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.
Neurochem Res ; 49(2): 388-401, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37847329

RESUMO

Endoplasmic reticulum (ER) dysfunction caused by excessive ER stress is a crucial mechanism underlying seizures-induced neuronal injury. Studies have shown that mitochondrial reactive oxygen species (ROS) are closely related to ER stress, and our previous study showed that activating transcription factor 5 (ATF5)-regulated mitochondrial unfolded protein response (mtUPR) modulated mitochondrial ROS generation in a hippocampal neuronal culture model of seizures. However, the effects of ATF5-regulated mtUPR on ER stress and the underlying mechanisms remain uncertain in epilepsy. In this study, ATF5 upregulation by lentivirus infection attenuated seizures-induced neuronal damage and apoptosis in a rat model of pilocarpine-induced epilepsy, whereas ATF5 downregulation by lentivirus infection had the opposite effects. ATF5 upregulation potentiated mtUPR by increasing the expression of mitochondrial chaperone heat shock protein 60 (HSP60) and caseinolytic protease proteolytic subunit (ClpP) and reducing mitochondrial ROS generation in pilocarpine-induced seizures in rats. Additionally, upregulation of ATF5 reduced the expression of glucose-regulated protein 78 (GRP78), protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor 4 (ATF4), and C/EBP homologous protein (CHOP), suggesting suppression of ER stress; Moreover, ATF5 upregulation attenuated apoptosis-related proteins such as B-cell lymphoma-2 (BCL2) downregulation, BCL2-associated X (BAX) and cleaved-caspase-3 upregulation. However, ATF5 downregulation exerted the opposite effects. Furthermore, pretreatment with the mitochondria-targeted antioxidant mito-TEMPO attenuated the harmful effects of ATF5 downregulation on ER stress and neuronal apoptosis by reducing mitochondrial ROS generation. Overall, our study suggested that ATF5-regulated mtUPR exerted neuroprotective effects against pilocarpine-induced seizures in rats and the underlying mechanisms might involve mitochondrial ROS-mediated ER stress.


Assuntos
Epilepsia , Infecções por Lentivirus , Ratos , Animais , Espécies Reativas de Oxigênio/metabolismo , Pilocarpina/toxicidade , Estresse do Retículo Endoplasmático , Resposta a Proteínas não Dobradas , Apoptose , Mitocôndrias/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Epilepsia/induzido quimicamente , Epilepsia/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Convulsões/induzido quimicamente , Convulsões/metabolismo , Neurônios/metabolismo , Infecções por Lentivirus/metabolismo
2.
Eur J Med Res ; 27(1): 5, 2022 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-35022077

RESUMO

BACKGROUND: Intervertebral disc degeneration (IDD) is a natural progression of age-related processes. Associated with IDD, degenerative disc disease (DDD) is a pathologic condition implicated as a major cause of chronic lower back pain, which can have a severe impact on the quality of life of patients. As degeneration progression is associated with elevated levels of inflammatory cytokines, enhanced aggrecan and collagen degradation, and changes in the disc cell phenotype. The purpose of this study was to investigate the biological and cytological characteristics of rabbit nucleus pulposus mesenchymal stem cells (NPMSCs)-a key factor in IDD-and to determine the effect of the growth and differentiation factor-5 (GDF5) on the differentiation of rabbit NPMSCs transduced with a lentivirus vector. METHODS: An in vitro culture model of rabbit NPMSCs was established and NPMSCs were identified by flow cytometry (FCM) and quantitative real-time PCR (qRT-PCR). Subsequently, NPMSCs were randomly divided into three groups: a transfection group (the lentiviral vector carrying GDF5 gene used to transfect NPMSCs); a control virus group (the NPMSCs transfected with an ordinary lentiviral vector); and a normal group (the NPMSCs alone). FCM, qRT-PCR, and western blot (WB) were used to detect the changes in NPMSCs. RESULTS: The GDF5-transfected NPMSCs displayed an elongated shape, with decreased cell density, and significantly increased GDF5 positivity rate in the transfected group compared to the other two groups (P < 0.01). The mRNA levels of Krt8, Krt18, and Krt19 in the transfected group were significantly higher in comparison with the other two groups (P < 0.01), and the WB results were consistent with that of qRT-PCR. CONCLUSIONS: GDF5 could induce the differentiation of NPMSCs. The lentiviral vector carrying the GDF5 gene could be integrated into the chromosome genome of NPMSCs and promoted differentiation of NPMSCs into nucleus pulposus cells. Our findings advance the development of feasible and effective therapies for IDD.


Assuntos
Regulação da Expressão Gênica , Fator 5 de Diferenciação de Crescimento/genética , Infecções por Lentivirus/virologia , Lentivirus , Células-Tronco Mesenquimais/citologia , Núcleo Pulposo/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Fator 5 de Diferenciação de Crescimento/biossíntese , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/patologia , Células-Tronco Mesenquimais/virologia , Núcleo Pulposo/patologia , Núcleo Pulposo/virologia , Coelhos
3.
Viruses ; 12(6)2020 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-32471198

RESUMO

Mammals have developed clever adaptive and innate immune defense mechanisms to protect against invading bacterial and viral pathogens. Human innate immunity is continuously evolving to expand the repertoire of restriction factors and one such family of intrinsic restriction factors is the APOBEC3 (A3) family of cytidine deaminases. The coordinated expression of seven members of the A3 family of cytidine deaminases provides intrinsic immunity against numerous foreign infectious agents and protects the host from exogenous retroviruses and endogenous retroelements. Four members of the A3 proteins-A3G, A3F, A3H, and A3D-restrict HIV-1 in the absence of virion infectivity factor (Vif); their incorporation into progeny virions is a prerequisite for cytidine deaminase-dependent and -independent activities that inhibit viral replication in the host target cell. HIV-1 encodes Vif, an accessory protein that antagonizes A3 proteins by targeting them for polyubiquitination and subsequent proteasomal degradation in the virus producing cells. In this review, we summarize our current understanding of the role of human A3 proteins as barriers against HIV-1 infection, how Vif overcomes their antiviral activity, and highlight recent structural and functional insights into A3-mediated restriction of lentiviruses.


Assuntos
Desaminases APOBEC/fisiologia , Infecções por Lentivirus/imunologia , Lentivirus/imunologia , Desaminases APOBEC/química , Animais , Sequência de Bases , Humanos , Lentivirus/metabolismo , Infecções por Lentivirus/metabolismo , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
4.
Sci Rep ; 8(1): 15036, 2018 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-30310119

RESUMO

Lentiviral vectors are a valuable tool to deliver exogenous genes for stable expression in cells. While much progress has been made in processing lentiviral vector-containing culture medium, it remains to be explored how the production of lentiviral vector from producer cells can be increased. We initially found that co-expression of the SPRY domain-containing SOCS box protein 1 (SPSB1) promotes the production of human immunodeficiency virus type 1 (HIV-1) and lentiviral vector with increased expression of the Gag and envelope proteins and activation of the HIV-1 LTR and CMV promoter. The presence of AP-1, NF-κB and CREB/ATF recognition sites in these promoters prompted us to utilize human T-lymphotropic virus type 1 (HTLV-1) Tax for lentiviral vector production because Tax activates all these transcription factors. Co-expression of a small amount of Tax markedly increased both the expression of viral structural proteins in producer cells and release of lentiviral vector particles, resulting in a more than 10-fold enhancement of transduction efficiency. Of note, the Tax protein was not detected in the lentiviral vector particles concentrated by ultracentrifugation, supporting the safety of this preparation. Collectively, these results indicate that promoter activation in producer cells represents a promising approach to preparing high-titer lentiviral vectors.


Assuntos
Engenharia Genética , Vetores Genéticos/genética , Lentivirus/fisiologia , Regiões Promotoras Genéticas , Replicação Viral , Linhagem Celular , Produtos do Gene tax/metabolismo , Técnicas de Transferência de Genes , HIV-1/genética , Interações Hospedeiro-Patógeno , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/virologia , Ligação Proteica , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Sequências Repetidas Terminais , Transdução Genética , Transfecção
5.
Mol Ther ; 26(2): 634-647, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29221804

RESUMO

Efficient transduction tools are a hallmark for both research and therapy development. Here, we introduce new insights into the generation of lentiviral vectors with improved performance by utilizing producer cells with increased production rates of extracellular vesicles through CD9 overexpression. Most human cells secrete small vesicles from their surface (microvesicles) or intraluminal endosome-derived membranes (exosomes). In particular, enhanced levels of the tetraspanin CD9 result in significantly increased numbers of extracellular vesicles with exosome-like features that were secreted from four different human cell lines. Intriguingly, exosomes and their biogenesis route display similarities to lentivirus and we examined the impact of CD9 expression on release and infectivity of recombinant lentiviral vectors. Although the titers of released viral particles were not increased upon production in high CD9 cells, we observed improved performance in terms of both speed and efficiency of lentiviral gene delivery into numerous human cell lines, including HEK293, HeLa, SH-SY5Y, as well as B and T lymphocytes. Here, we demonstrate that enhanced CD9 enables lentiviral transduction in the absence of any pseudotyping viral glycoprotein or fusogenic molecule. Our findings indicate an important role of CD9 for lentiviral vector and exosome biogenesis and point out a remarkable function of this tetraspanin in membrane fusion, viral infectivity, and exosome-mediated horizontal information transfer.


Assuntos
Exossomos/metabolismo , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/virologia , Lentivirus/fisiologia , Tetraspanina 29/metabolismo , Biomarcadores , Linhagem Celular , Vesículas Extracelulares/metabolismo , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Infecções por Lentivirus/genética , Tetraspanina 29/genética , Proteínas do Envelope Viral/metabolismo
6.
Mol Med Rep ; 10(5): 2249-54, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25174942

RESUMO

The present study aimed to investigate the effects of lentiviral infection of human umbilical cord mesenchymal stem cells (hUCMSCs) on the expression of octamer transcription factor 4 (Oct4). hUCMSCs were infected with lentivirus carrying the green fluorescent protein gene (GFP) at different multiplicities of infection (MOI), and the optimal MOI was determined by flow cytometry; the proliferation of non­infected and GFP-carrying lentivirus­infected hUCMSCs was evaluated by the MTT assay; and the expression of the Oct4 gene was measured by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and immunofluorescence staining in hUCMSCs cultured in vitro for eight weeks. Positive GFP staining of hUCMSCs was estimated at >75% at 48 h following infection with the GFP-carrying lentivirus (MOI=20); no effect on hUCMSC proliferation was detected by the MTT assay following the infection; immunofluorescence analysis detected positive Oct4 expression in the cell nuclei at two and eight weeks of culture, while the relative expression of Oct4 assessed by qRT-PCR was 0.9075±0.0124. The GFP gene carried by the lentivirus was successfully expressed in hUCMSCs and had no significant effect on Oct4 expression, which lays a solid foundation for future studies investigating gene functions via the use of exogenous markers.


Assuntos
Infecções por Lentivirus/metabolismo , Lentivirus/fisiologia , Células-Tronco Mesenquimais/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Proliferação de Células , Forma Celular , Células Cultivadas , Expressão Gênica , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Interações Hospedeiro-Patógeno , Humanos , Células-Tronco Mesenquimais/virologia , MicroRNAs/genética , Fator 3 de Transcrição de Octâmero/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução Genética
7.
J Virol ; 88(11): 6268-80, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24672033

RESUMO

UNLABELLED: Epithelial barrier dysfunction during human immunodeficiency virus (HIV) infection has largely been attributed to the rapid and severe depletion of CD4(+) T cells in the gastrointestinal (GI) tract. Although it is known that changes in mucosal gene expression contribute to intestinal enteropathy, the role of small noncoding RNAs, specifically microRNA (miRNA), has not been investigated. Using the simian immunodeficiency virus (SIV)-infected nonhuman primate model of HIV pathogenesis, we investigated the effect of viral infection on miRNA expression in intestinal mucosa. SIV infection led to a striking decrease in the expression of mucosal miRNA compared to that in uninfected controls. This decrease coincided with an increase in 5'-3'-exoribonuclease 2 protein and alterations in DICER1 and Argonaute 2 expression. Targets of depleted miRNA belonged to molecular pathways involved in epithelial proliferation, differentiation, and immune response. Decreased expression of several miRNA involved in maintaining epithelial homeostasis in the gut was localized to the proliferative crypt region of the intestinal epithelium. Our findings suggest that SIV-induced decreased expression of miRNA involved in epithelial homeostasis, disrupted expression of miRNA biogenesis machinery, and increased expression of XRN2 are involved in the development of epithelial barrier dysfunction and gastroenteropathy. IMPORTANCE: MicroRNA (miRNA) regulate the development and function of intestinal epithelial cells, and many viruses disrupt normal host miRNA expression. In this study, we demonstrate that SIV and HIV disrupt expression of miRNA in the small intestine during infection. The depletion of several key miRNA is localized to the proliferative crypt region of the gut epithelium. These miRNA are known to control expression of genes involved in inflammation, cell death, and epithelial maturation. Our data indicate that this disruption might be caused by altered expression of miRNA biogenesis machinery during infection. These findings suggest that the disruption of miRNA in the small intestine likely plays a role in intestinal enteropathy during HIV infection.


Assuntos
HIV , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatologia , Infecções por Lentivirus/metabolismo , MicroRNAs/metabolismo , Vírus da Imunodeficiência Símia , Adulto , Animais , Sequência de Bases , Linfócitos T CD4-Positivos/imunologia , Biologia Computacional , Densitometria , Citometria de Fluxo , Humanos , Mucosa Intestinal/imunologia , Microdissecção e Captura a Laser , Infecções por Lentivirus/fisiopatologia , Macaca mulatta , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Dados de Sequência Molecular , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Carga Viral
8.
J Virol ; 88(6): 3255-72, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24390322

RESUMO

UNLABELLED: BST2/tetherin inhibits the release of enveloped viruses from cells. Primate lentiviruses have evolved specific antagonists (Vpu, Nef, and Env). Here we characterized tetherin proteins of species representing both branches of the order Carnivora. Comparison of tiger and cat (Feliformia) to dog and ferret (Caniformia) genes demonstrated that the tiger and cat share a start codon mutation that truncated most of the tetherin cytoplasmic tail early in the Feliformia lineage (19 of 27 amino acids, including the dual tyrosine motif). Alpha interferon (IFN-α) induced tetherin and blocked feline immunodeficiency virus (FIV) replication in lymphoid and nonlymphoid feline cells. Budding of bald FIV and HIV particles was blocked by carnivore tetherins. However, infectious FIV particles were resistant, and spreading FIV replication was uninhibited. Antagonism mapped to the envelope glycoprotein (Env), which rescued FIV from carnivore tetherin restriction when expressed in trans but, in contrast to known antagonists, did not rescue noncognate particles. Also unlike the primate lentiviral antagonists, but similar to the Ebola virus glycoprotein, FIV Env did not reduce intracellular or cell surface tetherin levels. Furthermore, FIV-enveloped FIV particles actually required tetherin for optimal release from cells. The results show that FIV Envs mediate a distinctive tetherin evasion. Well adapted to a phylogenetically ancient tetherin tail truncation in the Felidae, it requires functional virion incorporation of Env, and it shields the budding particle without downregulating plasma membrane tetherin. Moreover, FIV has evolved dependence on this protein: particles containing FIV Env need tetherin for optimal release from the cell, while Env(-) particles do not. IMPORTANCE: HIV-1 antagonizes the restriction factor tetherin with the accessory protein Vpu, while HIV-2 and the filovirus Ebola use their envelope (Env) glycoproteins for this purpose. It turns out that the FIV tetherin antagonist is also its Env protein, but the mechanism is distinctive. Unlike other tetherin antagonists, FIV Env cannot act in trans to rescue vpu-deficient HIV-1. It must be incorporated specifically into FIV virions to be active. Also unlike other retroviral antagonists, but similar to Ebola virus Env, it does not act by downregulating or degrading tetherin. FIV Env might exclude tetherin locally or direct assembly to tetherin-negative membrane domains. Other distinctive features are apparent, including evidence that this virus evolved an equilibrium in which tetherin is both restriction factor and cofactor, as FIV requires tetherin for optimal particle release.


Assuntos
Antígenos CD/metabolismo , Doenças do Gato/metabolismo , Vírus da Imunodeficiência Felina/metabolismo , Infecções por Lentivirus/veterinária , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo , Sequência de Aminoácidos , Animais , Antígenos CD/química , Antígenos CD/genética , Doenças do Gato/genética , Doenças do Gato/virologia , Gatos , Cães , Vírus da Imunodeficiência Felina/química , Vírus da Imunodeficiência Felina/genética , Infecções por Lentivirus/genética , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/virologia , Dados de Sequência Molecular , Alinhamento de Sequência , Proteínas do Envelope Viral/genética , Vírion/genética
9.
Cancer Biol Ther ; 15(1): 108-18, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24153116

RESUMO

In recent years, the RNA-binding protein quaking 5 (QKI-5) has been recognized as a novel tumor suppressor in many cancers. To date, no studies have examined the role of QKI-5 in prostate cancer. The present study was designed to elucidate the correlation of QKI-5 expression with the clinical pathological features and prognosis of prostate cancer. In an overwhelming majority of the 184 cases of prostate cancer samples analyzed, the QKI-5 expression was significantly decreased, which was largely due to the high promoter methylation levels. Using lentiviral vectors, we established two stable prostate cancer cell lines with altered QKI-5 expression, including a QKI-5 overexpressing PC3 cell line and a DU145 cell line with knocked-down QKI-5 expression. The effects of the lentiviral-mediated QKI-5 knockdown on the PC3 cells and DU145 cells were assessed by cell growth curves, flow cytometry (FCM), and an invasion assay. The PC3 cells were transplanted into nude mice, and then, the tumor growth curves and TUNEL staining were determined. These results demonstrated that QKI-5 was highly expressed in benign prostatic hyperplasia (BPH) tissues but not in carcinomatous tissues and that QKI-5 effectively inhibited prostate cancer cell proliferation in vitro and in vivo. In addition, the decrease in QKI-5 expression was closely correlated with the prostate cancer Gleason score, poor differentiation, degree of invasion, lymph node metastasis, distant metastasis, TNM grading, and poor survival. These results indicate that the QKI-5 expression may be a novel, independent factor in the prognosis of prostate cancer patients.


Assuntos
Neoplasias da Próstata/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA , Humanos , Infecções por Lentivirus/metabolismo , Masculino , Camundongos , Camundongos Nus , Prognóstico , Regiões Promotoras Genéticas , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/patologia , Proteínas de Ligação a RNA/genética , Proteínas Supressoras de Tumor/genética
10.
J Exp Clin Cancer Res ; 32: 70, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-24294976

RESUMO

INTRODUCTION: Inflammatory breast cancer (IBC) is the most metastatic variant of breast cancer with the poorest survival in all types of breast cancer patients and presently therapeutic targets for IBC are very limited. Enhancer of zeste homolog 2 (EZH2) is frequently expressed in human IBC and its expression positively correlates with worse clinical outcome. However, the molecular basis for EZH2 promoting IBC has not been explored. Here, we investigated the functional role of EZH2 in IBC cells by examining the effects of its knockdown on the formation of tumor spheroids and invasion of these cells in vitro and in vivo in an orthotopic xenograft model. METHODS: SUM149 and a new IBC cell line-FC-IBC-02 derived from pleural effusion fluid of an IBC patient were used in this study. Specific knockdown of EZH2 was performed using short hairpin RNA (shRNA) specific to the human EZH2 gene. Cell growth and the formation of tumor spheroids were examined in vitro. The effects of EZH2 knockdown on IBC cell migration and invasion were examined by a Boyden chamber assay. For the in vivo tumor growth studies, IBC cells were orthotopically transplanted into the mammary fat pads of immunodeficient mice. RESULTS: The results showed that EZH2 is expressed at higher levels in human IBC cell lines compared with normal human mammary epithelial cells, and the knockdown of EZH2 expression significantly suppressed cell growth and tumor spheroid formation of human IBC cells in vitro. In addition, EZH2 knockdown inhibited the migration and invasion of IBC cells. Significantly, EZH2 knockdown suppressed the angiogenesis and tumor growth of IBC cells in vivo. CONCLUSIONS: Our results provide direct evidence that EZH2 is critical for the formation of tumor spheroids and invasion of human IBC cells and could be a potential target for developing novel therapeutic strategies for human IBC.


Assuntos
Neoplasias Inflamatórias Mamárias/genética , Complexo Repressor Polycomb 2/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Inflamatórias Mamárias/patologia , Infecções por Lentivirus/genética , Infecções por Lentivirus/metabolismo , Camundongos , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Complexo Repressor Polycomb 2/genética , Esferoides Celulares , Análise de Sobrevida
11.
J Virol ; 86(22): 12053-68, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22933280

RESUMO

Productive replication of human immunodeficiency virus type 1 (HIV-1) occurs efficiently only in humans. The posttranscriptional stages of the HIV-1 life cycle proceed poorly in mouse cells, with a resulting defect in viral assembly and release. Previous work has shown that the presence of human chromosome 2 increases HIV-1 production in mouse cells. Recent studies have shown that human chromosome region maintenance 1 (hCRM1) stimulates Gag release from rodent cells. Here we report that expressions of hCRM1 in murine cells resulted in marked increases in the production of infectious HIV-1 and feline immunodeficiency virus (FIV). HIV-1 production was also increased by hSRp40, and a combination of hCRM1 and hSRp40 resulted in a more-than-additive effect on HIV-1 release. In contrast, the overexpression of mouse CRM1 (mCRM1) minimally affected HIV-1 and FIV production and did not antagonize hCRM1. In the presence of hCRM1 there were large increases in the amounts of released capsid, which paralleled the increases in the infectious titers. Consistent with this finding, the ratios of unspliced to spliced HIV-1 mRNAs in mouse cells expressing hCRM1 and SRp40 became similar to those of human cells. Furthermore, imaging of intron-containing FIV RNA showed that hCRM1 increased RNA export to the cytoplasm.By testing chimeras between mCRM1 and hCRM1 and comparing those sequences to feline CRM1, we mapped the functional domain to HEAT (Huntingtin, elongation factor 3, protein phosphatase 2A, and the yeast kinase TOR1) repeats 4A to 9A and a triple point mutant in repeat 9A, which showed a loss of function. Structural analysis suggested that this region of hCRM1 may serve as a binding site for viral or cellular factors to facilitate lentiviral RNA nuclear export.


Assuntos
Infecções por HIV/metabolismo , HIV/metabolismo , Vírus da Imunodeficiência Felina/metabolismo , Carioferinas/fisiologia , Infecções por Lentivirus/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Transporte Ativo do Núcleo Celular , Alelos , Animais , Proteínas de Ciclo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Íntrons , Carioferinas/metabolismo , Camundongos , Conformação Molecular , Plasmídeos/metabolismo , RNA Viral/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Processamento de Serina-Arginina , Transfecção , Proteína Exportina 1
12.
J Immunol ; 185(11): 6876-82, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21041724

RESUMO

Interactions between the Fc segment of IgG and FcγRs on a variety of cells are likely to play an important role in the anti-HIV activity of Abs. Because the nature of the glycan structure on the Fc domain is a critical determinant of Fc-FcγR binding, proper Fc glycosylation may contribute to Ab-mediated protection. We have generated five different glycoforms of the broadly HIV-1-neutralizing mAb 2G12 in wild-type and glycoengineered plants and Chinese hamster ovary cells. Plant-derived 2G12 exhibited highly homogeneous glycosylation profiles with a single dominant N-glycan species. Using flow cytometry with FcγR-expressing cell lines, all 2G12 glycoforms demonstrated similar binding to FcγRI, FcγRIIa, and FcγRIIb. In contrast, two glycoforms derived from glycoengineered plants that lack plant-specific xylose and core α1,3-fucose, and instead carry human-like glycosylation with great uniformity, showed significantly enhanced binding to FcγRIIIa compared with Chinese hamster ovary or wild-type plant-derived 2G12. Using surface plasmon resonance, we show that binding of 2G12 to FcγRIIIa is markedly affected by core fucose, irrespective of its plant-specific α1,3 or mammalian-type α1,6 linkage. Consistent with this finding, 2G12 glycoforms lacking core fucose (and xylose) mediated higher antiviral activity against HIV-1 or simian immunodeficiency virus as measured by Ab-dependent cell-mediated virus inhibition. This is, to our knowledge, the first demonstration that specific alterations of Fc glycosylation can improve antiviral activity. Such alterations may result in better immunotherapeutic reagents. Moreover, biasing vaccine-induced immune responses toward optimal Fc glycosylation patterns could result in improved vaccine efficacy.


Assuntos
Fármacos Anti-HIV/farmacologia , Anticorpos Monoclonais/farmacologia , HIV-1/imunologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Receptores de IgG/metabolismo , Vacinas contra a AIDS/imunologia , Animais , Fármacos Anti-HIV/metabolismo , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Sítios de Ligação de Anticorpos/imunologia , Anticorpos Amplamente Neutralizantes , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Glicosilação , Anticorpos Anti-HIV , HIV-1/genética , HIV-1/crescimento & desenvolvimento , Humanos , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/prevenção & controle , Testes de Neutralização/métodos , Ligação Proteica/imunologia , Receptores de IgG/biossíntese , Receptores de IgG/genética , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Vírus da Imunodeficiência Símia/imunologia , Nicotiana/genética , Nicotiana/imunologia
13.
J Neurovirol ; 15(2): 139-52, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19115133

RESUMO

Infection by multiple lentiviral strains is recognized as a major driving force in the human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS) epidemic, but the neuropathogenic consequences of multivirus infections remain uncertain. Herein, we investigated the neurovirulence and underlying mechanisms of dual lentivirus infections with distinct viral strains. Experimental feline immunodeficiency virus (FIV) infections were performed using cultured cells and an in vivo model of AIDS neuropathogenesis. Dual infections were comprised of two FIV strains (FIV-Ch and FIV-PPR) as copassaged or superinfected viruses, with subsequent outcome analyses of host immune responses, viral load, neuropathological features, and neurobehavioral performance. Dual infections of feline macrophages resulted in greater IL-1beta (interleukin-1beta), TNF-alpha (tumor necrosis factor alpha), and IDO (indoleamine 2,3-dioxygenase) expression and associated neurotoxic properties. FIV coinfection and sequential superinfection in vivo also induced greater IL-1beta, TNF-alpha, and IDO expression in the basal ganglia (BG) and cortex (CTX), compared to the monovirus- and mock-infected groups, although viral loads were similar in single virus- and dual virus-infected animals. Immunoblot analyses disclosed lower synaptophysin immunoreactivity in the CTX resulting from FIV super- and coinfections. Cholinergic and GABAergic neuronal injury was evident in the CTX of animals with dual FIV infections. With increased glial activation and neuronal loss in dual FIV-infected brains, immunohistochemical analysis also revealed elevated detection of cleaved caspase-3 in dysmorphic neurons, which was associated with worsened neurobehavioral abnormalities among animals infected with the copassaged viruses. Dual lentivirus infections caused an escalation in neuroinflammation and ensuing neurodegeneration, underscoring the contribution of infection by multiple viruses to neuropathogenesis.


Assuntos
Vírus da Imunodeficiência Felina/patogenicidade , Inflamação/virologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Degeneração Neural/virologia , Animais , Gatos , Técnicas de Cultura de Células , Cérebro/metabolismo , Feminino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-1beta/biossíntese , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/metabolismo , Contagem de Linfócitos , Degeneração Neural/imunologia , Degeneração Neural/metabolismo , Gravidez , Sinaptofisina/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Virulência
14.
Bull Exp Biol Med ; 145(1): 133-6, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19024021

RESUMO

We developed a method for gene transfer into mesenchymal stromal cells. Lentivirus vector containing green fluorescent protein gene for labeling stromal and hemopoietic precursor cells was obtained using two plasmid sets from different sources. The vector was injected into the femur of mice in vivo and added into culture medium for in vitro infection of the stromal sublayer of long-term bone marrow culture. From 25 to 80% hemopoietic stem cells forming colonies in the spleen were infected with lentivirus vector in vivo and in vitro. Fibroblast colony-forming cells from the femoral bones of mice injected with the lentivirus vector carried no marker gene. The marker gene was detected in differentiated descendants from mesenchymal stem cells (bone cavity cells from the focus of ectopic hemopoiesis formed after implantation of the femoral bone marrow cylinder infected with lentivirus vector under the renal capsule of syngeneic recipient). In in vitro experiments, the marker gene was detected in sublayers of long-term bone marrow cultures infected after preliminary 28-week culturing, when hemopoiesis was completely exhausted. The efficiency of infection of stromal precursor cells depended on the source of lentivirus. The possibility of transfering the target gene into hemopoietic precursor cells in vivo is demonstrated. Stromal precursor cells can incorporate the provirus in vivo and in vitro, but conditions and infection system for effective infection should be thoroughly selected.


Assuntos
Células da Medula Óssea/virologia , Técnicas de Transferência de Genes , Vetores Genéticos/metabolismo , Células-Tronco Hematopoéticas/virologia , Lentivirus/metabolismo , Células Estromais/virologia , Animais , Células da Medula Óssea/fisiologia , Linhagem Celular , Feminino , Fêmur/citologia , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Humanos , Lentivirus/genética , Infecções por Lentivirus/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Estromais/fisiologia
15.
Virology ; 375(2): 452-63, 2008 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-18358512

RESUMO

Caprine arthritis-encephalitis virus (CAEV) is a lentivirus that causes natural inflammatory disease in goats, with chronic lesions in several different organs. CAEV infection of in vitro cultured cells is accompanied by apoptosis, but the involvement of the intrinsic and extrinsic pathways has not previously been elucidated. We have studied the activation of caspases-3, -8 and -9 by fluorescent assays in various goat cells infected in vitro by CAEV, and the effects of transfected dominant negative variants of theses caspases, to show that CAEV-associated apoptosis depends on activation of caspases-3 and -9, but not -8. A simultaneous disruption of mitochondrial membrane potential indicates an involvement of mitochondrial pathway.


Assuntos
Apoptose , Vírus da Artrite-Encefalite Caprina/fisiologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/virologia , Animais , Caspase 8/metabolismo , Células Cultivadas , Cabras , Macrófagos , Mitocôndrias/metabolismo , Transdução de Sinais , Replicação Viral
16.
Acta Microbiol Immunol Hung ; 53(1): 51-75, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16696550

RESUMO

The lentiviral protein Nef recruits cellular signalling proteins to lipid rafts at the cell membrane and acts thereby as a master regulator affecting the transcription of a series of cellular genes. By activating resting T cells, Nef creates an optimal environment for lentivirus replication. In human immunodeficiency virus (HIV) infected macrophages and microglial cells Nef activates the production of T-cell attracting chemokines and contributes to the development HIV infection associated brain damage. Nef also functions as an adaptor or connector protein downregulating CD4 and CCR5, the key receptor and one of the coreceptors for HIV. It also downregulates cell surface expression of a subset of class I MHC molecules which contributes to viral immune evasion. Extracellular, soluble Nef may facilitate the spread of T-cell-tropic HIV variants and mediate a switch in dominant replicating HIV strains (from macrophage-tropic to T-cell-tropic viruses) in AIDS (acquired immunodeficiency syndrome) patients. Virion-bound Nef enhances infectivity. Nef is a potential target of antiretroviral therapy and nef-deleted (attenuated) retroviruses have been considered as candidate vaccines against HIV. We suggest that nef-deleted or highly mutated defective HIV (dHIV) genomes interfere with replication of "wild type" HIV in certain long-term non-progressor individuals. This implies that introduction of artificially constructed dHIV genomes (by infusion of leukocytes carrying dHIV proviruses) into HIV infected individuals could slow disease progression and could be considered as a therapeutic possibility.


Assuntos
Produtos do Gene nef/fisiologia , Infecções por Lentivirus/virologia , Lentivirus/fisiologia , Síndrome da Imunodeficiência Adquirida/terapia , Animais , Encéfalo/patologia , Antígenos CD4/metabolismo , Quimiocinas/imunologia , Vírus Defeituosos , Produtos do Gene nef/deficiência , Produtos do Gene nef/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Lentivirus/patogenicidade , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/prevenção & controle , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Microglia/imunologia , Microglia/virologia , Receptores CCR5/metabolismo , Receptores Virais/metabolismo , Linfócitos T/imunologia , Virulência , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana
17.
Virology ; 348(2): 260-76, 2006 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-16492386

RESUMO

We investigated the neuropathogenic effects of feline immunodeficiency virus (FIV) envelope proteins in the context of both extracellular exposure and intracellular expression in feline neural cells. The envelope from the neurovirulent CSF-derived FIV V1 strain (V1-CSF) conferred infectivity to pseudotyped viruses in peripheral blood mononuclear cells (P < 0.01) in contrast to other cell types. Intracellular V1-CSF envelope expression in macrophages and microglia but not astrocytes resulted in the induction of host inflammatory genes contributing to neurotoxicity including IL-1beta, TNF-alpha, and indolamine 2',3'-dioxygenase (IDO) (P < 0.05) with concurrent neuronal death (P < 0.05). Upregulation of the endoplasmic reticulum stress genes was evident in brains from FIV-infected animals (P < 0.05) and in FIV-infected macrophages (P < 0.05) relative to controls. Intrastriatal implantation of an FIV envelope pseudotyped virus led to marked neuroinflammation and neuronal injury associated with neurobehavioral deficits (P < 0.01). Thus, lentivirus envelope proteins exert differential neuropathogenic effects through mechanisms that depend on the infected or exposed cell type.


Assuntos
Glicoproteínas/fisiologia , Vírus da Imunodeficiência Felina/patogenicidade , Proteínas do Envelope Viral/fisiologia , Animais , Sequência de Bases , Encéfalo/virologia , Gatos , Linhagem Celular , DNA Viral/genética , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Expressão Gênica , Genes Virais , Glicoproteínas/genética , Células HeLa , Humanos , Vírus da Imunodeficiência Felina/classificação , Vírus da Imunodeficiência Felina/genética , Vírus da Imunodeficiência Felina/fisiologia , Infecções por Lentivirus/genética , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/virologia , Leucócitos Mononucleares/virologia , Neurônios/virologia , Filogenia , Proteínas do Envelope Viral/genética , Virulência/genética
18.
J Biol Chem ; 281(1): 528-42, 2006 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-16195228

RESUMO

Retroviral Gag and Env glycoproteins (GPs) are expressed from distinct cellular areas and need to encounter to interact and assemble infectious particles. Retroviral particles may also incorporate GPs derived from other enveloped viruses via active or passive mechanisms, a process known as "pseudotyping." To further understand the mechanisms of pseudotyping, we have investigated the capacity of murine leukemia virus (MLV) or lentivirus core particles to recruit GPs derived from different virus families: the G protein of vesicular stomatitis virus (VSV-G), the hemagglutinin from an influenza virus, the E1E2 glycoproteins of hepatitis C virus (HCV-E1E2), and the retroviral Env glycoproteins of MLV and RD114 cat endogenous virus. The parameters that influenced the incorporation of viral GPs onto retroviral core particles were (i) the intrinsic cell localization properties of both viral GP and retroviral core proteins, (ii) the ability of the viral GP to interact with the retroviral core, and (iii) the expression of the lentiviral Nef protein. Whereas the hemagglutinin and VSV-G glycoproteins were recruited by MLV and lentivirus core proteins at the cell surface, the HCV and MLV GPs were most likely recruited in late endosomes. In addition, whereas these glycoproteins could be passively incorporated on either retrovirus type, the MLV GP was also actively recruited by MLV core proteins, which, through interactions with the cytoplasmic tail of the latter GP, induced its localization to late endosomal vesicles. Finally, the expression of Nef proteins specifically enhanced the incorporation of the retroviral GPs by increasing their localization in late endosomes.


Assuntos
Produtos do Gene gag/metabolismo , Produtos do Gene nef/metabolismo , Vírus da Leucemia Murina/fisiologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Células COS , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Membrana Celular/virologia , Chlorocebus aethiops , Endossomos/virologia , Produtos do Gene env/metabolismo , Hemaglutininas/metabolismo , Humanos , Rim/citologia , Lentivirus/crescimento & desenvolvimento , Lentivirus/fisiologia , Infecções por Lentivirus/metabolismo , Infecções por Lentivirus/virologia , Vírus da Leucemia Murina/crescimento & desenvolvimento , Neoplasias Hepáticas , Glicoproteínas de Membrana/metabolismo , Infecções por Retroviridae/metabolismo , Rabdomiossarcoma , Infecções Tumorais por Vírus/metabolismo , Proteínas do Envelope Viral/metabolismo
19.
Neurobiol Dis ; 20(1): 12-26, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16137563

RESUMO

Complement activation in the brain contributes to the pathology of neuroinflammatory and neurodegenerative diseases such as neuro-AIDS. Using semiquantitative in situ hybridization and immunohistochemistry, we observed an early and sustained increase in the expression of C1q, the initial recognition subcomponent of the classical complement cascade, in the CNS during simian immunodeficiency virus (SIV) infection of rhesus macaques. Cells of the microglial/macrophage lineage were the sources for C1q protein and transcripts. C1q expression was observed in proliferating and infiltrating cells in SIV-encephalitic brains. All SIV-positive cells were also C1q-positive. Treatment with the CNS-permeant antiretroviral agent 6-chloro-2',3'-dideoxyguanosine decreased C1q synthesis along with SIV burden and focal inflammatory reactions in the brains of AIDS-symptomatic monkeys. Thus, activation of the classical complement arm of innate immunity is an early event in neuro-AIDS and a possible target for intervention.


Assuntos
Complexo AIDS Demência/tratamento farmacológico , Complemento C1q/metabolismo , Didesoxinucleosídeos/uso terapêutico , Infecções por Lentivirus/tratamento farmacológico , Macrófagos/imunologia , Microglia/imunologia , Complexo AIDS Demência/imunologia , Complexo AIDS Demência/metabolismo , Animais , Antirretrovirais/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/virologia , Modelos Animais de Doenças , Encefalite/tratamento farmacológico , Encefalite/imunologia , Encefalite/metabolismo , Infecções por Lentivirus/imunologia , Infecções por Lentivirus/metabolismo , Macaca mulatta , Macrófagos/metabolismo , Macrófagos/virologia , Microglia/metabolismo , Microglia/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia , Carga Viral
20.
J Gen Virol ; 86(Pt 8): 2239-2248, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16033971

RESUMO

Infection of cats with Feline immunodeficiency virus (FIV) is an important model for understanding comparative lentivirus biology. In vivo, FIV infects lymphocytes and monocyte/macrophages, but in vitro infection is commonly investigated in epithelial Crandell-Reese Feline Kidney (CRFK) cells. In this study, the transcriptional responses of CRFK cells and primary lymphocytes to infection with FIV 34TF, a cloned subtype A virus, and FIV USgaB01, a biological subtype B isolate, were determined. Reverse-transcribed mRNA from both cell types was hybridized to microarrays containing 1700 human expressed sequence tags in duplicate and data were analysed with Significance Analysis of Microarrays (sam) software. Results from six experiments assessing homeostatic cross-species hybridization excluded 3.48 % inconsistently detected transcripts. Analysis of data from five time points over 48 h after infection identified 132 and 24 differentially expressed genes in epithelial cells and lymphocytes, respectively. Genes involved in protein synthesis, the cell cycle, structure and metabolism were affected. The magnitude of gene-expression changes ranged from 0.62 to 1.62 and early gene induction was followed by downregulation after 4 h. Transcriptional changes in CRFK cells were distinct from those in lymphocytes, except for heat-shock cognate protein 71, which was induced at multiple time points in both cell types. These findings indicate that FIV infection induces transcriptional changes of a modest magnitude in a wide range of genes, which is probably reflective of the relatively non-cytopathic nature of virus infection.


Assuntos
Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Vírus da Imunodeficiência Felina , Infecções por Lentivirus/genética , Linfócitos/metabolismo , Animais , Gatos , Células Cultivadas , Proteínas de Choque Térmico HSC70 , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Infecções por Lentivirus/metabolismo , Análise em Microsséries , Dados de Sequência Molecular , Especificidade de Órgãos , Especificidade da Espécie , Fatores de Tempo , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA