular membranes. These interactions are often evaluated in cell-cell fusion, gene-reporting systems with effector cells expressing Env and target cells expressing receptors. A common system utilizes vaccinia virus encoding T7 RNA polymerase (RNAP) in effector cells and a T7 promoted reporter plasmid in target cells. Fusion is quantified with expression of the reporter plasmid. However, direct activation of reporter plasmid from vaccinia virus can occur increasing background activity. We report here a modification of this assay in which T7 RNAP is expressed from a plasmid rather than vaccinia. This modification increased sensitivity with a ten-fold reduction in background. A novel dual T7/SP6 RNAP fusion assay was also developed to allow rapid screening for functional Env clones. Using these assays, we show that Envs from two CD4-independent HIV-2 isolates (VCP and ROD/B), which are able to fuse with chemokine receptor CXCR4 in a CD4-independent manner, are also able to fuse with alternative coreceptors GPR1 and GPR15 in the absence of CD4. The assay could also detect fusion of murine leukemia virus on target cells expressing the ecotropic MCAT-1 receptor showing its broad utility in other viral systems.


Assuntos
Fusão Celular/métodos , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Produtos do Gene env/metabolismo , HIV-2/patogenicidade , Plasmídeos , Proteínas Virais/genética , Proteínas Virais/metabolismo , Animais , Antígenos CD4/metabolismo , Linhagem Celular , Produtos do Gene env/genética , HIV-2/genética , HIV-2/metabolismo , Humanos , Vírus da Leucemia Murina/metabolismo , Vírus da Leucemia Murina/patogenicidade , Camundongos , Plasmídeos/genética , Receptores CXCR4/metabolismo , Receptores de HIV/metabolismo
15.
J Virol ; 78(15): 8392-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15254211

RESUMO

The murine leukemia virus (MLV) TR1.3 provides an excellent model to study the wide range of retrovirus-induced central nervous system (CNS) pathology and disease. TR1.3 rapidly induces thrombotic events in brain microvessels and causes cell-specific syncytium formation of brain capillary endothelial cells (BCEC). A single amino acid substitution, W102G, in the MLV envelope protein (Env) regulates the pathogenic effects. The role of Env in determining this disease phenotype compared to the induction of spongiform encephalomyelitis with a longer latency, as seen in several other MLV and in human retroviruses, was determined by studying in vitro-attenuated TR1.3. Virus cloned from this selection, termed TRM, induced progressive neurological disease characterized by ataxia and paralysis and the appearance of spongiform neurodegeneration throughout the brain stem and spinal cord. This disease was associated with virus replication in both BCEC and highly ramified glial cells. TRM did not induce syncytium formation, either in vivo or in vitro. Sequence and mutational analyses demonstrated that TRM contained a reversion of Env G102W but that neurological disease mapped to the single amino acid substitution Env S159P. The results demonstrate that single nucleotide changes within disparate regions of Env control dramatically different CNS disease patterns.


Assuntos
Doenças do Sistema Nervoso Central/etiologia , Vírus da Leucemia Murina/patogenicidade , Proteínas do Envelope Viral/química , Animais , Linhagem Celular , Doenças do Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso Central/virologia , Coturnix , Feminino , Fusão de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Tropismo , Proteínas do Envelope Viral/fisiologia
16.
AIDS ; 17(1): 33-42, 2003 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-12478067

RESUMO

OBJECTIVE: Under some circumstances the HIV virus may infect cells that do not express receptors essential to HIV-entry. We hypothesized that platelet- and megakaryocyte-derived microparticles (MP) could play a role in such infections. MP are circular membrane fragments shed from the surface of eukaryotic cells. After adhesion to target cells, MP may transfer membrane-associated proteins to these cells. We found that peripheral blood platelet- (PMP) and megakaryocyte-derived MP (MegaMP) that highly express CXCR4 may transfer this receptor from the surface of platelets or megakaryocytes to the surface of CXCR4-null cells. DESIGN: Since this mechanism could potentially allow CD4+/CXCR4-null cells to become infected by T-tropic HIV, we incubated several human CD4+/CXCR4-null cells such as normal erythroblasts, glioblastomas U87, MAGI and hematopoietic cell lines UT-7, HEL and TF-1 with PMP or MegaMP. We found that these cells became CXCR4+. We next exposed these cells to X4-HIV (IIIB) and evaluated their susceptibility to infection by PCR, ELISA, and morphological analysis. RESULTS: We observed in all instances that after CD4+/CXCR4-null cell lines 'acquired' CXCR4 from PMP or MegaMP, they could became infected by X4 HIV. CONCLUSIONS: We postulate that both PMP and MegaMP may play a novel and important role in spreading HIV-1 infection by transferring the CXCR4 co-receptor to CD4+/CXCR4-null cells.


Assuntos
Plaquetas/metabolismo , Infecções por HIV/metabolismo , HIV-1/patogenicidade , Megacariócitos/metabolismo , Receptores CXCR4/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Infecções por HIV/virologia , Humanos , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Reação em Cadeia da Polimerase/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA