Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
PLoS Pathog ; 17(1): e1009292, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33507952

RESUMO

The human airway epithelium is the initial site of SARS-CoV-2 infection. We used flow cytometry and single cell RNA-sequencing to understand how the heterogeneity of this diverse cell population contributes to elements of viral tropism and pathogenesis, antiviral immunity, and treatment response to remdesivir. We found that, while a variety of epithelial cell types are susceptible to infection, ciliated cells are the predominant cell target of SARS-CoV-2. The host protease TMPRSS2 was required for infection of these cells. Importantly, remdesivir treatment effectively inhibited viral replication across cell types, and blunted hyperinflammatory responses. Induction of interferon responses within infected cells was rare and there was significant heterogeneity in the antiviral gene signatures, varying with the burden of infection in each cell. We also found that heavily infected secretory cells expressed abundant IL-6, a potential mediator of COVID-19 pathogenesis.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Antivirais/farmacologia , COVID-19/imunologia , COVID-19/virologia , SARS-CoV-2/fisiologia , Tropismo Viral , Monofosfato de Adenosina/farmacologia , Alanina/farmacologia , COVID-19/genética , Epitélio/imunologia , Epitélio/virologia , Humanos , Interferons/genética , Interferons/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Pulmão/imunologia , Pulmão/virologia , SARS-CoV-2/efeitos dos fármacos , Tropismo Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
2.
Viruses ; 12(10)2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-33003350

RESUMO

The Gammacoronavirus infectious bronchitis virus (IBV) causes a highly contagious and economically important respiratory disease in poultry. In the laboratory, most IBV strains are restricted to replication in ex vivo organ cultures or in ovo and do not replicate in cell culture, making the study of their basic virology difficult. Entry of IBV into cells is facilitated by the large glycoprotein on the surface of the virion, the spike (S) protein, comprised of S1 and S2 subunits. Previous research showed that the S2' cleavage site is responsible for the extended tropism of the IBV Beaudette strain. This study aims to investigate whether protease treatment can extend the tropism of other IBV strains. Here we demonstrate that the addition of exogenous trypsin during IBV propagation in cell culture results in significantly increased viral titres. Using a panel of IBV strains, exhibiting varied tropisms, the effects of spike cleavage on entry and replication were assessed by serial passage cell culture in the presence of trypsin. Replication could be maintained over serial passages, indicating that the addition of exogenous protease is sufficient to overcome the barrier to infection. Mutations were identified in both S1 and S2 subunits following serial passage in cell culture. This work provides a proof of concept that exogenous proteases can remove the barrier to IBV replication in otherwise non-permissive cells, providing a platform for further study of elusive field strains and enabling sustainable vaccine production in vitro.


Assuntos
Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/virologia , Vírus da Bronquite Infecciosa/efeitos dos fármacos , Vírus da Bronquite Infecciosa/fisiologia , Tripsina/uso terapêutico , Tropismo Viral/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Gammacoronavirus/efeitos dos fármacos , Vírus da Bronquite Infecciosa/metabolismo , Cinética , Inoculações Seriadas , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Proteínas do Envelope Viral/metabolismo , Vírion/efeitos dos fármacos , Vírion/metabolismo , Replicação Viral/efeitos dos fármacos
3.
Braz. j. infect. dis ; 24(2): 150-159, Mar.-Apr. 2020. tab, graf
Artigo em Inglês | LILACS, Coleciona SUS (Brasil) | ID: biblio-1132431

RESUMO

ABSTRACT Background: Latent HIV-1 is a major hurdle in obtaining HIV-1 sustained virological remission (SVR). Here we explored histone deacetylation inhibition property of nicotinamide (NAM; n = 17) for the first time in comparison to a combination of methyltransferase inhibitors (MTIs; Chaetocin and BIX01294; n = 25) to reactivate latent HIV ex vivo in CD8-depleted PBMCs from antiretroviral treated aviremic individuals. Results: NAM reactivated HIV-1 from 13/17 (76.4%) samples compared to 20/25 (80.0%) using MTIs with mean viral load (VLs) of 4.32 and 3.22 log10 RNA copies/mL, respectively (p = 0.004). Mean purging time after NAM and MTIs stimulation was 5.1 and 6.75 days, respectively (p = 0.73). Viral purging in autologous cultures exhibited blunted HIV recovery with fluctuating VLs followed by a complete viral extinction when expanded in allogenic system. Electron microscopy from five supernatants revealed anomalous viral particles, with lack of complete viral genomes when characterized by ultradeep sequencing through metagenomics approach (n = 4). Conclusion: NAM alone was more potent HIV-1 activator than combination of MTIs, with potential of clinical use.


Assuntos
Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem , Quinazolinas/farmacologia , Azepinas/farmacologia , Ativação Viral/efeitos dos fármacos , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Niacinamida/farmacologia , Metiltransferases/antagonistas & inibidores , Piperazinas/farmacologia , Leucócitos Mononucleares/virologia , Linfócitos T CD4-Positivos , Regulação Viral da Expressão Gênica , Latência Viral , Carga Viral/efeitos dos fármacos , Tropismo Viral/efeitos dos fármacos
4.
Braz J Infect Dis ; 24(2): 150-159, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32105620

RESUMO

BACKGROUND: Latent HIV-1 is a major hurdle in obtaining HIV-1 sustained virological remission (SVR). Here we explored histone deacetylation inhibition property of nicotinamide (NAM; n=17) for the first time in comparison to a combination of methyltransferase inhibitors (MTIs; Chaetocin and BIX01294; n=25) to reactivate latent HIV ex vivo in CD8-depleted PBMCs from antiretroviral treated aviremic individuals. RESULTS: NAM reactivated HIV-1 from 13/17 (76.4%) samples compared to 20/25 (80.0%) using MTIs with mean viral load (VLs) of 4.32 and 3.22 log10 RNA copies/mL, respectively (p=0.004). Mean purging time after NAM and MTIs stimulation was 5.1 and 6.75 days, respectively (p=0.73). Viral purging in autologous cultures exhibited blunted HIV recovery with fluctuating VLs followed by a complete viral extinction when expanded in allogenic system. Electron microscopy from five supernatants revealed anomalous viral particles, with lack of complete viral genomes when characterized by ultradeep sequencing through metagenomics approach (n=4). CONCLUSION: NAM alone was more potent HIV-1 activator than combination of MTIs, with potential of clinical use.


Assuntos
Azepinas/farmacologia , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Metiltransferases/antagonistas & inibidores , Niacinamida/farmacologia , Quinazolinas/farmacologia , Ativação Viral/efeitos dos fármacos , Adulto , Linfócitos T CD4-Positivos , Feminino , Regulação Viral da Expressão Gênica , Humanos , Leucócitos Mononucleares/virologia , Masculino , Pessoa de Meia-Idade , Piperazinas/farmacologia , Carga Viral/efeitos dos fármacos , Tropismo Viral/efeitos dos fármacos , Latência Viral , Adulto Jovem
5.
Acta Biomater ; 97: 93-104, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31386928

RESUMO

Adenovirus (Ad) is a promising viral carrier in gene therapy because of its unique attribution. However, clinical applications of Ad vectors are currently restricted by their immunogenicity and broad native tropism. To address these obstacles, a variety of nonimmunogenic polymers are utilized to modify Ad vectors chemically or physically. In this review, we systemically discuss the functions of polymers in Ad-mediated gene delivery from two aspects: evading the host immune responses to Ads and redirecting Ad tropism. With polyethylene glycol (PEG) first in order, a variety of polymers have been developed to shield the surface of Ad vectors and well accomplished to evade the host immune response, block CAR-dependant cellular uptake, and reduce accumulation in the liver. In addition, shielding Ad vectors with targeted polymers (including targeting ligand-conjugated polymers and bio-responsive polymers) can also efficiently retarget Ad vectors to tumor tissues and reduce their distribution in nontargeted tissues. With its potential to evade the immune response and retarget Ad vectors, modification with polymers has been generally regarded as a promising strategy to facilitate the clinical applications of Ad vectors for virotherapy. STATEMENT OF SIGNIFICANCE: There is no doubt that Adenovirus (Ads) are attractive vectors for gene therapy, with high sophistication and effectiveness in overcoming both extra- and intracellular barriers, which cannot be exceeded by any other nonviral gene vectors. Unfortunately, their clinical applications are still restricted by some critical hurdles, including immunogenicity and native broad tropism. Therefore, a variety of elegant strategies have been developed from various angles to address these hurdles. Among these various strategies, coating Ads with nonimmunogenic polymers has attracted much attention. In this review, we systemically discuss the functions of polymers in Ad-mediated gene delivery from two aspects: evading the host immune responses to Ads and redirecting Ad tropism. In addition, the key factors in Ad modification with polymers have been highlighted and summarized to provide guiding theory for the design of more effective and safer polymer-Ad hybrid gene vectors.


Assuntos
Adenoviridae , Terapia Genética , Vetores Genéticos , Evasão da Resposta Imune/efeitos dos fármacos , Polietilenoglicóis/uso terapêutico , Transdução Genética , Tropismo Viral , Animais , Vetores Genéticos/imunologia , Vetores Genéticos/uso terapêutico , Humanos , Tropismo Viral/efeitos dos fármacos , Tropismo Viral/imunologia
6.
PLoS Pathog ; 15(6): e1007790, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31194854

RESUMO

Bourbon virus (BRBV) is an emerging tick-borne RNA virus in the orthomyxoviridae family that was discovered in 2014. Although fatal human cases of BRBV have been described, little is known about its pathogenesis, and no antiviral therapies or vaccines exist. We obtained serum from a fatal case in 2017 and successfully recovered the second human infectious isolate of BRBV. Next-generation sequencing of the St. Louis isolate of BRBV (BRBV-STL) showed >99% nucleotide identity to the original reference isolate. Using BRBV-STL, we developed a small animal model to study BRBV-STL tropism in vivo and evaluated the prophylactic and therapeutic efficacy of the experimental antiviral drug favipiravir against BRBV-induced disease. Infection of Ifnar1-/- mice lacking the type I interferon receptor, but not congenic wild-type animals, resulted in uniformly fatal disease 6 to 10 days after infection. RNA in situ hybridization and viral yield assays demonstrated a broad tropism of BRBV-STL with highest levels detected in liver and spleen. In vitro replication and polymerase activity of BRBV-STL were inhibited by favipiravir. Moreover, administration of favipiravir as a prophylaxis or as post-exposure therapy three days after infection prevented BRBV-STL-induced mortality in immunocompromised Ifnar1-/- mice. These results suggest that favipiravir may be a candidate treatment for humans who become infected with BRBV.


Assuntos
Amidas/farmacologia , Antivirais/farmacologia , Infecções por Orthomyxoviridae/prevenção & controle , Pirazinas/farmacologia , Thogotovirus/imunologia , Animais , Chlorocebus aethiops , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/imunologia , Thogotovirus/patogenicidade , Células Vero , Tropismo Viral/efeitos dos fármacos , Tropismo Viral/genética , Tropismo Viral/imunologia
7.
Viruses ; 11(3)2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30841507

RESUMO

Propagation of human cytomegalovirus (CMV) in cultured cells results in genetic adaptations that confer improved growth in vitro and significant attenuation in vivo. Mutations in RL13 arise quickly, while mutations in the UL128-131A locus emerge later during fibroblast passage and disrupt formation of a glycoprotein complex that is important for entry into epithelial and endothelial cells. As CMV replicates in the context of host antibodies in vivo, we reasoned that antibodies might mitigate the accumulation of adaptive mutations during cell culture passage. To test this, CMV in infant urine was used to infect replicate fibroblast cultures. One lineage was passaged in the absence of CMV-hyperimmuneglobulin (HIG) while the other was passaged with HIG in the culture medium. The former lost epithelial tropism and acquired mutations disrupting RL13 and UL131A expression, whereas the latter retained epithelial tropism and both gene loci remained intact after 22 passages. Additional mutations resulting in single amino acid changes also occurred in UL100 encoding glycoprotein M, UL102 encoding a subunit of the helicase/primase complex, and UL122 encoding the Immediate Early 2 protein. An epitheliotropic RL13+/UL131A+ virus was isolated by limiting dilution in the presence of HIG and expanded to produce a working stock sufficient to conduct cell tropism experiments. Thus, production of virus stocks by culture in the presence of antibodies may facilitate in vitro experiments using viruses that are genetically more authentic than previously available.


Assuntos
Anticorpos Antivirais/farmacologia , Meios de Cultura/química , Citomegalovirus/genética , Citomegalovirus/fisiologia , Fibroblastos/virologia , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos , Técnicas de Cultura de Células , Linhagem Celular , Citomegalovirus/efeitos dos fármacos , Humanos , Recém-Nascido , Mutação , Tropismo Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
8.
Nano Lett ; 19(3): 1701-1705, 2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30773888

RESUMO

Mesenchymal stem cell (MSC) has been increasingly applied to cancer therapy because of its tumor-tropic capability. However, short retention at target tissue and limited payload option hinder the progress of MSC-based cancer therapy. Herein, we proposed a hybrid spheroid/nanomedicine system, comprising MSC spheroid entrapping drug-loaded nanocomposite, to address these limitations. Spheroid formulation enhanced MSC's tumor tropism and facilitated loading of different types of therapeutic payloads. This system acted as an active drug delivery platform seeking and specifically targeting glioblastoma cells. It enabled effective delivery of combinational protein and chemotherapeutic drugs by engineered MSC and nanocomposite, respectively. In an in vivo migration model, the hybrid spheroid showed higher nanocomposite retention in the tumor tissue compared with the single MSC approach, leading to enhanced tumor inhibition in a heterotopic glioblastoma murine model. Taken together, this system integrates the merits of cell- and nanoparticle- mediated drug delivery with the tumor-homing characteristics of MSC to advance targeted combinational cancer therapy.


Assuntos
Sistemas de Liberação de Medicamentos , Glioblastoma/tratamento farmacológico , Células-Tronco Mesenquimais/química , Esferoides Celulares/transplante , Engenharia Celular/tendências , Movimento Celular/efeitos dos fármacos , Terapia Combinada , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Células-Tronco Mesenquimais/citologia , Nanomedicina/tendências , Esferoides Celulares/química , Tropismo Viral/efeitos dos fármacos
9.
PLoS One ; 13(12): e0204099, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30586365

RESUMO

Detailed clonal phenotypic/genotypic analyses explored viral-escape mechanisms during maraviroc-based therapy in highly treatment-experienced participants from the MOTIVATE trials. To allow real-time assessment of samples while maintaining a blind trial, the first 267 enrolled participants were selected for evaluation. At failure, plasma samples from 20/50 participants (16/20 maraviroc-treated) with CXCR4-using virus and all 38 (13 maraviroc-treated) with CCR5-tropic virus were evaluated. Of those maraviroc-treated participants with CXCR4-using virus at failure, genotypic and phenotypic clonal tropism determinations showed >90% correspondence in 14/16 at Day 1 and 14/16 at failure. Phylogenetic analysis of clonal sequences detected pre-treatment progenitor CXCR4-using virus, or on-treatment virus highly divergent from the Day 1 R5 virus, excluding possible co-receptor switch through maraviroc-mediated evolution. Re-analysis of pre-treatment samples using the enhanced-sensitivity Trofile® assay detected CXCR4-using virus pre-treatment in 16/20 participants failing with CXCR4-using virus. Post-maraviroc reversion of CXCR4-use to CCR5-tropic occurred in 7/8 participants with follow-up, suggesting selective maraviroc inhibition of CCR5-tropic variants in a mixed-tropic viral population, not emergence of de novo mutations in CCR5-tropic virus, as the main virologic escape mechanism. Maraviroc-resistant CCR5-tropic virus was observed in plasma from 5 treated participants with virus displaying reduced maximal percent inhibition (MPI) but no evidence of IC50 change. Env clones with reduced MPI showed 1-5 amino acid changes specific to each V3-loop region of env relative to Day 1. However, transferring on-treatment resistance-associated changes using site-directed mutagenesis did not always establish resistance in Day 1 virus, and key 'signature' mutation patterns associated with reduced susceptibility to maraviroc were not identified. Evolutionary divergence of the CXCR4-using viruses is confirmed, emphasizing natural selection not influenced directly by maraviroc; maraviroc simply unmasks pre-existing lineages by inhibiting the R5 virus. For R5-viral failure, resistance development through drug selection pressure was uncommon and manifested through reduced MPI and with virus strain-specific mutational patterns.


Assuntos
Genótipo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , HIV-1/genética , Maraviroc/administração & dosagem , Filogenia , Tropismo Viral/genética , Adulto , Feminino , Infecções por HIV/patologia , Humanos , Masculino , Maraviroc/efeitos adversos , Pessoa de Meia-Idade , Falha de Tratamento , Tropismo Viral/efeitos dos fármacos
10.
Curr Opin HIV AIDS ; 13(4): 341-345, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29771694

RESUMO

PURPOSE OF REVIEW: Even in the era of modern HAART, antiretroviral (ARV) failure and emergence of drug resistance is still a problem worldwide. New classes with different mechanisms of action are needed to overcome this challenge. After the integrase inhibitors were launched, more than a decade ago, no new classes were added to the ARV armamentarium. RECENT FINDINGS: Fostemsavir (FTR) is an attachment inhibitor, active regardless of viral tropism, without cross-resistance to any of the existing ARV compounds. A phase 3 study showed a reduction in plasma viral RNA of 1.21-1.73 log10 copies/ml from baseline after 8 days of functional monotherapy; at 48 weeks, up to 82% of patients treated with FTR and an optimized background ARV regimen achieved virological suppression below 50 copies/ml. SUMMARY: FTR is an investigational HIV drug with a novel mechanism of action that demonstrates virologic activity in HIV-infected treatment-experienced individuals.


Assuntos
Fármacos Anti-HIV/administração & dosagem , Linfócitos T CD4-Positivos/virologia , Infecções por HIV/tratamento farmacológico , HIV/efeitos dos fármacos , Organofosfatos/administração & dosagem , Piperazinas/administração & dosagem , Ligação Viral/efeitos dos fármacos , Fármacos Anti-HIV/farmacologia , Ensaios Clínicos Fase III como Assunto , HIV/fisiologia , Infecções por HIV/virologia , Humanos , Organofosfatos/farmacologia , Piperazinas/farmacologia , Tropismo Viral/efeitos dos fármacos
11.
Clin Infect Dis ; 64(3): 295-300, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27838645

RESUMO

BACKGROUND: In the absence of therapy, CXCR4 (X4)-tropic human immunodeficiency virus type 1 (HIV-1) increases over time, associated with accelerated disease progression. In contrast, the majority of patients receiving long-term combination antiretroviral therapy (cART) present with CCR5 (R5)-tropic HIV-1 variants. It is unclear whether cART itself mediates the reduction of X4-tropic HIV-1. The current study aimed at assessing the tropism of viral integrates in patients' blood during fully suppressive cART. METHODS: The relative frequencies of X4-tropic proviral HIV-1 variants were determined by means of next-generation sequencing (False Positive Rate (FPR), 3.5%; R5- or X4 tropic variants occurring at less than 2% of the total virus population) for 35 treated patients in the Swiss HIV Cohort Study and followed longitudinally over time. Full viral suppression and a continuous CD4 T-cell recovery during cART were documented for all patients. Viral phylogenetic changes and sequence evolution were analyzed. RESULTS: The majority of patients (80%) experienced no frequency increase in X4-tropic proviruses during therapy. Although some proviral sequence evolution was demonstrable in >50% of these patients during therapy, this growing viral diversity was in no case paralleled by the emergence or expansion of X4-tropic provirus variants. In the remaining 20% of patients, the documented expansion of X4-tropic provirus was based on the outgrowth of single viral variants from minority populations already present before therapy initiation. CONCLUSION: Our study demonstrates that X4-tropic HIV sharply declines in most patients during successful therapy, which indicates a preferential tropism-dependent provirus elimination in the immunocompetent host. The recently implemented World Health Organization strategies of immediate therapy initiation are fully in line with this gradual loss of X4 tropism during therapy. Moreover, the early use of coreceptor antagonists against the remaining CCR5-tropic viruses may be indicated.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/fisiologia , Receptores CXCR4/imunologia , Resposta Viral Sustentada , Tropismo Viral , Adulto , Contagem de Linfócito CD4 , Estudos de Coortes , Quimioterapia Combinada , Feminino , Infecções por HIV/imunologia , HIV-1/efeitos dos fármacos , HIV-1/genética , Heterossexualidade , Homossexualidade Masculina , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Receptores CCR5/imunologia , Carga Viral , Tropismo Viral/efeitos dos fármacos , Tropismo Viral/genética
12.
Proc Natl Acad Sci U S A ; 113(50): 14408-14413, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27911847

RESUMO

The rapid spread of Zika virus (ZIKV) and its association with abnormal brain development constitute a global health emergency. Congenital ZIKV infection produces a range of mild to severe pathologies, including microcephaly. To understand the pathophysiology of ZIKV infection, we used models of the developing brain that faithfully recapitulate the tissue architecture in early to midgestation. We identify the brain cell populations that are most susceptible to ZIKV infection in primary human tissue, provide evidence for a mechanism of viral entry, and show that a commonly used antibiotic protects cultured brain cells by reducing viral proliferation. In the brain, ZIKV preferentially infected neural stem cells, astrocytes, oligodendrocyte precursor cells, and microglia, whereas neurons were less susceptible to infection. These findings suggest mechanisms for microcephaly and other pathologic features of infants with congenital ZIKV infection that are not explained by neural stem cell infection alone, such as calcifications in the cortical plate. Furthermore, we find that blocking the glia-enriched putative viral entry receptor AXL reduced ZIKV infection of astrocytes in vitro, and genetic knockdown of AXL in a glial cell line nearly abolished infection. Finally, we evaluate 2,177 compounds, focusing on drugs safe in pregnancy. We show that the macrolide antibiotic azithromycin reduced viral proliferation and virus-induced cytopathic effects in glial cell lines and human astrocytes. Our characterization of infection in the developing human brain clarifies the pathogenesis of congenital ZIKV infection and provides the basis for investigating possible therapeutic strategies to safely alleviate or prevent the most severe consequences of the epidemic.


Assuntos
Azitromicina/farmacologia , Encéfalo/embriologia , Encéfalo/virologia , Tropismo Viral/efeitos dos fármacos , Infecção por Zika virus/tratamento farmacológico , Zika virus/efeitos dos fármacos , Zika virus/fisiologia , Encéfalo/patologia , Linhagem Celular , Efeito Citopatogênico Viral/efeitos dos fármacos , Feminino , Humanos , Recém-Nascido , Testes de Sensibilidade Microbiana , Microcefalia/tratamento farmacológico , Microcefalia/embriologia , Microcefalia/patologia , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Neuroglia/virologia , Gravidez , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/fisiologia , Tropismo Viral/fisiologia , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Zika virus/patogenicidade , Infecção por Zika virus/embriologia , Infecção por Zika virus/patologia , Receptor Tirosina Quinase Axl
13.
Antimicrob Agents Chemother ; 60(10): 6398-401, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27480849

RESUMO

TROCAI is a phenotypic tropism test developed using the virological response to a short-term exposure to maraviroc monotherapy (Maraviroc Clinical Test [MCT]). It was found that with TROCAI, a cutoff of <0.5% of dual/mixed viruses was needed to predict R5 HIV tropism. Here, we have validated TROCAI, using this cutoff, in a new cohort of 42 patients, finding a very high concordance between TROCAI and MCT (98%), and a good concordance (71 to 87%) with other genotypic/phenotypic methods.


Assuntos
Cicloexanos/farmacologia , Inibidores da Fusão de HIV/farmacologia , HIV/efeitos dos fármacos , Triazóis/farmacologia , Tropismo Viral/efeitos dos fármacos , Virologia/métodos , HIV/fisiologia , Humanos , Concentração Inibidora 50 , Maraviroc , Tropismo Viral/fisiologia
14.
Nature ; 530(7588): 108-12, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26814968

RESUMO

Adeno-associated virus (AAV) vectors are currently the leading candidates for virus-based gene therapies because of their broad tissue tropism, non-pathogenic nature and low immunogenicity. They have been successfully used in clinical trials to treat hereditary diseases such as haemophilia B (ref. 2), and have been approved for treatment of lipoprotein lipase deficiency in Europe. Considerable efforts have been made to engineer AAV variants with novel and biomedically valuable cell tropisms to allow efficacious systemic administration, yet basic aspects of AAV cellular entry are still poorly understood. In particular, the protein receptor(s) required for AAV entry after cell attachment remains unknown. Here we use an unbiased genetic screen to identify proteins essential for AAV serotype 2 (AAV2) infection in a haploid human cell line. The most significantly enriched gene of the screen encodes a previously uncharacterized type I transmembrane protein, KIAA0319L (denoted hereafter as AAV receptor (AAVR)). We characterize AAVR as a protein capable of rapid endocytosis from the plasma membrane and trafficking to the trans-Golgi network. We show that AAVR directly binds to AAV2 particles, and that anti-AAVR antibodies efficiently block AAV2 infection. Moreover, genetic ablation of AAVR renders a wide range of mammalian cell types highly resistant to AAV2 infection. Notably, AAVR serves as a critical host factor for all tested AAV serotypes. The importance of AAVR for in vivo gene delivery is further highlighted by the robust resistance of Aavr(-/-) (also known as Au040320(-/-) and Kiaa0319l(-/-)) mice to AAV infection. Collectively, our data indicate that AAVR is a universal receptor involved in AAV infection.


Assuntos
Dependovirus/fisiologia , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Tropismo Viral , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Linhagem Celular , Dependovirus/classificação , Dependovirus/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Feminino , Deleção de Genes , Terapia Genética/métodos , Especificidade de Hospedeiro , Humanos , Masculino , Camundongos , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores Virais/antagonistas & inibidores , Receptores Virais/deficiência , Receptores Virais/genética , Tropismo Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Rede trans-Golgi/efeitos dos fármacos
15.
Sci Rep ; 5: 16944, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26585833

RESUMO

HIV-1 coreceptor usage must be accurately determined before starting CCR5 antagonist-based treatment as the presence of undetected minor CXCR4-using variants can cause subsequent virological failure. Ultra-deep pyrosequencing of HIV-1 V3 env allows to detect low levels of CXCR4-using variants that current genotypic approaches miss. However, the computation of the mass of sequence data and the need to identify true minor variants while excluding artifactual sequences generated during amplification and ultra-deep pyrosequencing is rate-limiting. Arbitrary fixed cut-offs below which minor variants are discarded are currently used but the errors generated during ultra-deep pyrosequencing are sequence-dependant rather than random. We have developed an automated processing of HIV-1 V3 env ultra-deep pyrosequencing data that uses biological filters to discard artifactual or non-functional V3 sequences followed by statistical filters to determine position-specific sensitivity thresholds, rather than arbitrary fixed cut-offs. It allows to retain authentic sequences with point mutations at V3 positions of interest and discard artifactual ones with accurate sensitivity thresholds.


Assuntos
Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Fragmentos de Peptídeos/genética , Sequência de Aminoácidos , Cicloexanos/farmacologia , Genótipo , Proteína gp120 do Envelope de HIV/metabolismo , Inibidores da Fusão de HIV/farmacologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/metabolismo , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Maraviroc , Dados de Sequência Molecular , Fragmentos de Peptídeos/metabolismo , Fenótipo , Mutação Puntual , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Reprodutibilidade dos Testes , Triazóis/farmacologia , Tropismo Viral/efeitos dos fármacos , Tropismo Viral/genética , Tropismo Viral/fisiologia
16.
PLoS One ; 10(7): e0131894, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26154172

RESUMO

BACKGROUND: Insertion of T4 lysozyme (T4L) into the GPCR successfully enhanced GPCR protein stability and solubilization. However, the biological functions of the recombinant GPCR protein have not been analyzed. METHODS: We engineered the CCR5-T4L mutant and expressed and purified the soluble recombinant protein using an E.coli expression system. The antiviral effects of this recombinant protein in THP-1 cell lines, primary human macrophages, and PBMCs from different donors were investigated. We also explored the possible mechanisms underlying the observed antiviral effects. RESULTS: Our data showed the biphasic inhibitory and promotion effects of different concentrations of soluble recombinant CCR5-T4L protein on R5 tropic human immunodeficiency virus-1 (HIV-1) infection in THP-1 cell lines, human macrophages, and PBMCs from clinical isolates. We demonstrated that soluble recombinant CCR5-T4L acts as a HIV-1 co-receptor, interacts with wild type CCR5, down-regulates the surface CCR5 expression in human macrophages, and interacts with CCL5 to inhibit macrophage migration. Using binding assays, we further determined that recombinant CCR5-T4L and [125I]-CCL5 compete for the same binding site on wild type CCR5. CONCLUSIONS: Our results suggest that recombinant CCR5-T4L protein marginally promotes HIV-1 infection at low concentrations and markedly inhibits infection at higher concentrations. This recombinant protein may be helpful in the future development of anti-HIV-1 therapeutic agents.


Assuntos
Bacteriófago T4/enzimologia , Infecções por HIV/tratamento farmacológico , Muramidase/metabolismo , Receptores CCR5/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Células 3T3 , Animais , Antivirais/farmacologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Movimento Celular/efeitos dos fármacos , Quimiocina CCL5/farmacologia , Fatores Quimiotáticos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Escherichia coli/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Infecções por HIV/patologia , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Monócitos/patologia , Ligação Proteica/efeitos dos fármacos , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/farmacologia , Solubilidade , Doadores de Tecidos , Tropismo Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
17.
Ann N Y Acad Sci ; 1346(1): 7-17, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25962411

RESUMO

Assays to identify infectious organisms are critical for diagnosis and enabling the development of therapeutic agents. The demonstration that individuals with a 32-bp deletion within the CCR5 locus were resistant to human immunodeficiency virus (HIV) infection, while those heterozygous for the mutation progressed more slowly, led to the discovery of maraviroc (MVC), a CCR5 antagonist. As MVC is only active against CCR5-tropic strains of HIV, it was critical to develop a diagnostic assay to identify appropriate patients. Trofile™, a novel phenotypic tropism assay, was used to identify patients with CCR5-tropic virus for the MVC development program. Results of these clinical studies demonstrated that the assay correctly identified patients likely to respond to MVC. Over time, the performance characteristics of the phenotypic assay were enhanced, necessitating retesting of study samples. Genotypic tropism tests that have the potential to allow for local use and more rapid turnaround times are also being developed.


Assuntos
Fármacos Anti-HIV , Cicloexanos , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , Triazóis , Tropismo Viral/efeitos dos fármacos , Fármacos Anti-HIV/síntese química , Fármacos Anti-HIV/química , Fármacos Anti-HIV/uso terapêutico , Bioensaio/métodos , Ensaios Clínicos como Assunto , Cicloexanos/síntese química , Cicloexanos/química , Cicloexanos/farmacologia , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , HIV-1/efeitos dos fármacos , Humanos , Maraviroc , Receptores CCR5/efeitos dos fármacos , Receptores CCR5/metabolismo , Triazóis/síntese química , Triazóis/química , Triazóis/farmacologia
18.
PLoS One ; 10(3): e0121491, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25816132

RESUMO

The severity of influenza-related illness is mediated by many factors, including in vivo cell tropism, timing and magnitude of the immune response, and presence of pre-existing immunity. A direct way to study cell tropism and virus spread in vivo is with an influenza virus expressing a reporter gene. However, reporter gene-expressing influenza viruses are often attenuated in vivo and may be genetically unstable. Here, we describe the generation of an influenza A virus expressing GFP from a tri-cistronic NS segment. To reduce the size of this engineered gene segment, we used a truncated NS1 protein of 73 amino acids combined with a heterologous dimerization domain to increase protein stability. GFP and nuclear export protein coding information were fused in frame with the truncated NS1 open reading frame and separated from each other by 2A self-processing sites. The resulting PR8-NS1(1-73)GFP virus was successfully rescued and replicated as efficiently as the parental PR8 virus in vitro and was slightly attenuated in vivo. Flow cytometry-based monitoring of cells isolated from PR8-NS1(1-73)GFP virus infected BALB/c mice revealed that GFP expression peaked on day two in all cell types tested. In particular respiratory epithelial cells and myeloid cells known to be involved in antigen presentation, including dendritic cells (CD11c+) and inflammatory monocytes (CD11b+ GR1+), became GFP positive following infection. Prophylactic treatment with anti-M2e monoclonal antibody or oseltamivir reduced GFP expression in all cell types studied, demonstrating the usefulness of this reporter virus to analyze the efficacy of antiviral treatments in vivo. Finally, deep sequencing analysis, serial in vitro passages and ex vivo analysis of PR8-NS1(1-73)GFP virus, indicate that this virus is genetically and phenotypically stable.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Proteínas de Fluorescência Verde/metabolismo , Vírus da Influenza A/fisiologia , Infecções por Orthomyxoviridae/prevenção & controle , Proteínas não Estruturais Virais/metabolismo , Tropismo Viral/efeitos dos fármacos , Animais , Anticorpos Monoclonais/uso terapêutico , Antivirais/administração & dosagem , Antivirais/farmacologia , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Cães , Proteínas de Fluorescência Verde/genética , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/genética , Células Madin Darby de Rim Canino , Camundongos , Monócitos/metabolismo , Monócitos/virologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Oseltamivir/administração & dosagem , Oseltamivir/farmacologia , Estabilidade Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas da Matriz Viral/antagonistas & inibidores , Proteínas não Estruturais Virais/genética , Replicação Viral/efeitos dos fármacos
19.
PLoS One ; 10(2): e0116158, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25646764

RESUMO

Human parechoviruses (HPeVs), members of the family Picornaviridae, are associated with severe human clinical conditions such as gastrointestinal disease, encephalitis, meningitis, respiratory disease and neonatal sepsis. A new contemporary strain of HPeV1, KVP6 (accession no. KC769584), was isolated from a clinical specimen. Full-genome alignment revealed that HPeV1 KVP6 shares high genome homology with the German strain of HPeV1, 7555312 (accession no. FM178558) and could be classified in the clade 1B group. An intertypic recombination was shown within the P2-P3 genome regions of HPeV1. Cell-type tropism test showed that T84 cells (colon carcinoma cells), A549 cells (lung carcinoma cells) and DBTRG-5MG cells (glioblastoma cells) were susceptible to HPeV1 infection, which might be relevant clinically. A facilitated cytopathic effect and increased viral titers were reached after serial viral passages in Vero cells, with viral genome mutation found in later passages. HPeV1 is sensitive to elevated temperature because 39C incubation impaired virion production. HPeV1 induced innate immunity with phosphorylation of interferon (IFN) regulatory transcription factor 3 and production of type I IFN in A549 but not T84 cells. Furthermore, type I IFN inhibited HPeV1 production in A549 cells but not T84 cells; T84 cells may be less responsive to type I IFN stimulation. Moreover, HPeV1-infected cells showed downregulated type I IFN activation, which indicated a type I IFN evasion mechanism. The characterization of the complete genome and infection features of HPeV1 provide comprehensive information about this newly isolated HPeV1 for further diagnosis, prevention or treatment strategies.


Assuntos
Antivirais/farmacologia , Genoma Viral/genética , Interferon Tipo I/farmacologia , Parechovirus/genética , Parechovirus/fisiologia , Infecções por Picornaviridae , Animais , Antivirais/metabolismo , Linhagem Celular , Genômica , Humanos , Interferon Tipo I/metabolismo , Cinética , Dados de Sequência Molecular , Parechovirus/efeitos dos fármacos , Transdução de Sinais , Temperatura , Tropismo Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
20.
Rinsho Byori ; 63(11): 1323-7, 2015 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-26995879

RESUMO

Currently, the treatment of human immunodeficiency virus involves combination therapy, as antiretroviral therapy(ART). The treatment has improved steadily since the advent of potent combination therapy in 1996. New drugs that offer new mechanisms of action, improvements in potency and activity even against multidrug-resistant viruses, dosing convenience, and tolerability have been approved. Among ART with useful drugs, there are two important examinations before starting the treatment using the two kinds of drug. CCR5 co-receptor antagonists, maraviroc, prevent HIV entry into target cells by binding to CCR5 receptors. Genotypic assays have been developed that can determine or predict the co-receptor tropism(i.e., CCR5, CXCR4, or both) of the patient's dominant virus population. The assay for HIV-1 co-receptor usage should be performed whenever the use of a CCR5 antagonist is being considered. One of the nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), abacavir, is an important agent to develop recommended regimens for antiretroviral therapy. Serious and sometimes fatal hypersensitivity reactions have been associated with abacavir-containing products, ZIAGEN, Epzicom, and Triumeq. Patients who carry the HLA-B*5701 allele are at high-risk of a hypersensitivity reaction to abacavir. Prior to initiating therapy with abacavir, performing a screening test for the HLA-B*5701 allele is recommended. [Review].


Assuntos
Antirretrovirais/administração & dosagem , Infecções por HIV/diagnóstico , Infecções por HIV/tratamento farmacológico , HIV-1 , Alelos , Antirretrovirais/farmacologia , Antagonistas dos Receptores CCR5 , Cicloexanos/administração & dosagem , Cicloexanos/farmacologia , Didesoxinucleosídeos/administração & dosagem , Didesoxinucleosídeos/farmacologia , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/fisiologia , Antígenos HLA-B/genética , Humanos , Maraviroc , Farmacogenética , Medicina de Precisão , Inibidores da Transcriptase Reversa , Triazóis/administração & dosagem , Triazóis/farmacologia , Tropismo Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA