Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(48): e2306374120, 2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-37983491

RESUMO

The rather few cases of humans infected by HIV-1 N, O, or P raise the question of their incomplete adaptation to humans. We hypothesized that early postentry restrictions may be relevant for the impaired spread of these HIVs. One of the best-characterized species-specific restriction factors is TRIM5α. HIV-1 M can escape human (hu) TRIM5α restriction by binding cyclophilin A (CYPA, also known as PPIA, peptidylprolyl isomerase A) to the so-called CYPA-binding loop of its capsid protein. How non-M HIV-1s interact with huTRIM5α is ill-defined. By testing full-length reporter viruses (Δ env) of HIV-1 N, O, P, and SIVgor (simian IV of gorillas), we found that in contrast to HIV-1 M, the nonpandemic HIVs and SIVgor showed restriction by huTRIM5α. Work to identify capsid residues that mediate susceptibility to huTRIM5α revealed that residue 88 in the capsid CYPA-binding loop was important for such differences. There, HIV-1 M uses alanine to resist, while non-M HIV-1s have either valine or methionine, which avail them for huTRIM5α. Capsid residue 88 determines the sensitivity to TRIM5α in an unknown way. Molecular simulations indicated that capsid residue 88 can affect trans-to-cis isomerization patterns on the capsids of the viruses we tested. These differential CYPA usages by pandemic and nonpandemic HIV-1 suggest that the enzymatic activity of CYPA on the viral core might be important for its protective function against huTRIM5α.


Assuntos
Infecções por HIV , Soropositividade para HIV , HIV-1 , Humanos , Ciclofilina A/metabolismo , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , HIV-1/fisiologia , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Infecções por HIV/metabolismo
2.
Chin J Physiol ; 64(5): 251-256, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34708717

RESUMO

Heat shock protein 90α (HSP90α) has been confirmed to be upregulated in the blood in various types of tumors and may therefore serve as a potential tumor marker. However, whether HSP90α exists in nipple discharge remains unknown, and its expression and diagnostic value in nipple discharge remain unclear. In this study, the expression of HSP90α, carcinoembryonic antigen (CEA), and cancer antigen 153 in nipple discharge and blood from 128 patients was measured. Receiver operating characteristic curve was used to assess the diagnostic value of HSP90α. Further, its relationship with clinicopathological parameters of patients with breast cancer was analyzed. The results showed that the expression of HSP90α in nipple discharge was significantly higher in patients with breast cancer than in those with benign disease, and its diagnostic value was better than that of CEA. Combination of HSP90α and CEA showed better diagnostic efficacy than HSP90α or CEA alone. Moreover, the expression of HSP90α displayed a stepwise increase from benign lesions, followed by carcinoma in situ to invasive ductal carcinoma. HSP90α was positively correlated with Ki67 expression. However, there was no significant difference in the expression of HSP90α in blood between patients with breast cancer and benign disease. Further, the expression of HSP90α was higher in nipple discharge than in blood. In summary, HSP90α was upregulated in the nipple discharge of patients with breast cancer, and it may be related to the occurrence and progression of breast cancer. HSP90α in nipple discharge may serve as a potential diagnostic marker for breast cancer.


Assuntos
Neoplasias da Mama , Proteínas de Choque Térmico HSP90/genética , Derrame Papilar , Biomarcadores Tumorais , Neoplasias da Mama/diagnóstico , Feminino , Humanos
3.
J Virol ; 93(23)2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31511384

RESUMO

Lassa virus (LASV) is the causative agent of a fatal hemorrhagic fever in humans. The glycoprotein (GP) of LASV mediates viral entry into host cells, and correct processing and modification of GP by host factors is a prerequisite for virus replication. Here, using an affinity purification-coupled mass spectrometry (AP-MS) strategy, 591 host proteins were identified as interactors of LASV GP. Gene ontology analysis was performed to functionally annotate these proteins, and the oligosaccharyltransferase (OST) complex was highly enriched. Functional studies conducted by using CRISPR-Cas9-mediated knockouts showed that STT3A and STT3B, the two catalytically active isoforms of the OST complex, are essential for the propagation of the recombinant arenavirus rLCMV/LASV glycoprotein precursor, mainly via affecting virus infectivity. Knockout of STT3B, but not STT3A, caused hypoglycosylation of LASV GP, indicating a preferential requirement of LASV for the STT3B-OST isoform. Furthermore, double knockout of magnesium transporter 1 (MAGT1) and tumor suppressor candidate 3 (TUSC3), two specific subunits of STT3B-OST, also caused hypoglycosylation of LASV GP and affected virus propagation. Site-directed mutagenesis analysis revealed that the oxidoreductase CXXC active-site motif of MAGT1 or TUSC3 is essential for the glycosylation of LASV GP. NGI-1, a small-molecule OST inhibitor, can effectively reduce virus infectivity without affecting cell viability. The STT3B-dependent N-glycosylation of GP is conserved among other arenaviruses, including both the Old World and New World groups. Our study provided a systematic view of LASV GP-host interactions and revealed the preferential requirement of STT3B for LASV GP N-glycosylation.IMPORTANCE Glycoproteins play vital roles in the arenavirus life cycle by facilitating virus entry and participating in the virus budding process. N-glycosylation of GPs is responsible for their proper functioning; however, little is known about the host factors on which the virus depends for this process. In this study, a comprehensive LASV GP interactome was characterized, and further study revealed that STT3B-dependent N-glycosylation was preferentially required by arenavirus GPs and critical for virus infectivity. The two specific thioredoxin subunits of STT3B-OST MAGT1 and TUSC3 were found to be essential for the N-glycosylation of viral GP. NGI-1, a small-molecule inhibitor of OST, also showed a robust inhibitory effect on arenavirus. Our study provides new insights into LASV GP-host interactions and extends the potential targets for the development of novel therapeutics against Lassa fever in the future.


Assuntos
Glicoproteínas/metabolismo , Hexosiltransferases/metabolismo , Febre Lassa/metabolismo , Vírus Lassa/metabolismo , Proteínas de Membrana/metabolismo , Sistemas CRISPR-Cas , Proteínas de Transporte de Cátions , Linhagem Celular , Técnicas de Inativação de Genes , Glicosilação , Células HEK293 , Células HeLa , Hexosiltransferases/genética , Humanos , Vírus Lassa/genética , Vírus Lassa/patogenicidade , Proteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso , Oxirredutases/metabolismo , Isoformas de Proteínas , Receptores de Superfície Celular , Proteínas Supressoras de Tumor/genética , Internalização do Vírus
4.
Molecules ; 24(23)2019 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-31757042

RESUMO

Herpes simplex virus type 1 (HSV-1) is widespread double-stranded DNA (dsDNA) virus that establishes life-long latency and causes diverse severe symptoms. The mechanisms of HSV-1 infection and HSV-1's interactions with various host cells have been studied and reviewed extensively. Type I interferons were secreted by host cells upon HSV infection and play a vital role in controlling virus proliferation. A few studies, however, have focused on HSV-1 infection without the presence of interferon (IFN) signaling. In this study, HEK 293T cells with low toll-like receptor (TLR) and stimulator of interferon genes protein (STING) expression were infected with HSV-1 and subjected to a quantitative proteomic analysis. By using a subcellular fractionation strategy and high-performance mass spectrometry, a total of 6607 host proteins were quantified, of which 498 proteins were differentially regulated. A bioinformatics analysis indicated that multiple signaling pathways might be involved in HSV-1 infection. A further functional study indicated the role of Interferon-induced transmembrane protein 3 (IFITM3), Coiled-coil-helix-coiled-coil-helix domain-containing protein 2 (CHCHD2), and Tripartite motif-containing protein 27 (TRIM27) in inhibiting viral DNA replication and proliferation. Our data provide a global view of host responses to HSV-1 infection in HEK 293T cells and identify the proteins involved in the HSV-1 infection process.


Assuntos
Replicação do DNA/fisiologia , DNA Viral , Proteínas de Ligação a DNA , Herpesvirus Humano 1/fisiologia , Proteínas de Membrana , Proteínas Nucleares , Proteômica , Proteínas de Ligação a RNA , Fatores de Transcrição , Replicação Viral/fisiologia , DNA Viral/biossíntese , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Proteomics ; 17(5)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28067018

RESUMO

Sendai virus (SeV) is an enveloped nonsegmented negative-strand RNA virus that belongs to the genus Respirovirus of the Paramyxoviridae family. As a model pathogen, SeV has been extensively studied to define the basic biochemical and molecular biologic properties of the paramyxoviruses. In addition, SeV-infected host cells were widely employed to uncover the mechanism of innate immune response. To identify proteins involved in the SeV infection process or the SeV-induced innate immune response process, system-wide evaluations of SeV-host interactions have been performed. cDNA microarray, siRNA screening and phosphoproteomic analysis suggested that multiple signaling pathways are involved in SeV infection process. Here, to study SeV-host interaction, a global quantitative proteomic analysis was performed on SeV-infected HEK 293T cells. A total of 4699 host proteins were quantified, with 742 proteins being differentially regulated. Bioinformatics analysis indicated that regulated proteins were mainly involved in "interferon type I (IFN-I) signaling pathway" and "defense response to virus," suggesting that these processes play roles in SeV infection. Further RNAi-based functional studies indicated that the regulated proteins, tripartite motif (TRIM24) and TRIM27, affect SeV-induced IFN-I production. Our data provided a comprehensive view of host cell response to SeV and identified host proteins involved in the SeV infection process or the SeV-induced innate immune response process.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Proteoma/análise , Infecções por Respirovirus/metabolismo , Vírus Sendai/patogenicidade , Citoplasma/química , Citoplasma/metabolismo , Citoplasma/virologia , Células HEK293/virologia , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Proteínas Nucleares/análise , Proteínas Nucleares/metabolismo , Reação em Cadeia da Polimerase/métodos , Proteoma/genética , Proteoma/metabolismo , Proteômica/métodos , Reprodutibilidade dos Testes , Infecções por Respirovirus/virologia , Fatores de Transcrição/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Replicação Viral
6.
J Virol ; 90(22): 10259-10270, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27605671

RESUMO

The family Arenaviridae includes several important human pathogens that can cause severe hemorrhagic fever and greatly threaten public health. As a major component of the innate immune system, the RLR/MAVS signaling pathway is involved in recognizing viral components and initiating antiviral activity. It has been reported that arenavirus infection can suppress the innate immune response, and NP and Z proteins of pathogenic arenaviruses can disrupt RLR/MAVS signaling, thus inhibiting production of type I interferon (IFN-I). However, recent studies have shown elevated IFN-I levels in certain arenavirus-infected cells. The mechanism by which arenavirus infection induces IFN-I responses remains unclear. In this study, we determined that the L polymerase (Lp) of Mopeia virus (MOPV), an Old World (OW) arenavirus, can activate the RLR/MAVS pathway and thus induce the production of IFN-I. This activation is associated with the RNA-dependent RNA polymerase activity of Lp. This study provides a foundation for further studies of interactions between arenaviruses and the innate immune system and for the elucidation of arenavirus pathogenesis. IMPORTANCE: Distinct innate immune responses are observed when hosts are infected with different arenaviruses. It has been widely accepted that NP and certain Z proteins of arenaviruses inhibit the RLR/MAVS signaling pathway. The viral components responsible for the activation of the RLR/MAVS signaling pathway remain to be determined. In the current study, we demonstrate for the first time that the Lp of MOPV, an OW arenavirus, can activate the RLR/MAVS signaling pathway and thus induce the production of IFN-I. Based on our results, we proposed that dynamic interactions exist among Lp-produced RNA, NP, and the RLR/MAVS signaling pathway, and the outcome of these interactions may determine the final IFN-I response pattern: elevated or reduced. Our study provides a possible explanation for how IFN-I can become activated during arenavirus infection and may help us gain insights into the interactions that form between different arenavirus components and the innate immune system.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por Arenaviridae/metabolismo , Arenavirus do Velho Mundo/metabolismo , Transdução de Sinais/fisiologia , Proteínas Virais/metabolismo , Animais , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/virologia , Arenavirus/imunologia , Arenavirus/metabolismo , Arenavirus do Velho Mundo/imunologia , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , RNA Polimerases Dirigidas por DNA/metabolismo , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Imunidade Inata/imunologia , Interferon Tipo I/metabolismo , Células Vero
7.
Proteomics ; 15(22): 3784-96, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26350028

RESUMO

Enterovirus 71 (EV71) is one of the leading causes of hand, foot and mouth disease with neurological complications in some cases. To study the pathogenesis of EV71 infection, large-scale analyses of EV71 infected cells have been performed. However, most of these studies employed rhabdomyosarcoma (RD) cells or used transcriptomic strategy. Here, we performed SILAC-based quantitative proteomic analysis of EV71-infected U251 cells, a human glioma cell line. A total of 3125 host proteins were quantified, in which 451 were differentially regulated as a result of EV71 infection at 8 or 20 hpi or both. Gene Ontology analysis indicates the regulated proteins were enriched in "metabolic process", "biological regulation" and "cellular process", implying that these biological processes were affected by EV71 infection. Furthermore, functional study indicated that TRAF2 and TRAF6 among the up-regulated proteins could inhibit the replication of EV71 at the early phase post infection, and the anti-EV71 function of both proteins was independent of interferon ß. Our study not only provided an overview of cellular response to EV71 infection in a human glioma cell line, but also found that TRAF2 and TRAF6 might be potential targets to inhibit the replication of EV71. All MS data have been deposited in the ProteomeXchange with identifier PXD002454 (http://proteomecentral.proteomexchange.org/dataset/PXD002454).


Assuntos
Enterovirus Humano A/fisiologia , Interações Hospedeiro-Patógeno , Proteoma/análise , Linhagem Celular Tumoral , Biologia Computacional , Glioma , Doença de Mão, Pé e Boca/virologia , Humanos , Fator 2 Associado a Receptor de TNF/análise , Fator 6 Associado a Receptor de TNF/análise , Replicação Viral
8.
ACS Appl Mater Interfaces ; 15(16): 20563-20570, 2023 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-37040160

RESUMO

The oxygen evolution reaction (OER) is the primary bottleneck for electrochemical splitting of water into H2. Developing robust and active OER electrocatalysts through understanding the OER mechanism is essential. However, the mechanism for OER is not yet well understood even for the most studied rutile Ru-based oxide, especially in a water-solvent environment. It is still disputed whether the adsorbate evolving mechanism (AEM) is competitive with the lattice oxygen mechanism (LOM). In this article, the AEM and LOM for OER in transition metal (TM)-doped rutile RuO2 with different ratios of TM and Ru are discussed through density functional theory + U calculation. In low TM doping concentration, the evolved O2 is generated through the AEM, and the OER activity is limited by the scaling relationship of OER intermediates. In higher TM doping concentration, the evolved O2 is generated through the LOM for Cu- or Ni-doped RuO2. We find that the distribution of Ru 4d and O 2p orbitals and the adsorption energy of H and O are the major factors that affect the conversion of AEM into LOM. By explicitly considering the water-solvent environment, the LOM can result in higher theoretical OER activity arising from the effects of hydrogen-bond networks.

9.
ACS Infect Dis ; 7(6): 1409-1422, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33183004

RESUMO

Arenaviruses are a large family of enveloped negative-strand RNA viruses that include several causative agents of severe hemorrhagic fevers. Currently, there are no FDA-licensed drugs to treat arenavirus infection except for the off-labeled use of ribavirin. Here, we performed antiviral drug screening against the Old World arenavirus lymphocytic choriomeningitis virus (LCMV) using an FDA-approved drug library. Five drug candidates were identified, including mycophenolic acid, benidipine hydrochloride, clofazimine, dabrafenib, and apatinib, for having strong anti-LCMV effects. Further analysis indicated that benidipine hydrochloride inhibited LCMV membrane fusion, and an adaptive mutation on the LCMV glycoprotein D414 site was found to antagonize the anti-LCMV activity of benidipine hydrochloride. Mycophenolic acid inhibited LCMV replication by depleting GTP production. We also found mycophenolic acid, clofazimine, dabrafenib, and apatinib can inhibit the newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Owing to their FDA-approved status, these drug candidates can potentially be used rapidly in the clinical treatment of arenavirus and SARS-CoV-2 infection.


Assuntos
COVID-19 , Preparações Farmacêuticas , Ensaios de Triagem em Larga Escala , Humanos , SARS-CoV-2 , Replicação Viral
10.
Antiviral Res ; 165: 11-22, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30822450

RESUMO

Respiratory syncytial virus (RSV) infection causes significant disease in the lower respiratory tract of young children, and there is currently no licensed vaccine to prevent RSV infection. The F glycoprotein is considered a major antigenic target for RSV vaccine development. Recent evidence indicates that the pre-fusion F state, compared with the post-fusion F state, is a superior antigen for generation of neutralizing antibodies. In this study, we developed a novel vaccine antigen, RSV glycoprotein F fused with an IgG Fc fragment (F-Fc). The F-Fc fusion protein is predominantly a hexamer and could be recognized by the pre-fusion F-specific monoclonal antibody D25. Intranasal immunization with the F-Fc fusion protein promoted a protective Th1-biased cellular immune response relative to that promoted by immunization with the F protein. This immunization strategy significantly reduced the lung viral load in mice. Furthermore, immunization with F-Fc reduced lung pathology and the production of pro-inflammatory cytokines and chemokines in the lung after RSV infection. These results suggest that the F-Fc protein may be a safe and effective RSV vaccine candidate.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Lesão Pulmonar/virologia , Vacinas contra Vírus Sincicial Respiratório/uso terapêutico , Proteínas Virais de Fusão/imunologia , Animais , Fragmentos Fc das Imunoglobulinas/imunologia , Pulmão/virologia , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/patologia , Camundongos , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/patologia , Vírus Sinciciais Respiratórios/imunologia
11.
Artigo em Inglês | MEDLINE | ID: mdl-27917375

RESUMO

TLR2-dependent cellular signaling in Mycobacterium tuberculosis-infected macrophages causes apoptosis and inhibits class II major histocompatibility complex (MHC-II) molecules antigen processing, leading to evasion of surveillance. Mycobacterium tuberculosis (MTB) lipoproteins are an important class of Toll-like receptor (TLR) ligand, and identified as specific components that mediate these effects. In this study, we identified and characterized MTB lipoprotein Rv1016c (lpqT) as a cell wall associated-protein that was exposed on the cell surface and enhanced the survival of recombinants M. smegmatis_Rv1016c under stress conditions. We found that Rv1016c lipoprotein was a novel TLR2 ligand and able to induce macrophage apoptosis in a both dose- and time-dependent manner. Additionally, apoptosis induced by Rv1016c was reserved in THP-1 cells blocked with anti-TLR-2 Abs or in TLR2-/- mouse macrophages, indicating that Rv1016c-induced apoptosis is dependent on TLR2. Moreover, we demonstrated that Rv1016c lipoprotein inhibited IFN-γ-induced MHC-II expression and processing of soluble antigens in a TLR2 dependent manner. Class II transactivator (CIITA) regulates MHC II expression. In this context, Rv1016c lipoprotein diminished IFN-γ-induced expression of CIITA IV through TLR2 and MAPK Signaling. TLR2-dependent apoptosis and inhibition of MHC-II Ag processing induced by Rv1016c during mycobacteria infection may promote the release of residual bacilli from apoptotic cells and decrease recognition by CD4+ T cells. These mechanisms may allow intracellular MTB to evade immune surveillance and maintain chronic infection.


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/farmacologia , Antígenos de Histocompatibilidade Classe II/efeitos dos fármacos , Lipoproteínas/genética , Macrófagos/microbiologia , Mycobacterium tuberculosis/metabolismo , Proteínas Recombinantes/farmacologia , Receptor 2 Toll-Like/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Parede Celular/química , DNA Bacteriano , Modelos Animais de Doenças , Regulação da Expressão Gênica , Genes Bacterianos/genética , Humanos , Interferon gama/efeitos dos fármacos , Lipoproteínas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium smegmatis , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Proteínas Nucleares/efeitos dos fármacos , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Superfície Celular/fisiologia , Proteínas Recombinantes/genética , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Receptor 2 Toll-Like/genética , Receptores Toll-Like/classificação , Receptores Toll-Like/metabolismo , Transativadores/efeitos dos fármacos , Tuberculose/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA