Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
J Biol Chem ; 295(13): 4252-4264, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32075911

RESUMO

SAM and HD domain-containing protein 1 (SAMHD1) is a host factor that restricts reverse transcription of lentiviruses such as HIV in myeloid cells and resting T cells through its dNTP triphosphohydrolase (dNTPase) activity. Lentiviruses counteract this restriction by expressing the accessory protein Vpx or Vpr, which targets SAMHD1 for proteasomal degradation. SAMHD1 is conserved among mammals, and the feline and bovine SAMHD1 proteins (fSAM and bSAM) restrict lentiviruses by reducing cellular dNTP concentrations. However, the functional regions of fSAM and bSAM that are required for their biological functions are not well-characterized. Here, to establish alternative models to investigate SAMHD1 in vivo, we studied the restriction profile of fSAM and bSAM against different primate lentiviruses. We found that both fSAM and bSAM strongly restrict primate lentiviruses and that Vpx induces the proteasomal degradation of both fSAM and bSAM. Further investigation identified one and five amino acid sites in the C-terminal domain (CTD) of fSAM and bSAM, respectively, that are required for Vpx-mediated degradation. We also found that the CTD of bSAM is directly involved in mediating bSAM's antiviral activity by regulating dNTPase activity, whereas the CTD of fSAM is not. Our results suggest that the CTDs of fSAM and bSAM have important roles in their antiviral functions. These findings advance our understanding of the mechanism of fSAM- and bSAM-mediated viral restriction and might inform strategies for improving HIV animal models.


Assuntos
HIV/genética , Lentivirus/genética , Transcrição Reversa/genética , Proteína 1 com Domínio SAM e Domínio HD/genética , Animais , Gatos , Bovinos , Células HEK293 , HIV/patogenicidade , Interações Hospedeiro-Patógeno/genética , Humanos , Lentivirus/patogenicidade , Células Mieloides/virologia , Domínios Proteicos/genética , Proteína 1 com Domínio SAM e Domínio HD/química , Linfócitos T/virologia , Replicação Viral/genética
2.
Trends Biochem Sci ; 40(2): 108-16, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25555456

RESUMO

To achieve productive infection, retroviruses such as HIV stably integrate their reverse transcribed RNA genome into a host chromosome. Each retroviral family preferentially integrates near a unique subset of genomic features. HIV integrase (IN) is targeted to the body of active transcription units through interaction with lens epithelium-derived growth factor (LEDGF/p75). We describe the successful effort to develop inhibitors of the interaction between IN and LEDGF/p75, referred to as LEDGINs. Gammaretroviruses display a distinct integration pattern. Recently, BET (bromo- and extraterminal domain) proteins were identified as the LEDGF/p75 counterparts that target the integration of gammaretroviruses. The identification of the chromatin-readers LEDGF/p75 and BET as cellular cofactors that orchestrate lentiviral or gammaretroviral integration opens new avenues to developing safer viral vectors for gene therapy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por HIV/tratamento farmacológico , Inibidores de Integrase de HIV/administração & dosagem , Fatores de Transcrição/metabolismo , Integração Viral/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Cromatina/efeitos dos fármacos , Gammaretrovirus/efeitos dos fármacos , Gammaretrovirus/genética , Gammaretrovirus/patogenicidade , Infecções por HIV/virologia , Integrase de HIV/metabolismo , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Humanos , Lentivirus/efeitos dos fármacos , Lentivirus/genética , Lentivirus/patogenicidade , Fatores de Transcrição/genética , Integração Viral/efeitos dos fármacos
3.
J Virol ; 90(15): 6724-6737, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27170760

RESUMO

UNLABELLED: Nonhuman primates (NHPs) are a historically important source of zoonotic viruses and are a gold-standard model for research on many human pathogens. However, with the exception of simian immunodeficiency virus (SIV) (family Retroviridae), the blood-borne viruses harbored by these animals in the wild remain incompletely characterized. Here, we report the discovery and characterization of two novel simian pegiviruses (family Flaviviridae) and two novel simian arteriviruses (family Arteriviridae) in wild African green monkeys from Zambia (malbroucks [Chlorocebus cynosuros]) and South Africa (vervet monkeys [Chlorocebus pygerythrus]). We examine several aspects of infection, including viral load, genetic diversity, evolution, and geographic distribution, as well as host factors such as age, sex, and plasma cytokines. In combination with previous efforts to characterize blood-borne RNA viruses in wild primates across sub-Saharan Africa, these discoveries demonstrate that in addition to SIV, simian pegiviruses and simian arteriviruses are widespread and prevalent among many African cercopithecoid (i.e., Old World) monkeys. IMPORTANCE: Primates are an important source of viruses that infect humans and serve as an important laboratory model of human virus infection. Here, we discover two new viruses in African green monkeys from Zambia and South Africa. In combination with previous virus discovery efforts, this finding suggests that these virus types are widespread among African monkeys. Our analysis suggests that one of these virus types, the simian arteriviruses, may have the potential to jump between different primate species and cause disease. In contrast, the other virus type, the pegiviruses, are thought to reduce the disease caused by human immunodeficiency virus (HIV) in humans. However, we did not observe a similar protective effect in SIV-infected African monkeys coinfected with pegiviruses, possibly because SIV causes little to no disease in these hosts.


Assuntos
Infecções por Arterivirus/epidemiologia , Evolução Biológica , Infecções por Flaviviridae/epidemiologia , Variação Genética , Infecções por Lentivirus/epidemiologia , Carga Viral , África/epidemiologia , Animais , Animais Selvagens , Arterivirus/genética , Arterivirus/patogenicidade , Infecções por Arterivirus/genética , Infecções por Arterivirus/virologia , Flaviviridae/genética , Flaviviridae/patogenicidade , Infecções por Flaviviridae/genética , Infecções por Flaviviridae/virologia , Genoma Viral , Haplorrinos , Humanos , Lentivirus/genética , Lentivirus/patogenicidade , Infecções por Lentivirus/genética , Infecções por Lentivirus/virologia , Filogenia , Prevalência
4.
J Virol ; 89(10): 5204-12, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25762730

RESUMO

Pathogens such as HIV-1, with their minimalist genomes, must navigate cellular networks and rely on hijacking and manipulating the host machinery for successful replication. Limited overlap of host factors identified as vital for pathogen replication may be explained by considering that pathogens target, rather than specific cellular factors, crucial cellular pathways by targeting different, functionally equivalent, protein-protein interactions within that pathway. The ability to utilize alternative routes through cellular pathways may be essential for pathogen survival when restricted and provide flexibility depending on the viral replication stage and the environment in the infected host. In this minireview, we evaluate evidence supporting this notion, discuss specific HIV-1 examples, and consider the molecular mechanisms which allow pathogens to flexibly exploit different routes.


Assuntos
Infecções por HIV/virologia , HIV-1/patogenicidade , Transporte Ativo do Núcleo Celular , Animais , Citidina Desaminase/fisiologia , HIV-1/genética , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Lentivirus/patogenicidade , Lentivirus/fisiologia , Infecções por Lentivirus/virologia , Modelos Biológicos , Integração Viral , Replicação Viral , Produtos do Gene vif do Vírus da Imunodeficiência Humana/fisiologia
5.
Mol Ther ; 22(4): 774-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24441399

RESUMO

Self-inactivating (SIN) lentiviral vectors (LV) have an excellent therapeutic potential as demonstrated in preclinical studies and clinical trials. However, weaker mechanisms of insertional mutagenesis could still pose a significant risk in clinical applications. Taking advantage of novel in vivo genotoxicity assays, we tested a battery of LV constructs, including some with clinically relevant designs, and found that oncogene activation by promoter insertion is the most powerful mechanism of early vector-induced oncogenesis. SIN LVs disabled in their capacity to activate oncogenes by promoter insertion were less genotoxic and induced tumors by enhancer-mediated activation of oncogenes with efficiency that was proportional to the strength of the promoter used. On the other hand, when enhancer activity was reduced by using moderate promoters, oncogenesis by inactivation of tumor suppressor gene was revealed. This mechanism becomes predominant when the enhancer activity of the internal promoter is shielded by the presence of a synthetic chromatin insulator cassette. Our data provide both mechanistic insights and quantitative readouts of vector-mediated genotoxicity, allowing a relative ranking of different vectors according to these features, and inform current and future choices of vector design with increasing biosafety.


Assuntos
Carcinogênese/genética , Terapia Genética , Vetores Genéticos/efeitos adversos , Lentivirus/genética , Vetores Genéticos/uso terapêutico , Humanos , Lentivirus/patogenicidade , Mutagênese Insercional/genética , Regiões Promotoras Genéticas
6.
Uirusu ; 65(2): 173-178, 2015.
Artigo em Japonês | MEDLINE | ID: mdl-27760915

RESUMO

Membrane-associated RING-CH 8 (MARCH8) is one of 11 members of the recently discovered MARCH family of RING-finger E3 ubiquitin ligases. MARCH8 downregulates several host transmembrane proteins; however, its physiological roles remain unknown. Here we identify MARCH8 as a novel antiviral factor. The overexpression of MARCH8 in virus producing cells did not affect levels of lentivirus production, but markedly reduced viral infectivity. MARCH8 blocked the incorporation of HIV-1 envelope glycoprotein into virions by downregulating it from the cell surface, probably through their interaction, resulting in reduced viral entry efficiency. The inhibitory effect of MARCH8 on vesicular stomatitis virus G-glycoprotein was even more remarkable, suggesting a broad-spectrum inhibition of enveloped viruses by MARCH8. Importantly, the endogenous expression of MARCH8 was high in monocyte-derived macrophages and dendritic cells, and MARCH8 depletion in macrophages significantly increased the infectivity of virions produced from these cells. Our findings thus indicate that MARCH8, which is highly expressed in terminally differentiated myeloid cells, is a potent antiviral host transmembrane protein that reduces virion incorporation of viral envelope glycoproteins.


Assuntos
Antivirais , HIV-1/patogenicidade , Lentivirus/patogenicidade , Ubiquitina-Proteína Ligases/fisiologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Glicoproteínas/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/virologia , Glicoproteínas de Membrana/metabolismo , Células Mieloides/metabolismo , Células Mieloides/virologia , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo
7.
J Virol ; 85(24): 12950-61, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21976643

RESUMO

Lentiviruses, unlike the gammaretroviruses, are able to infect nondividing cells by transiting through nuclear pores to access the host genomic DNA. Several nuclear import and nuclear pore components have been implicated as playing a role in nuclear import, including transportin 3 (TNPO3), a member of the importin-ß family of nuclear import proteins. We demonstrated that TNPO3 was required by several lentiviruses, with simian immunodeficiency virus mac239 (SIVmac239) and equine infectious anemia virus (EIAV) the most dependent and human immunodeficiency virus type 1 (HIV-1) and feline immunodeficiency virus (FIV) the least. Analysis of HIV-1/SIVmac239 chimeric viruses showed that dependence on TNPO3 mapped to the SIVmac239 capsid. Mutation of a single amino acid, A76V in the SIVmac239 capsid, rendered the virus TNPO3 independent and resistant to mCPSF6-358, a truncated splicing factor that prevents HIV-1 nuclear import. Using a complementation assay based on 293T cells that express a TNPO3-targeted short hairpin RNA (shRNA), we showed that the Drosophila TNPO3 homologue can substitute for its human counterpart and that it mapped a key functional domain of TNPO3 to the carboxy-terminal cargo-binding domain. Within the cargo-binding domain, two hydrophobic motifs were required for TNPO3-dependent infection. The mutated TNPO3 proteins maintained their ability to localize to the nucleus, suggesting that their inability to restore lentivirus infection resulted from an inability to bind to a host or viral cargo protein.


Assuntos
Transporte Ativo do Núcleo Celular , Interações Hospedeiro-Patógeno , Lentivirus/patogenicidade , beta Carioferinas/metabolismo , Animais , Linhagem Celular , Drosophila , Teste de Complementação Genética , Humanos , Ligação Proteica , Estrutura Terciária de Proteína
8.
J Virol ; 84(16): 8193-201, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519393

RESUMO

All lentiviruses except equine infectious anemia virus (EIAV) use the small accessory protein Vif to counteract the restriction activity of the relevant APOBEC3 (A3) proteins of their host species. Prior studies have suggested that the Vif-A3 interaction is species specific. Here, using the APOBEC3H (Z3)-type proteins from five distinct mammals, we report that this is generally not the case: some lentiviral Vif proteins are capable of triggering the degradation of both the A3Z3-type protein of their normal host species and those of several other mammals. For instance, SIV(mac) Vif can mediate the degradation of the human, macaque, and cow A3Z3-type proteins but not of the sheep or cat A3Z3-type proteins. Maedi-visna virus (MVV) Vif is similarly promiscuous, degrading not only sheep A3Z3 but also the A3Z3-type proteins of humans, macaques, cows, and cats. In contrast to the neutralization capacity of these Vif proteins, human immunodeficiency virus (HIV), bovine immunodeficiency virus (BIV), and feline immunodeficiency virus (FIV) Vif appear specific to the A3Z3-type protein of their hosts. We conclude, first, that the Vif-A3Z3 interaction can be promiscuous and, second, despite this tendency, that each lentiviral Vif protein is optimized to degrade the A3Z3 protein of its mammalian host. Our results thereby suggest that the Vif-A3Z3 interaction is relevant to lentivirus biology.


Assuntos
Citosina Desaminase/antagonistas & inibidores , Produtos do Gene vif/metabolismo , Lentivirus/patogenicidade , Fatores de Virulência/metabolismo , Animais , Gatos , Bovinos , Humanos , Macaca , Ovinos
9.
J Virol ; 84(1): 397-406, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19846519

RESUMO

Recent genome-wide screens have highlighted an important role for transportin 3 in human immunodeficiency virus type 1 (HIV-1) infection and preintegration complex (PIC) nuclear import. Moreover, HIV-1 integrase interacted with recombinant transportin 3 protein under conditions whereby Moloney murine leukemia virus (MLV) integrase failed to do so, suggesting that integrase-transportin 3 interactions might underscore active retroviral PIC nuclear import. Here we correlate infectivity defects in transportin 3 knockdown cells with in vitro protein binding affinities for an expanded set of retroviruses that include simian immunodeficiency virus (SIV), bovine immunodeficiency virus (BIV), equine infectious anemia virus (EIAV), feline immunodeficiency virus (FIV), and Rous sarcoma virus (RSV) to critically address the role of integrase-transportin 3 interactions in viral infection. Lentiviruses, with the exception of FIV, display a requirement for transportin 3 in comparison to MLV and RSV, yielding an infection-based dependency ranking of SIV > HIV-1 > BIV and EIAV > MLV, RSV, and FIV. In vitro pulldown and surface plasmon resonance assays, in contrast, define a notably different integrase-transportin 3 binding hierarchy: FIV, HIV-1, and BIV > SIV and MLV > EIAV. Our results therefore fail to support a critical role for integrase binding in dictating transportin 3 dependency during retrovirus infection. In addition to integrase, capsid has been highlighted as a retroviral nuclear import determinant. Accordingly, MLV/HIV-1 chimera viruses pinpoint the genetic determinant of sensitization to transportin 3 knockdown to the HIV-1 capsid protein. We therefore conclude that capsid, not integrase, is the dominant viral factor that dictates transportin 3 dependency during HIV-1 infection.


Assuntos
Capsídeo/fisiologia , Infecções por HIV/etiologia , HIV-1/patogenicidade , Integrases/fisiologia , Carioferinas/fisiologia , beta Carioferinas/fisiologia , Animais , Capsídeo/metabolismo , Linhagem Celular , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/fisiologia , Humanos , Integrases/metabolismo , Carioferinas/deficiência , Carioferinas/genética , Carioferinas/metabolismo , Lentivirus/patogenicidade , Vírus da Leucemia Murina , Ligação Proteica , beta Carioferinas/genética , beta Carioferinas/metabolismo
10.
J Immunother Cancer ; 9(9)2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34518288

RESUMO

BACKGROUND: Chimeric antigen receptor (CAR) T cells have shown considerable promise as a personalized cellular immunotherapy against B cell malignancies. However, the complex and lengthy manufacturing processes involved in generating CAR T cell products ex vivo result in substantial production time delays and high costs. Furthermore, ex vivo expansion of T cells promotes cell differentiation that reduces their in vivo replicative capacity and longevity. METHODS: Here, to overcome these limitations, CAR-T cells are engineered directly in vivo by administering a lentivirus expressing a mutant Sindbis envelope, coupled with a bispecific antibody binder that redirects the virus to CD3+ human T cells. RESULTS: This redirected lentiviral system offers exceptional specificity and efficiency; a single dose of the virus delivered to immunodeficient mice engrafted with human peripheral blood mononuclear cells generates CD19-specific CAR-T cells that markedly control the growth of an aggressive pre-established xenograft B cell tumor. CONCLUSIONS: These findings underscore in vivo engineering of CAR-T cells as a promising approach for personalized cancer immunotherapy.


Assuntos
Anticorpos Biespecíficos/metabolismo , Lentivirus/patogenicidade , Receptores de Antígenos Quiméricos/metabolismo , Engenharia Tecidual/métodos , Animais , Modelos Animais de Doenças , Humanos , Camundongos
11.
Sci Rep ; 11(1): 16201, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34376765

RESUMO

Optical spectroscopic techniques have been commonly used to detect the presence of biofilm-forming pathogens (bacteria and fungi) in the agro-food industry. Recently, near-infrared (NIR) spectroscopy revealed that it is also possible to detect the presence of viruses in animal and vegetal tissues. Here we report a platform based on visible and NIR (VNIR) hyperspectral imaging for non-contact, reagent free detection and quantification of laboratory-engineered viral particles in fluid samples (liquid droplets and dry residue) using both partial least square-discriminant analysis and artificial feed-forward neural networks. The detection was successfully achieved in preparations of phosphate buffered solution and artificial saliva, with an equivalent pixel volume of 4 nL and lowest concentration of 800 TU·[Formula: see text]L-1. This method constitutes an innovative approach that could be potentially used at point of care for rapid mass screening of viral infectious diseases and monitoring of the SARS-CoV-2 pandemic.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Infecções por Lentivirus/diagnóstico , Técnicas de Diagnóstico Molecular/métodos , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Células HEK293 , Humanos , Processamento de Imagem Assistida por Computador/normas , Lentivirus/isolamento & purificação , Lentivirus/patogenicidade , Infecções por Lentivirus/virologia , Técnicas de Diagnóstico Molecular/normas , Sistemas Automatizados de Assistência Junto ao Leito , Saliva/virologia , Sensibilidade e Especificidade , Espectroscopia de Luz Próxima ao Infravermelho/normas
12.
Bioengineered ; 12(2): 12498-12508, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34927536

RESUMO

At present, the mechanism of siSCN9A in Vincristine (VCR)-induced neuropathic pain (NP) is still unclear. This study aimed to explore the analgesic mechanism of lentivirus-siSCN9A (LV-siSCN9A) infected neurons against NP. 40 male Sprague-Dawley (SD) rats were divided into a control group (injected with normal saline), a model group (VCR-induced NP model), a LV-SC group (NP model mice were injected with LV-SC-infected dorsal root ganglia (DRG) neuron cells under the microscope), and a LV-siSCN9A group (NP model mice were injected with LV-siSCN9A-infected DRG neuron cells under the microscope, with 10 rats in each group. The changes of mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) of rats in different groups were detected by behavior testing, the Nav1.7 changes in each group were detected by immunofluorescence double standard and Western-blot method. It was found that compared with the control group, the MWT and TWL of the rats in model group were significantly decreased (P < 0.05), and the expression levels of Nav1.7 messenger ribonucleic acid (mRNA) and proteins were significantly increased (P < 0.05). Compared with LV-SC group, the MWT and TWL of rats in LV-siSCN9A group were significantly increased (P < 0.05), the expression levels of Nav1.7 mRNA and proteins were significantly decreased (P < 0.05), and the CGRP expression of spinal dorsal horn was significantly decreased. It was concluded that the LV-siSCN9A infected neurons could play an analgesic role by down-regulating Nav1.7 expression induced by VCR in NP model.


Assuntos
Infecções por Lentivirus/virologia , Lentivirus/patogenicidade , Neuralgia/induzido quimicamente , Neuralgia/virologia , Neurônios/efeitos dos fármacos , Neurônios/virologia , Vincristina/farmacologia , Analgesia/métodos , Animais , Modelos Animais de Doenças , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/virologia , Infecções por Lentivirus/genética , Masculino , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Neuralgia/genética , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
13.
Elife ; 92020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33226340

RESUMO

Systemic oxygen restriction (SOR) is prevalent in numerous clinical conditions, including chronic obstructive pulmonary disease (COPD), and is associated with increased susceptibility to viral infections. However, the influence of SOR on T cell immunity remains uncharacterized. Here we show the detrimental effect of hypoxia on mitochondrial-biogenesis in activated mouse CD8+ T cells. We find that low oxygen level diminishes CD8+ T cell anti-viral response in vivo. We reveal that respiratory restriction inhibits ATP-dependent matrix processes that are critical for mitochondrial-biogenesis. This respiratory restriction-mediated effect could be rescued by TCA cycle re-stimulation, which yielded increased mitochondrial matrix-localized ATP via substrate-level phosphorylation. Finally, we demonstrate that the hypoxia-arrested CD8+ T cell anti-viral response could be rescued in vivo through brief exposure to atmospheric oxygen pressure. Overall, these findings elucidate the detrimental effect of hypoxia on mitochondrial-biogenesis in activated CD8+ T cells, and suggest a new approach for reducing viral infections in COPD.


Assuntos
Trifosfato de Adenosina/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Hipóxia/metabolismo , Ativação Linfocitária , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Biogênese de Organelas , Fosforilação Oxidativa , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Hipóxia/imunologia , Lentivirus/patogenicidade , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/imunologia , Transdução de Sinais
14.
Top HIV Med ; 17(2): 30-4, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19401604

RESUMO

The 16th Conference on Retroviruses and Opportunistic Infections featured a strong and balanced program that showcased exciting research into cellular restrictions that defend the cell against viral infection as well as cellular cofactors that regulate central steps in viral replication. Immunopathogenesis presentations continued to reveal some surprises such as evidence for pathogenicity in natural simian immunodeficiency virus infection. The identification of novel cellular restrictions and cellular cofactors of viral replication indicate the possibility of numerous opportunities for the development of novel therapeutic agents for the treatment of HIV and AIDS.


Assuntos
Infecções por HIV/fisiopatologia , Infecções por Retroviridae/fisiopatologia , Replicação Viral/fisiologia , Desaminase APOBEC-3G , Animais , Citidina Desaminase , Produtos do Gene nef , Infecções por HIV/imunologia , Humanos , Lentivirus/patogenicidade , Primatas , Infecções por Retroviridae/imunologia , Transcrição Reversa , Replicação Viral/genética
15.
J Vis Exp ; (145)2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30985756

RESUMO

The use of hiPSC-derived cells represents a valuable approach to study human neurodegenerative diseases. Here, we describe an optimized protocol for the differentiation of hiPSCs derived from a patient with the triplication of the alpha-synuclein gene (SNCA) locus into Parkinson's disease (PD)-relevant dopaminergic neuronal populations. Accumulating evidence has shown that high levels of SNCA are causative for the development of PD. Recognizing the unmet need to establish novel therapeutic approaches for PD, especially those targeting the regulation of SNCA expression, we recently developed a CRISPR/dCas9-DNA-methylation-based system to epigenetically modulate SNCA transcription by enriching methylation levels at the SNCA intron 1 regulatory region. To deliver the system, consisting of a dead (deactivated) version of Cas9 (dCas9) fused with the catalytic domain of the DNA methyltransferase enzyme 3A (DNMT3A), a lentiviral vector is used. This system is applied to cells with the triplication of the SNCA locus and reduces the SNCA-mRNA and protein levels by about 30% through the targeted DNA methylation of SNCA intron 1. The fine-tuned downregulation of the SNCA levels rescues disease-related cellular phenotypes. In the current protocol, we aim to describe a step-by-step procedure for differentiating hiPSCs into neural progenitor cells (NPCs) and the establishment and validation of pyrosequencing assays for the evaluation of the methylation profile in the SNCA intron 1. To outline in more detail the lentivirus-CRISPR/dCas9 system used in these experiments, this protocol describes how to produce, purify, and concentrate lentiviral vectors and to highlight their suitability for epigenome- and genome-editing applications using hiPSCs and NPCs. The protocol is easily adaptable and can be used to produce high titer lentiviruses for in vitro and in vivo applications.


Assuntos
Epigenoma/genética , Epigenômica/métodos , Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lentivirus/patogenicidade , Diferenciação Celular , Humanos
16.
Res Vet Sci ; 118: 107-114, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29421479

RESUMO

Small Ruminant Lentivirus (SRLV) subtype E1, also known as Roccaverano strain, is considered a low pathogenic virus on the basis of natural genetic deletions, in vitro properties and on-farm observations. In order to gain more knowledge on this atypical lentivirus we investigated the in vivo tropism of Roccaverano strain in both, experimentally and naturally infected goats. Antibody responses were monitored as well as tissue distribution and viral load, evaluated by real time PCR on single spliced (gag/env) and multiple spliced (rev) RNA targets respectively, that were compared to histopathological lesions. Lymph nodes, spleen, alveolar macrophages and mammary gland turned out to be the main tissue reservoirs of genotype E1-provirus. Moreover, mammary gland and/or mammary lymph nodes acted as active replication sites in dairy goats, supporting the lactogenic transmission of this virus. Notably, a direct association between viral load and concomitant infection or inflammatory processes was evident within organs such as spleen, lung and testis. Our results validate the low pathogenicity designation of SRLV genotype E1 in vivo, and confirm the monocyte-macrophage cell lineage as the main virus reservoir of this genotype. Accordingly, SRLV genotype E displays a tropism towards all tissues characterized by an abundant presence of these cells, either for their own anatomical structure or for an occasional infectious/inflammatory status.


Assuntos
Doenças das Cabras/patologia , Doenças das Cabras/virologia , Infecções por Lentivirus/veterinária , Animais , Genótipo , Cabras , Lentivirus/isolamento & purificação , Lentivirus/patogenicidade , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Ruminantes , Ovinos , Doenças dos Ovinos , Distribuição Tecidual , Carga Viral/veterinária
17.
PLoS One ; 13(9): e0204134, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30212571

RESUMO

Small ruminant lentivirus (SRLV) infection manifests itself mainly with chronic progressive arthritis affecting mainly carpal joints. The data from serological and questionnaire surveys were retrospectively analyzed to determine how the dissemination of SRLV infection in the herd influenced farmer's subjective opinion on the occurrence of swelling of carpal joints (considered as a proxy of arthritis). Between 1996 and 2017 153 different Polish dairy goat herds counting at least 20 adult goats were serologically screened for CAE and their owners were asked about their opinion on the occurrence of arthritis (never, rarely, often). Of them 73 SRLV-seropositive herds, in which true seroprevalence had been estimated, were included in the analysis. The ordinal logistic regression model was developed to determine the relationship between the true within-herd seroprevalence and the probability that the farmer would observe arthritis in the herd never, rarely or often. True within-herd seroprevalence ranged from 0.2% to 100% with the median of 34.6%. Farmers declared not to have observed arthritis in 40 (54.8%) herds, to have seen it rarely in 9 (12.3%) of herds, and to have observed it often in 24 (32.9%) of herds. The model proved that the probability of observing goats with carpal arthritis in the herd was significantly linked to the true within-herd seroprevalence (OR = 1.058, CI 95% from 1.037 to 1.078; p<0.001), but this relationship was not linear and SRLV infection proved to remain unapparent to farmers even when a considerable part of the herd had already become infected. Concluding, the study shows that when the farmer realizes that goats in the herd suffer from arthritis, SRLV infection is almost certainly already widespread in the herd.


Assuntos
Anticorpos Antivirais/sangue , Artrite/veterinária , Articulações do Carpo/virologia , Fazendeiros/psicologia , Doenças das Cabras/epidemiologia , Cabras/virologia , Infecções por Lentivirus/veterinária , Criação de Animais Domésticos , Animais , Artrite/epidemiologia , Artrite/patologia , Artrite/virologia , Articulações do Carpo/imunologia , Articulações do Carpo/patologia , Feminino , Doenças das Cabras/imunologia , Doenças das Cabras/patologia , Doenças das Cabras/virologia , Lentivirus/patogenicidade , Lentivirus/fisiologia , Infecções por Lentivirus/epidemiologia , Infecções por Lentivirus/patologia , Infecções por Lentivirus/virologia , Modelos Logísticos , Masculino , Polônia/epidemiologia , Estudos Soroepidemiológicos , Inquéritos e Questionários
18.
Trends Biotechnol ; 25(8): 333-7, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17574286

RESUMO

AIDS is among the most devastating diseases of our time, claiming the lives of approximately 3 million people per year. The primary cause of AIDS, human immunodeficiency virus type 1 (HIV-1), is a pathogen that is highly specific for humans and generally does not infect or cause disease in other species. This property complicates the generation of animal models that are urgently needed to test new antiretroviral therapies and vaccines. The most practical animal models developed to date consist of infection of rhesus macaques with a simian immunodeficiency virus (SIV) or chimeric HIV/SIV viruses. Although these models are useful for particular applications, the fact that SIV is a distinct virus compared with HIV-1 represents a significant limitation to their use. Here, we discuss the uses and limitations of existing models and recent advances that might lead to better animal models for HIV/AIDS.


Assuntos
Modelos Animais de Doenças , Infecções por HIV/classificação , Infecções por HIV/virologia , Lentivirus/genética , Lentivirus/patogenicidade , Animais , Humanos
19.
J Clin Virol ; 39(1): 27-33, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17409017

RESUMO

BACKGROUND: Highly pathogenic avian influenza (HPAI) H5N1 has spread globally in birds and infected over 270 humans with an apparently high mortality rate. Serologic studies to determine the extent of asymptomatic H5N1 infection in humans and other mammals and to investigate the immunogenicity of current H5N1 vaccine candidates have been hampered by the biosafety requirements needed for H5N1 micro-neutralization tests. OBJECTIVE: Development of a serodiagnostic tool for highly pathogenic influenza that reproduces H5N1 biology but can be used with less biohazard. STUDY DESIGN: We have generated and evaluated H5 hemagglutinin pseudotyped lentiviral particles encoding the luciferase reporter (H5pp). RESULTS: H5pp entry into target cells depends on alpha2-3 cell surface sialic acids and requires low pH for membrane fusion. H5pp infectivity is specifically neutralized by sera from patients and animals infected with H5N1 and correlates well with conventional microneutralization test. CONCLUSIONS: H5pp reproduce H5N1 influenza virus entry into target cells and potentially provides a high-throughput and safe method for sero-epidemiology.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Aviária/diagnóstico , Influenza Aviária/virologia , Influenza Humana/diagnóstico , Influenza Humana/virologia , Lentivirus/fisiologia , Vírion/fisiologia , Animais , Aves , Cães , Glicoproteínas de Hemaglutininação de Vírus da Influenza/biossíntese , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Humanos , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/sangue , Influenza Humana/sangue , Lentivirus/genética , Lentivirus/patogenicidade , Luciferases/genética , Testes de Neutralização/métodos , Testes Sorológicos/métodos , Vírion/genética , Vírion/patogenicidade
20.
Comp Med ; 57(1): 33-43, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17348289

RESUMO

Human immunodeficiency virus (HIV), the causative agent for acquired immune deficiency syndrome, was described over 25 y ago. Since that time, much progress has been made in characterizing the pathogenesis, etiology, transmission, and disease syndromes resulting from this devastating pathogen. However, despite decades of study by many investigators, basic questions about HIV biology still remain, and an effective prophylactic vaccine has not been developed. This review provides an overview of the viruses related to HIV that have been used in experimental animal models to improve our knowledge of lentiviral disease. Viruses discussed are grouped as causing (1) nonlentiviral immunodeficiency-inducing diseases, (2) naturally occurring pathogenic infections, (3) experimentally induced lentiviral infections, and (4) nonpathogenic lentiviral infections. Each of these model types has provided unique contributions to our understanding of HIV disease; further, a comparative overview of these models both reinforces the unique attributes of each agent and provides a basis for describing elements of lentiviral disease that are similar across mammalian species.


Assuntos
Síndrome da Imunodeficiência Adquirida , Modelos Animais de Doenças , Infecções por Lentivirus/virologia , Lentivirus/classificação , Lentivirus/patogenicidade , Animais , Filogenia , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA