Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Rev. cuba. salud pública ; 48(4)dic. 2022.
Artigo em Espanhol | LILACS, CUMED | ID: biblio-1441841

RESUMO

Introducción: En la transmisión de la COVID-19 en Santiago de Cuba se distinguieron tres brotes epidémicos entre 2020 y 2021. Objetivo: Identificar las diferencias entre los contagios intra y extradomiciliarios en tres brotes epidémicos de COVID-19 en Santiago de Cuba entre marzo de 2020 y mayo de 2021. Métodos: Se realizó un estudio descriptivo transversal de los casos de COVID-19 del territorio y el período referidos, mediante las técnicas bivariadas habituales de la estadística y el análisis estadístico implicativo, con una muestra de 6408 que se eligió por muestreo aleatorio simple de la base de datos de casos confirmados. Resultados: El contagio extradomiciliario fue significativamente mayor que el intradomiciliario sin diferencias por sexo, pero sí según grupos de edades y municipios dentro y entre ambos grupos. Fue significativo el predominio de los adultos mayores en el contagio intradomiciliario y de los adultos jóvenes en el extradomiciliario. Primaron los sintomáticos en el intradomiciliario; y, los asintomáticos, en el extradomiciliario, sin diferencias significativas entre ambas formas. Los menores de 20 años de edad, adultos mayores, asintomáticos y el municipio Mella fueron las características que se asociaron con el contagio intradomiciliario, mientras, con el extradomiciliario, los adultos jóvenes sintomáticos. Conclusiones: Las formas de contagio intra y extradomiciliaria se modularon según la conducta de las personas y el aislamiento propio de cada grupo de edades. La extradomiciliaria predominó en edades intermedias de la vida, como expresión de la conducta mediada por su responsabilidad económica en el hogar, mientras las edades extremas, que permanecieron en casa por cumplir medidas de aislamiento, fueron más propensas a la intradomiciliaria(AU)


Introduction: In the transmission of COVID-19 in Santiago de Cuba province, three epidemic outbreaks were observed between 2020 and 2021. Objective: To identify the differences between intra- and extra-domiciliary infections in three epidemic outbreaks of COVID-19 in Santiago de Cuba between March 2020 and May 2021. Methods: A cross-sectional descriptive study of COVID-19 cases in the territory and period above mentioned was carried out, using the usual bivariate techniques of statistics and implicative statistical analysis, to a sample of 6408 cass that was chosen by simple random sampling from the database of confirmed cases. Results: Extra-domiciliary contagion was significantly higher than intra-domiciliary contagion without differences by sex, but according to age groups and municipalities within and between both groups. The predominance of older adults in intra-domiciliary contagion and of young adults in extra-domiciliary contagion was significant. Symptomatic patients prevailed in the intra-domiciliary; and, the asymptomatic, in the extra-domiciliary, without significant differences between both forms. Children under 20 years of age, older adults, asymptomatic and Mella municipality were the characteristics that were associated with intra-domiciliary contagion, while, with the extra-domiciliary were related symptomatic young adults. Conclusions: The forms of intra- and extra-domiciliary contagion were modulated according to the behavior of the people and the isolation of each age group. Extra-domiciliary predominated in intermediate ages of life, as an expression of the behavior mediated by their economic responsibility at home, while extreme ages, who remained at home to comply with isolation measures, were more prone to intra-domiciliary contagion(AU)


Assuntos
Humanos , Masculino , Feminino , Liberação de Vírus/imunologia , COVID-19/transmissão , COVID-19/epidemiologia , Epidemiologia Descritiva , Estudos Transversais
2.
Nat Commun ; 12(1): 4629, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34330917

RESUMO

Since the outbreak of the SARS-CoV-2 pandemic, there have been intense structural studies on purified viral components and inactivated viruses. However, structural and ultrastructural evidence on how the SARS-CoV-2 infection progresses in the native cellular context is scarce, and there is a lack of comprehensive knowledge on the SARS-CoV-2 replicative cycle. To correlate cytopathic events induced by SARS-CoV-2 with virus replication processes in frozen-hydrated cells, we established a unique multi-modal, multi-scale cryo-correlative platform to image SARS-CoV-2 infection in Vero cells. This platform combines serial cryoFIB/SEM volume imaging and soft X-ray cryo-tomography with cell lamellae-based cryo-electron tomography (cryoET) and subtomogram averaging. Here we report critical SARS-CoV-2 structural events - e.g. viral RNA transport portals, virus assembly intermediates, virus egress pathway, and native virus spike structures, in the context of whole-cell volumes revealing drastic cytppathic changes. This integrated approach allows a holistic view of SARS-CoV-2 infection, from the whole cell to individual molecules.


Assuntos
COVID-19/imunologia , SARS-CoV-2/imunologia , Montagem de Vírus/imunologia , Liberação de Vírus/imunologia , Replicação Viral/imunologia , Animais , COVID-19/epidemiologia , COVID-19/virologia , Chlorocebus aethiops , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Humanos , Pandemias/prevenção & controle , SARS-CoV-2/fisiologia , SARS-CoV-2/ultraestrutura , Células Vero , Montagem de Vírus/fisiologia , Liberação de Vírus/fisiologia , Replicação Viral/fisiologia
3.
Mol Cell ; 81(12): 2656-2668.e8, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33930332

RESUMO

A deficient interferon (IFN) response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been implicated as a determinant of severe coronavirus disease 2019 (COVID-19). To identify the molecular effectors that govern IFN control of SARS-CoV-2 infection, we conducted a large-scale gain-of-function analysis that evaluated the impact of human IFN-stimulated genes (ISGs) on viral replication. A limited subset of ISGs were found to control viral infection, including endosomal factors inhibiting viral entry, RNA binding proteins suppressing viral RNA synthesis, and a highly enriched cluster of endoplasmic reticulum (ER)/Golgi-resident ISGs inhibiting viral assembly/egress. These included broad-acting antiviral ISGs and eight ISGs that specifically inhibited SARS-CoV-2 and SARS-CoV-1 replication. Among the broad-acting ISGs was BST2/tetherin, which impeded viral release and is antagonized by SARS-CoV-2 Orf7a protein. Overall, these data illuminate a set of ISGs that underlie innate immune control of SARS-CoV-2/SARS-CoV-1 infection, which will facilitate the understanding of host determinants that impact disease severity and offer potential therapeutic strategies for COVID-19.


Assuntos
Antígenos CD/genética , Interações Hospedeiro-Patógeno/genética , Fatores Reguladores de Interferon/genética , Interferon Tipo I/genética , SARS-CoV-2/genética , Proteínas Virais/genética , Animais , Antígenos CD/química , Antígenos CD/imunologia , Sítios de Ligação , Linhagem Celular Tumoral , Chlorocebus aethiops , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/virologia , Proteínas Ligadas por GPI/química , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Regulação da Expressão Gênica , Complexo de Golgi/genética , Complexo de Golgi/imunologia , Complexo de Golgi/virologia , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Fatores Reguladores de Interferon/classificação , Fatores Reguladores de Interferon/imunologia , Interferon Tipo I/imunologia , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , SARS-CoV-2/imunologia , Transdução de Sinais , Células Vero , Proteínas Virais/química , Proteínas Virais/imunologia , Internalização do Vírus , Liberação de Vírus/genética , Liberação de Vírus/imunologia , Replicação Viral/genética , Replicação Viral/imunologia
4.
J Virol ; 94(19)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32699087

RESUMO

Chikungunya fever, a mosquito-borne disease manifested by fever, rash, myalgia, and arthralgia, is caused by chikungunya virus (CHIKV), which belongs to the genus Alphavirus of the family Togaviridae Anti-CHIKV IgG from convalescent patients is known to directly neutralize CHIKV, and the state of immunity lasts throughout life. Here, we examined the epitope of a neutralizing mouse monoclonal antibody against CHIKV, CHE19, which inhibits viral fusion and release. In silico docking analysis showed that the epitope of CHE19 was localized in the viral E2 envelope and consisted of two separate segments, an N-linker and a ß-ribbon connector, and that its bound Fab fragment on E2 overlapped the position that the E3 glycoprotein originally occupied. We showed that CHIKV-E2 is lost during the viral internalization and that CHE19 inhibits the elimination of CHIKV-E2. These findings suggested that CHE19 stabilizes the E2-E1 heterodimer instead of E3 and inhibits the protrusion of the E1 fusion loop and subsequent membrane fusion. In addition, the antigen-bound Fab fragment configuration showed that CHE19 connects to the CHIKV spikes existing on the two individual virions, leading us to conclude that the CHE19-CHIKV complex was responsible for the large virus aggregations. In our subsequent filtration experiments, large viral aggregations by CHE19 were trapped by a 0.45-µm filter. This virion-connecting characteristic of CHE19 could explain the inhibition of viral release from infected cells by the tethering effect of the virion itself. These findings provide clues toward the development of effective prophylactic and therapeutic monoclonal antibodies against the Alphavirus infection.IMPORTANCE Recent outbreaks of chikungunya fever have increased its clinical importance. Neither a specific antiviral drug nor a commercial vaccine for CHIKV infection are available. Here, we show a detailed model of the docking between the envelope glycoprotein of CHIKV and our unique anti-CHIKV-neutralizing monoclonal antibody (CHE19), which inhibits CHIKV membrane fusion and virion release from CHIKV-infected cells. Homology modeling of the neutralizing antibody CHE19 and protein-protein docking analysis of the CHIKV envelope glycoprotein and CHE19 suggested that CHE19 inhibits the viral membrane fusion by stabilizing the E2-E1 heterodimer and inhibits virion release by facilitating the formation of virus aggregation due to the connecting virions, and these predictions were confirmed by experiments. Sequence information of CHE19 and the CHIKV envelope glycoprotein and their docking model will contribute to future development of an effective prophylactic and therapeutic agent.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Febre de Chikungunya/imunologia , Vírus Chikungunya/imunologia , Proteínas do Envelope Viral/imunologia , Internalização do Vírus/efeitos dos fármacos , Liberação de Vírus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/química , Anticorpos Antivirais/farmacologia , Vírus Chikungunya/genética , Vírus Chikungunya/fisiologia , Chlorocebus aethiops , Epitopos/imunologia , Feminino , Fusão de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Simulação de Acoplamento Molecular , Domínios e Motivos de Interação entre Proteínas , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/imunologia , Liberação de Vírus/efeitos dos fármacos
5.
Virus Res ; 287: 198096, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-32682818

RESUMO

During herpesvirus replication, newly synthesized nucleocapsids exit the nucleus by a vesicle-mediated transport, which requires the nuclear egress complex (NEC), composed of the conserved viral proteins designated as pUL31 and pUL34 in the alphaherpesviruses pseudorabies virus (PrV) and herpes simplex viruses. Oligomerization of the heterodimeric NEC at the inner nuclear membrane (INM) results in membrane bending and budding of virus particles into the perinuclear space. The INM-derived primary envelope then fuses with the outer nuclear membrane to release nucleocapsids into the cytoplasm. The two NEC components are necessary and sufficient for induction of vesicle budding and scission as shown after co-expression in eukaryotic cells or in synthetic membranes. However, where and when the NEC is formed, how membrane curvature is mediated and how it is regulated, remains unclear. While monospecific antisera raised against the different components of the PrV NEC aided in the characterization and intracellular localization of the individual proteins, no NEC specific tools have been described yet for any herpesvirus. To gain more insight into vesicle budding and scission, we aimed at generating NEC specific monoclonal antibodies (mAbs). To this end, mice were immunized with bacterially expressed soluble PrV NEC, which was previously used for structure determination. Besides pUL31- and pUL34-specific mAbs, we also identified mAbs, which reacted only in the presence of both proteins indicating specificity for the complex. Confocal microscopy with those NEC-specific mAbs revealed small puncta (approx. 0.064 µm2) along the nuclear rim in PrV wild type infected cells. In contrast, ca. 5-fold larger speckles (approx. 0.35 µm2) were detectable in cells infected with a PrV mutant lacking the viral protein kinase pUS3, which is known to accumulate primary enveloped virions in the PNS within large invaginations of the INM, or in cells co-expressing pUL31 and pUL34. Kinetic experiments showed that while the individual proteins were detectable already between 2-4 hours after infection, the NEC-specific mAbs produced significant staining only after 4-6 hours in accordance with timing of nuclear egress. Taken together, the data indicate that these mAbs specifically label the PrV NEC.


Assuntos
Anticorpos Monoclonais/imunologia , Herpesvirus Suídeo 1/imunologia , Membrana Nuclear/metabolismo , Proteínas Virais/imunologia , Liberação de Vírus/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Linhagem Celular , Feminino , Herpesvirus Suídeo 1/genética , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Imunoeletrônica , Membrana Nuclear/imunologia , Nucleocapsídeo/metabolismo , Coelhos , Proteínas Virais/genética
6.
Methods Mol Biol ; 1911: 441-450, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30593644

RESUMO

Hepatitis C virus (HCV) pseudoparticles (HCVpp) are generated by cotransfection of HCV envelope (E1 and E2) genes along with a retroviral packaging/reporter construct into HEK293T cells. Enveloped particles bearing HCV E1E2 proteins on their surface are released through a retroviral budding process into the supernatant. Viral E1E2 glycoproteins facilitate a single round of receptor-mediated entry of HCVpp into hepatoma cells, which can be quantified by reporter gene expression. These HCVpp have been employed to study mechanisms of HCV entry into hepatoma cells, as well as HCV neutralization by immune sera or HCV-specific monoclonal antibodies.


Assuntos
Anticorpos Anti-Hepatite C/imunologia , Hepatite C/imunologia , Testes de Neutralização/métodos , Internalização do Vírus , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Linhagem Celular Tumoral , Genes Reporter/genética , Células HEK293 , Hepacivirus/imunologia , Hepatite C/sangue , Hepatite C/virologia , Anticorpos Anti-Hepatite C/sangue , Humanos , Testes de Neutralização/instrumentação , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vírion/imunologia , Liberação de Vírus/imunologia
7.
J Cell Biol ; 217(8): 2911-2929, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29921601

RESUMO

Septins are conserved components of the cytoskeleton that play important roles in many fundamental cellular processes including division, migration, and membrane trafficking. Septins can also inhibit bacterial infection by forming cage-like structures around pathogens such as Shigella We found that septins are recruited to vaccinia virus immediately after its fusion with the plasma membrane during viral egress. RNA interference-mediated depletion of septins increases virus release and cell-to-cell spread, as well as actin tail formation. Live cell imaging reveals that septins are displaced from the virus when it induces actin polymerization. Septin loss, however, depends on the recruitment of the SH2/SH3 adaptor Nck, but not the activity of the Arp2/3 complex. Moreover, it is the recruitment of dynamin by the third Nck SH3 domain that displaces septins from the virus in a formin-dependent fashion. Our study demonstrates that septins suppress vaccinia release by "entrapping" the virus at the plasma membrane. This antiviral effect is overcome by dynamin together with formin-mediated actin polymerization.


Assuntos
Septinas/fisiologia , Vacínia/imunologia , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Membrana Celular/virologia , Clatrina/análise , Clatrina/metabolismo , Dinaminas/metabolismo , Dinaminas/fisiologia , Células HeLa , Humanos , Proteínas Oncogênicas/metabolismo , Proteínas Oncogênicas/fisiologia , Fosforilação , Interferência de RNA , Septinas/análise , Septinas/antagonistas & inibidores , Transdução de Sinais , Liberação de Vírus/imunologia
8.
Viral Immunol ; 30(7): 500-507, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28598267

RESUMO

Intravenous immunoglobulin (IVIG) is used to treat or prevent severe viral infection, especially cytomegalovirus (CMV) infections. IVIG was characterized to understand its interaction with CMV-infected cells. IVIG retarded CMV spread and reduced virus yields depending on the neutralizing (NT) antibody titer. Immediate early protein synthesis was reduced by IVIG in 3 to 15 h, and IVIG specifically reduced the ratio of 66/68k protein synthesis among immediate early proteins in an NT antibody-dependent manner between 4 and 8 h after infection, indicating that antigenic modulation of CMV-infected cells by IVIG reduced viral protein synthesis and virus production. The half-life of antibody bound to CMV-infected cells was 3.8 h. NT antibody titers to varicella-zoster virus (VZV) and CMV in IVIG were dose dependently absorbed by cells infected with VZV and CMV, respectively, but the antibody titers to CMV and VZV, respectively, were not affected. NT antibody in 0.3 mL of IVIG (15 mg) was specifically absorbed by 108 CMV-infected cells and 107 VZV-infected cells, suggesting that the NT antibody in IVIG might be inactivated by one-tenth of a similar volume of CMV-infected or VZV-infected cells. Various antiviral activities of IVIG may contribute to control and alleviation of CMV infection.


Assuntos
Anticorpos Neutralizantes/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Imunoglobulina G/imunologia , Anticorpos Antivirais/imunologia , Modulação Antigênica , Antivirais/imunologia , Células Cultivadas , Humanos , Proteínas Imediatamente Precoces/metabolismo , Imunoglobulina G/metabolismo , Imunoglobulinas Intravenosas/imunologia , Testes de Neutralização , Liberação de Vírus/imunologia
9.
Biochim Biophys Acta Biomembr ; 1859(4): 550-560, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27793589

RESUMO

The HIV gp160 envelope fusion protein is situated in the viral membrane and mediates virus entry into its host cell. Increasing evidence suggests that virtually all parts of the HIV envelope are structurally and functionally dependent on membranes. Protein-lipid interactions and membrane properties influence the dynamics of a manifold of gp160 biological activities such as membrane fusion, immune suppression and gp160 incorporation into virions during HIV budding and assembly. In the following we will summarize our current understanding of this interdependence between membrane interaction, structural conformation and functionality of the different gp160 domains. This article is part of a Special Issue entitled: Lipid order/lipid defects and lipid-control of protein activity edited by Dirk Schneider.


Assuntos
Proteína gp120 do Envelope de HIV/química , Proteína gp160 do Envelope de HIV/química , Proteína gp41 do Envelope de HIV/química , HIV-1/química , Microdomínios da Membrana/química , Esfingomielinas/química , Sequência de Aminoácidos , Expressão Gênica , Proteína gp120 do Envelope de HIV/genética , Proteína gp120 do Envelope de HIV/imunologia , Proteína gp160 do Envelope de HIV/genética , Proteína gp160 do Envelope de HIV/imunologia , Proteína gp41 do Envelope de HIV/genética , Proteína gp41 do Envelope de HIV/imunologia , HIV-1/imunologia , Interações Hospedeiro-Patógeno , Humanos , Fusão de Membrana , Microdomínios da Membrana/imunologia , Microdomínios da Membrana/virologia , Conformação Proteica , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Esfingomielinas/imunologia , Linfócitos T/imunologia , Linfócitos T/virologia , Montagem de Vírus/imunologia , Liberação de Vírus/imunologia
10.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795429

RESUMO

Influenza A virus (IAV) matrix protein 2 (M2) plays multiple roles in the early and late phases of viral infection. Once synthesized, M2 is translocated to the endoplasmic reticulum (ER), travels to the Golgi apparatus, and is sorted at the trans-Golgi network (TGN) for transport to the apical plasma membrane, where it functions in virus budding. We hypothesized that M2 trafficking along with its secretory pathway must be finely regulated, and host factors could be involved in this process. However, no studies examining the role of host factors in M2 posttranslational transport have been reported. Here, we used a yeast two-hybrid (Y2H) system to screen for host proteins that interact with the M2 protein and identified transport protein particle complex 6A (TRAPPC6A) as a potential binding partner. We found that both TRAPPC6A and its N-terminal internal-deletion isoform, TRAPPC6A delta (TRAPPC6AΔ), interact with M2. Truncation and mutation analyses showed that the highly conserved leucine residue at position 96 of M2 is critical for mediating this interaction. The role of TRAPPC6AΔ in the viral life cycle was investigated by the knockdown of endogenous TRAPPC6AΔ with small interfering RNA (siRNA) and by generating a recombinant virus that was unable to interact with TRAPPC6A/TRAPPC6AΔ. The results indicated that TRAPPC6AΔ, through its interaction with M2, slows M2 trafficking to the apical plasma membrane, favors viral replication in vitro, and positively modulates virus virulence in mice. IMPORTANCE: The influenza A virus M2 protein regulates the trafficking of not only other proteins but also itself along the secretory pathway. However, the host factors involved in the regulation of the posttranslational transport of M2 are largely unknown. In this study, we identified TRAPPC6A and its N-terminal internal-deletion isoform, TRAPPC6AΔ, as interacting partners of M2. We found that the leucine (L) residue at position 96 of M2 is critical for mediating this interaction, which leads us to propose that the high level of conservation of 96L is a consequence of M2 adaptation to its interacting host factor TRAPPC6A/TRAPPC6AΔ. Importantly, we discovered that TRAPPC6AΔ can positively regulate viral replication in vitro by modulating M2 trafficking to the plasma membrane.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H1N1/genética , Virus da Influenza A Subtipo H5N1/genética , Infecções por Orthomyxoviridae/virologia , Proteínas Recombinantes de Fusão/química , Proteínas de Transporte Vesicular/química , Proteínas da Matriz Viral/química , Animais , Linhagem Celular Tumoral , Membrana Celular/imunologia , Membrana Celular/virologia , Cães , Células Epiteliais/virologia , Feminino , Expressão Gênica , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H1N1/química , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/patogenicidade , Virus da Influenza A Subtipo H5N1/química , Virus da Influenza A Subtipo H5N1/crescimento & desenvolvimento , Virus da Influenza A Subtipo H5N1/patogenicidade , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Neuroglia/virologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/mortalidade , Ligação Proteica , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Análise de Sobrevida , Técnicas do Sistema de Duplo-Híbrido , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/imunologia , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/imunologia , Liberação de Vírus/genética , Liberação de Vírus/imunologia , Replicação Viral/genética , Replicação Viral/imunologia , Rede trans-Golgi/virologia
11.
J Virol ; 89(18): 9631-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26178987

RESUMO

UNLABELLED: The "shock and kill" model of human immunodeficiency virus type 1 (HIV-1) eradication involves the induction of transcription of HIV-1 genes in latently infected CD4(+) T cells, followed by the elimination of these infected CD4(+) T cells by CD8(+) T cells or other effector cells. CD8(+) T cells may also be needed to control the spread of new infection if residual infected cells are present at the time combination antiretroviral therapy (cART) is discontinued. In order to determine the time frame needed for CD8(+) T cells to effectively prevent the spread of HIV-1 infection, we examined the kinetics of HIV transcription and virus release in latently infected cells reactivated ex vivo. Isolated resting, primary CD4(+) T cells from HIV-positive (HIV+) subjects on suppressive regimens were found to upregulate cell-associated HIV-1 mRNA within 1 h of stimulation and produce extracellular virus as early as 6 h poststimulation. In spite of the rapid kinetics of virus production, we show that CD8(+) T cells from 2 out of 4 viremic controllers were capable of effectively eliminating reactivated autologous CD4(+) cells that upregulate cell-associated HIV-1 mRNA. The results have implications for devising strategies to prevent rebound viremia due to reactivation of rare latently infected cells that persist after potentially curative therapy. IMPORTANCE: A prominent HIV-1 cure strategy termed "shock and kill" involves the induction of HIV-1 transcription in latently infected CD4(+) T cells with the goal of elimination of these cells by either the cytotoxic T lymphocyte response or other immune cell subsets. However, the cytotoxic T cell response may also be required after curative treatment if residual latently infected cells remain. The kinetics of HIV-1 reactivation indicate rapid upregulation of cell-associated HIV-1 mRNA and a 5-h window between transcription and virus release. Thus, HIV-specific CD8(+) T cell responses likely have a very short time frame to eliminate residual latently infected CD4(+) T cells that become reactivated after discontinuation of antiretroviral therapy following potentially curative treatment strategies.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Modelos Imunológicos , Ativação Viral/imunologia , Latência Viral/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/patologia , Feminino , Infecções por HIV/patologia , Humanos , Masculino , Liberação de Vírus/imunologia
12.
J Virol ; 89(3): 1550-63, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25392225

RESUMO

UNLABELLED: The envelope of influenza A viruses contains two large antigens, hemagglutinin (HA) and neuraminidase (NA). Conventional influenza virus vaccines induce neutralizing antibodies that are predominantly directed to the HA globular head, a domain that is subject to extensive antigenic drift. Antibodies directed to NA are induced at much lower levels, probably as a consequence of the immunodominance of the HA antigen. Although antibodies to NA may affect virus release by inhibiting the sialidase function of the glycoprotein, the antigen has been largely neglected in past vaccine design. In this study, we characterized the protective properties of monospecific immune sera that were generated by vaccination with recombinant RNA replicon particles encoding NA. These immune sera inhibited hemagglutination in an NA subtype-specific and HA subtype-independent manner and interfered with infection of MDCK cells. In addition, they inhibited the sialidase activities of various influenza viruses of the same and even different NA subtypes. With this, the anti-NA immune sera inhibited the spread of H5N1 highly pathogenic avian influenza virus and HA/NA-pseudotyped viruses in MDCK cells in a concentration-dependent manner. When chickens were immunized with NA recombinant replicon particles and subsequently infected with low-pathogenic avian influenza virus, inflammatory serum markers were significantly reduced and virus shedding was limited or eliminated. These findings suggest that NA antibodies can inhibit virus dissemination by interfering with both virus attachment and egress. Our results underline the potential of high-quality NA antibodies for controlling influenza virus replication and place emphasis on NA as a vaccine antigen. IMPORTANCE: The neuraminidase of influenza A viruses is a sialidase that acts as a receptor-destroying enzyme facilitating the release of progeny virus from infected cells. Here, we demonstrate that monospecific anti-NA immune sera inhibited not only sialidase activity, but also influenza virus hemagglutination and infection of MDCK cells, suggesting that NA antibodies can interfere with virus attachment. Inhibition of both processes, virus release and virus binding, may explain why NA antibodies efficiently blocked virus dissemination in vitro and in vivo. Anti-NA immune sera showed broader reactivity than anti-HA sera in hemagglutination inhibition tests and demonstrated cross-subtype activity in sialidase inhibition tests. These remarkable features of NA antibodies highlight the importance of the NA antigen for the development of next-generation influenza virus vaccines.


Assuntos
Soros Imunes/imunologia , Vírus da Influenza A/imunologia , Neuraminidase/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Galinhas , Cães , Influenza Aviária/prevenção & controle , Neuraminidase/administração & dosagem , Suínos , Proteínas Virais/administração & dosagem , Internalização do Vírus , Liberação de Vírus/imunologia , Eliminação de Partículas Virais
13.
AIDS Rev ; 16(4): 223-35, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25350531

RESUMO

Dendritic cells are professional antigen-presenting cells and key elements of both innate and adaptive immunity. Tissues like skin and mucosal epithelium, more exposed to the environment, are particularly rich in dendritic cells. Given that HIV is mainly transmitted through mucosal surfaces, the cellular mechanisms governing the initial interactions between HIV and dendritic cells are crucial for establishing systemic infection in a new host. Upon HIV/dendritic cell interaction, viral particles carried by exposed dendritic cells are transmitted to activated CD4+ T-cells during the antigen presentation process. Such dendritic cell/T-cell transmission of HIV plays an important role in the viral dissemination and immune dysregulation associated with HIV infection, subverting the bridge between innate and adaptive immune responses. Thus, defining how HIV interacts with dendritic cells remains a critical area of research, with downstream implications in the knowledge of pathogenic mechanisms, transmission, vaccine development, and molecular targets for therapeutic intervention. In this review we will, therefore, delve into the mechanisms involved in HIV/dendritic cell interactions that govern viral persistence, cellular trafficking, transmission and restriction, compiling the present knowledge on these subjects and attempting to postulate how some uncertain pathways may shape up and intertwine.


Assuntos
Comunicação Celular , Células Dendríticas/fisiologia , Infecções por HIV/fisiopatologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Imunidade Adaptativa , Moléculas de Adesão Celular , Comunicação Celular/fisiologia , Células Dendríticas/imunologia , Infecções por HIV/imunologia , Infecções por HIV/transmissão , HIV-1/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/fisiologia , Receptor Cross-Talk , Linfócitos T/imunologia , Liberação de Vírus/imunologia , Replicação Viral
14.
PLoS Pathog ; 10(7): e1004237, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25058429

RESUMO

Herpes simplex viruses (HSV) are significant human pathogens that provide one of the best-described examples of viral latency and reactivation. HSV latency occurs in sensory neurons, being characterized by the absence of virus replication and only fragmentary evidence of protein production. In mouse models, HSV latency is especially stable but the detection of some lytic gene transcription and the ongoing presence of activated immune cells in latent ganglia have been used to suggest that this state is not entirely quiescent. Alternatively, these findings can be interpreted as signs of a low, but constant level of abortive reactivation punctuating otherwise silent latency. Using single cell analysis of transcription in mouse dorsal root ganglia, we reveal that HSV-1 latency is highly dynamic in the majority of neurons. Specifically, transcription from areas of the HSV genome associated with at least one viral lytic gene occurs in nearly two thirds of latently-infected neurons and more than half of these have RNA from more than one lytic gene locus. Further, bioinformatics analyses of host transcription showed that progressive appearance of these lytic transcripts correlated with alterations in expression of cellular genes. These data show for the first time that transcription consistent with lytic gene expression is a frequent event, taking place in the majority of HSV latently-infected neurons. Furthermore, this transcription is of biological significance in that it influences host gene expression. We suggest that the maintenance of HSV latency involves an active host response to frequent viral activity.


Assuntos
Herpes Simples/imunologia , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Células Receptoras Sensoriais/imunologia , Latência Viral/imunologia , Liberação de Vírus/imunologia , Animais , Modelos Animais de Doenças , Herpes Simples/patologia , Humanos , Camundongos , Camundongos Transgênicos , Células Receptoras Sensoriais/patologia , Células Receptoras Sensoriais/virologia
15.
J Virol ; 88(16): 8813-25, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24872578

RESUMO

UNLABELLED: Human immunodeficiency virus type 1 (HIV-1) exploits dendritic cells (DCs) to promote its transmission to T cells. We recently reported that the capture of HIV-1 by mature dendritic cells (MDCs) is mediated by an interaction between the glycosphingolipid (GSL) GM3 on virus particles and CD169/Siglec-1 on MDCs. Since HIV-1 preferentially buds from GSL-enriched lipid microdomains on the plasma membrane, we hypothesized that the virus assembly and budding site determines the ability of HIV-1 to interact with MDCs. In support of this hypothesis, mutations in the N-terminal basic domain (29/31KE) or deletion of the membrane-targeting domain of the HIV-1 matrix (MA) protein that altered the virus assembly and budding site to CD63(+)/Lamp-1-positive intracellular compartments resulted in lower levels of virion incorporation of GM3 and attenuation of virus capture by MDCs. Furthermore, MDC-mediated capture and transmission of MA mutant viruses to T cells were decreased, suggesting that HIV-1 acquires GSLs via budding from the plasma membrane to access the MDC-dependent trans infection pathway. Interestingly, MDC-mediated capture of Nipah and Hendra virus (recently emerged zoonotic paramyxoviruses) M (matrix) protein-derived virus-like particles that bud from GSL-enriched plasma membrane microdomains was also dependent on interactions between virion-incorporated GSLs and CD169. Moreover, capture and transfer of Nipah virus envelope glycoprotein-pseudotyped lentivirus particles by MDCs were severely attenuated upon depletion of GSLs from virus particles. These results suggest that GSL incorporation into virions is critical for the interaction of diverse enveloped RNA viruses with DCs and that the GSL-CD169 recognition nexus might be a conserved viral mechanism of parasitization of DC functions for systemic virus dissemination. IMPORTANCE: Dendritic cells (DCs) can capture HIV-1 particles and transfer captured virus particles to T cells without establishing productive infection in DCs, a mechanism of HIV-1 trans infection. We have recently identified CD169-mediated recognition of GM3, a host-derived glycosphingolipid (GSL) incorporated into the virus particle membrane, as the receptor and ligand for the DC-HIV trans infection pathway. In this study, we have identified the matrix (MA) domain of Gag to be the viral determinant that governs incorporation of GM3 into HIV-1 particles, a previously unappreciated function of the HIV-1 MA. In addition, we demonstrate that the GSL-CD169-dependent trans infection pathway is also utilized as a dissemination mechanism by henipaviruses. GSL incorporation in henipaviruses was also dependent on the viral capsid (M) protein-directed assembly and budding from GSL-enriched lipid microdomains. These findings provide evidence of a conserved mechanism of retrovirus and henipavirus parasitization of cell-to-cell recognition pathways for systemic virus dissemination.


Assuntos
Células Dendríticas/imunologia , Glicoesfingolipídeos/imunologia , HIV-1/imunologia , Henipavirus/imunologia , Vírion/imunologia , Liberação de Vírus/imunologia , Linhagem Celular , Infecções por HIV/imunologia , Infecções por Henipavirus , Humanos , Microdomínios da Membrana/imunologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Montagem de Vírus/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia
16.
Immunity ; 40(3): 400-13, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24631155

RESUMO

The contribution of different DC subsets to effector and memory CD8(+) T cell generation during infection and the mechanism by which DCs controls these fate decisions is unclear. Here we demonstrated that the CD103(+) and CD11b(hi) migratory respiratory DC (RDC) subsets after influenza virus infection activated naive virus-specific CD8(+) T cells differentially. CD103(+) RDCs supported the generation of CD8(+) T effector (Teff) cells, which migrate from lymph nodes to the infected lungs. In contrast, migrant CD11b(hi) RDCs activated CD8(+) T cells characteristic of central memory CD8(+) T (CD8(+) Tcm) cells including retention within the draining lymph nodes. CD103(+) RDCs expressed CD24 at an elevated level, contributing to the propensity of this DC subpopulation to support CD8(+) Teff cell differentiation. Mechanistically, CD24 was shown to regulate CD8(+) T cell activation through HMGB1-mediated engagement of T cell RAGE. Thus, there is distribution of labor among DC subsets in regulating CD8(+) T cell differentiation.


Assuntos
Antígeno CD24/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Memória Imunológica , Animais , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/virologia , Feminino , Imunofenotipagem , Cadeias alfa de Integrinas/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/virologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Fenótipo , Ligação Proteica , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Liberação de Vírus/imunologia
17.
J Mol Biol ; 425(24): 4995-5008, 2013 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24095857

RESUMO

Post-translational protein modification is an important strategy for the regulation of the cell proteome independent of the need for new gene expression. Ubiquitin and ubiquitin-like modifiers mediate the regulation of protein levels, signaling pathways, vesicular trafficking, and many other cellular processes through their covalent conjugation to proteins. Interferon stimulated gene 15 (ISG15) is a ubiquitin-like modifier induced by type I interferon. In addition to conjugating to potentially hundreds of target proteins, ISG15 can be found in an unconjugated form both inside of the cell and released from interferon stimulated cells into the extracellular environment. Due to its robust expression after type I interferon stimulation and the broad panel of proteins that it targets, ISG15 has drawn much attention as a potential regulator of the immune response and has been shown to mediate protection in a number of different viral infection models. Here we will review the current state of the field of ISG15, the viruses against which ISG15 mediates protection, and the mechanisms by which ISG15 exerts antiviral activity.


Assuntos
Antivirais/imunologia , Citocinas/imunologia , Imunidade Inata , Interferon Tipo I/metabolismo , Processamento de Proteína Pós-Traducional , Ubiquitinas/imunologia , Antivirais/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Ubiquitinas/genética , Ubiquitinas/metabolismo , Liberação de Vírus/imunologia , Replicação Viral/imunologia
18.
J Exp Med ; 210(12): 2523-38, 2013 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-24145510

RESUMO

HIV-1-infected macrophages likely represent viral reservoirs, as they accumulate newly formed virions in internal virus-containing compartments (VCCs). However, the nature and biogenesis of VCCs remain poorly defined. We show that upon HIV-1 infection of primary human macrophages, Gag is recruited to preexisting compartments containing the scavenger receptor CD36, which then become VCCs. Silencing of CD36 in HIV-1-infected macrophages decreases the amount of virions released. Strikingly, soluble anti-CD36 antibodies, but not the natural ligands of CD36, inhibit release of virions from HIV-1-infected macrophages and the transmission of virus to CD4(+) T cells. The effect of the antibodies is potent, rapid, and induces the retention of virions within VCCs. Ectopic expression of CD36 in HeLa cells renders them susceptible to the inhibitory effect of the anti-CD36 mAb upon HIV-1 infection. We show that the anti-CD36 mAb inhibits HIV-1 release by clustering newly formed virions at their site of budding, and that signaling via CD36 is not required. Thus, HIV-1 reservoirs in macrophages may be tackled therapeutically using anti-CD36 antibodies to prevent viral dissemination.


Assuntos
Antígenos CD36/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Sequência de Aminoácidos , Anticorpos Biespecíficos , Anticorpos Bloqueadores , Anticorpos Monoclonais , Especificidade de Anticorpos , Antígenos CD36/antagonistas & inibidores , Antígenos CD36/genética , Células Cultivadas , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/transmissão , HIV-1/patogenicidade , HIV-1/fisiologia , Células HeLa , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Dados de Sequência Molecular , Vírion/imunologia , Vírion/patogenicidade , Vírion/fisiologia , Montagem de Vírus/imunologia , Liberação de Vírus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA