Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell ; 178(4): 1004-1015.e14, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31398326

RESUMO

Lassa virus (LASV) causes hemorrhagic fever and is endemic in West Africa. Protective antibody responses primarily target the LASV surface glycoprotein (GPC), and GPC-B competition group antibodies often show potent neutralizing activity in humans. However, which features confer potent and broadly neutralizing antibody responses is unclear. Here, we compared three crystal structures of LASV GPC complexed with GPC-B antibodies of varying neutralization potency. Each GPC-B antibody recognized an overlapping epitope involved in binding of two adjacent GPC monomers and preserved the prefusion trimeric conformation. Differences among GPC-antibody interactions highlighted specific residues that enhance neutralization. Using structure-guided amino acid substitutions, we increased the neutralization potency and breadth of these antibodies to include all major LASV lineages. The ability to define antibody residues that allow potent and broad neutralizing activity, together with findings from analyses of inferred germline precursors, is critical to develop potent therapeutics and for vaccine design and assessment.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Células Germinativas/imunologia , Febre Lassa/imunologia , Vírus Lassa/imunologia , Glicoproteínas de Membrana/química , Proteínas do Envelope Viral/química , Animais , Antígenos Virais/imunologia , Chlorocebus aethiops , Drosophila/citologia , Epitopos/química , Epitopos/imunologia , Células HEK293 , Humanos , Febre Lassa/virologia , Glicoproteínas de Membrana/imunologia , Estrutura Secundária de Proteína , Células Vero , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia
2.
J Virol ; 90(9): 4556-62, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26912609

RESUMO

UNLABELLED: The arenavirus matrix protein Z is highly multifunctional and occurs in both monomeric and oligomeric forms. The crystal structure of a dodecamer of Z from Lassa virus, presented here, illustrates a ring-like structure with a highly basic center. Mutagenesis demonstrates that the dimeric interface within the dodecamer and a Lys-Trp-Lys triad at the center of the ring are important for oligomerization. This structure provides an additional template to explore the many functions of Z. IMPORTANCE: The arenavirus Lassa virus causes hundreds of thousands of infections each year, many of which develop into fatal hemorrhagic fever. The arenavirus matrix protein Z is multifunctional, with at least four distinct roles. Z exists in both monomeric and oligomeric forms, each of which likely serves a specific function in the viral life cycle. Here we present the dodecameric form of Lassa virus Z and demonstrate that Z forms a "wreath" with a highly basic center. This structure and that of monomeric Z now provide a pair of critical templates by which the multiple roles of Z in the viral life cycle may be interpreted.


Assuntos
Proteínas de Transporte/química , Vírus Lassa , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Proteínas da Matriz Viral/química , Cristalografia por Raios X , Espectrometria de Massas , Ressonância Magnética Nuclear Biomolecular , Proteínas de Ligação a RNA , Proteínas Recombinantes de Fusão/química , Relação Estrutura-Atividade
3.
PLoS Pathog ; 8(9): e1002916, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23028316

RESUMO

Filoviruses, including Marburg virus (MARV) and Ebola virus (EBOV), cause fatal hemorrhagic fever in humans and non-human primates. All filoviruses encode a unique multi-functional protein termed VP35. The C-terminal double-stranded (ds)RNA-binding domain (RBD) of VP35 has been implicated in interferon antagonism and immune evasion. Crystal structures of the VP35 RBD from two ebolaviruses have previously demonstrated that the viral protein caps the ends of dsRNA. However, it is not yet understood how the expanses of dsRNA backbone, between the ends, are masked from immune surveillance during filovirus infection. Here, we report the crystal structure of MARV VP35 RBD bound to dsRNA. In the crystal structure, molecules of dsRNA stack end-to-end to form a pseudo-continuous oligonucleotide. This oligonucleotide is continuously and completely coated along its sugar-phosphate backbone by the MARV VP35 RBD. Analysis of dsRNA binding by dot-blot and isothermal titration calorimetry reveals that multiple copies of MARV VP35 RBD can indeed bind the dsRNA sugar-phosphate backbone in a cooperative manner in solution. Further, MARV VP35 RBD can also cap the ends of the dsRNA in solution, although this arrangement was not captured in crystals. Together, these studies suggest that MARV VP35 can both coat the backbone and cap the ends, and that for MARV, coating of the dsRNA backbone may be an essential mechanism by which dsRNA is masked from backbone-sensing immune surveillance molecules.


Assuntos
Evasão da Resposta Imune , Interferons/antagonistas & inibidores , Marburgvirus/química , Marburgvirus/imunologia , RNA de Cadeia Dupla/metabolismo , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/metabolismo , Linhagem Celular , Cristalografia por Raios X , Ebolavirus/química , Ebolavirus/genética , Ebolavirus/imunologia , Ebolavirus/metabolismo , Células HEK293 , Humanos , Marburgvirus/genética , Marburgvirus/metabolismo , Modelos Moleculares , Ligação Proteica , Estrutura Terciária de Proteína , RNA de Cadeia Dupla/química , Proteínas de Ligação a RNA/metabolismo
4.
Proc Natl Acad Sci U S A ; 108(6): 2396-401, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21262835

RESUMO

Lassa fever virus, a member of the family Arenaviridae, is a highly endemic category A pathogen that causes 300,000-500,000 infections per year in Western Africa. The arenaviral nucleoprotein NP has been implicated in suppression of the host innate immune system, but the mechanism by which this occurs has remained elusive. Here we present the crystal structure at 1.5 Å of the immunosuppressive C-terminal portion of Lassa virus NP and illustrate that, unexpectedly, its 3D fold closely mimics that of the DEDDh family of exonucleases. Accompanying biochemical experiments illustrate that NP indeed has a previously unknown, bona fide exonuclease activity, with strict specificity for double-stranded RNA substrates. We further demonstrate that this exonuclease activity is essential for the ability of NP to suppress translocation of IFN regulatory factor 3 and block activation of the innate immune system. Thus, the nucleoprotein is a viral exonuclease with anti-immune activity, and this work provides a unique opportunity to combat arenaviral infections.


Assuntos
Exorribonucleases/química , Vírus Lassa/enzimologia , Nucleoproteínas/química , RNA de Cadeia Dupla/química , Proteínas Virais/química , Linhagem Celular , Cristalografia por Raios X , Exorribonucleases/imunologia , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/imunologia , Vírus Lassa/imunologia , Nucleoproteínas/imunologia , Estrutura Terciária de Proteína , RNA de Cadeia Dupla/imunologia , Proteínas Virais/imunologia
5.
Proc Natl Acad Sci U S A ; 108(48): 19365-70, 2011 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-22084115

RESUMO

Arenaviruses cause disease in industrialized and developing nations alike. Among them, the hemorrhagic fever virus Lassa is responsible for ~300,000-500,000 infections/y in Western Africa. The arenavirus nucleoprotein (NP) forms the protein scaffold of the genomic ribonucleoprotein complexes and is critical for transcription and replication of the viral genome. Here, we present crystal structures of the RNA-binding domain of Lassa virus NP in complex with ssRNA. This structure shows, in contrast to the predicted model, that RNA binds in a deep, basic crevice located entirely within the N-terminal domain. Furthermore, the NP-ssRNA structures presented here, combined with hydrogen-deuterium exchange/MS and functional studies, suggest a gating mechanism by which NP opens to accept RNA. Directed mutagenesis and functional studies provide a unique look into how the arenavirus NPs bind to and protect the viral genome and also suggest the likely assembly by which viral ribonucleoprotein complexes are organized.


Assuntos
Vírus Lassa/química , Modelos Moleculares , Conformação Proteica , RNA Viral/metabolismo , Ribonucleoproteínas/química , Proteínas Virais/química , Linhagem Celular , Cristalização , Ensaio de Imunoadsorção Enzimática , Humanos , Ribonucleoproteínas/metabolismo , Proteínas Virais/metabolismo
6.
Cell Rep ; 42(5): 112421, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37083327

RESUMO

Therapeutic antibodies are an important tool in the arsenal against coronavirus infection. However, most antibodies developed early in the pandemic have lost most or all efficacy against newly emergent strains of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), particularly those of the Omicron lineage. Here, we report the identification of a panel of vaccinee-derived antibodies that have broad-spectrum neutralization activity. Structural and biochemical characterization of the three broadest-spectrum antibodies reveal complementary footprints and differing requirements for avidity to overcome variant-associated mutations in their binding footprints. In the K18 mouse model of infection, these three antibodies exhibit protective efficacy against BA.1 and BA.2 infection. This study highlights the resilience and vulnerabilities of SARS-CoV-2 antibodies and provides road maps for further development of broad-spectrum therapeutics.


Assuntos
Anticorpos Neutralizantes , COVID-19 , Animais , Camundongos , SARS-CoV-2 , Anticorpos Antivirais/uso terapêutico , Anticorpos Amplamente Neutralizantes
7.
bioRxiv ; 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38168261

RESUMO

The 800 million human infections with SARS-CoV-2 and the likely emergence of new variants and additional coronaviruses necessitate a better understanding of the essential spike glycoprotein and the development of immunogens that foster broader and more durable immunity. The S2 fusion subunit is more conserved in sequence, is essential to function, and would be a desirable immunogen to boost broadly reactive antibodies. It is, however, unstable in structure and in its wild-type form, cannot be expressed alone without irreversible collapse into a six-helix bundle. In addition to the irreversible conformational changes of fusion, biophysical measurements indicate that spike also undergoes a reversible breathing action. However, spike in an open, "breathing" conformation has not yet been visualized at high resolution. Here we describe an S2-only antigen, engineered to remain in its relevant, pre-fusion viral surface conformation in the absence of S1. We also describe a panel of natural human antibodies specific for S2 from vaccinated and convalescent individuals. One of these mAbs, from a convalescent individual, afforded a high-resolution cryo-EM structure of the prefusion S2. The structure reveals a complex captured in an "open" conformation with greater stabilizing intermolecular interactions at the base and a repositioned fusion peptide. Together, this work provides an antigen for advancement of next-generation "booster" immunogens and illuminates the likely breathing adjustments of the coronavirus spike.

8.
Antiviral Res ; 212: 105580, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36940916

RESUMO

Although severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) initially infects the respiratory tract, it also directly or indirectly affects other organs, including the brain. However, little is known about the relative neurotropism of SARS-CoV-2 variants of concern (VOCs), including Omicron (B.1.1.529), which emerged in November 2021 and has remained the dominant pathogenic lineage since then. To address this gap, we examined the relative ability of Omicron, Beta (B.1.351), and Delta (B.1.617.2) to infect the brain in the context of a functional human immune system by using human angiotensin-converting enzyme 2 (hACE2) knock-in triple-immunodeficient NGC mice with or without reconstitution with human CD34+ stem cells. Intranasal inoculation of huCD34+-hACE2-NCG mice with Beta and Delta resulted in productive infection of the nasal cavity, lungs, and brain on day 3 post-infection, but Omicron was surprisingly unique in its failure to infect either the nasal tissue or brain. Moreover, the same infection pattern was observed in hACE2-NCG mice, indicating that antiviral immunity was not responsible for the lack of Omicron neurotropism. In independent experiments, we demonstrate that nasal inoculation with Beta or with D614G, an ancestral SARS-CoV-2 with undetectable replication in huCD34+-hACE2-NCG mice, resulted in a robust response by human innate immune cells, T cells, and B cells, confirming that exposure to SARS-CoV-2, even without detectable infection, is sufficient to induce an antiviral immune response. Collectively, these results suggest that modeling of the neurologic and immunologic sequelae of SARS-CoV-2 infection requires careful selection of the appropriate SARS-CoV-2 strain in the context of a specific mouse model.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Humanos , Camundongos , Encéfalo , Antivirais , Modelos Animais de Doenças
9.
Cell Rep ; 39(8): 110841, 2022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35613585

RESUMO

Lassa virus (LASV) is the etiologic agent of Lassa Fever, a hemorrhagic disease that is endemic to West Africa. During LASV infection, LASV glycoprotein (GP) engages with multiple host receptors for cell entry. Neutralizing antibodies against GP are rare and principally target quaternary epitopes displayed only on the metastable, pre-fusion conformation of GP. Currently, the structural features of the neutralizing GPC-A antibody competition group are understudied. Structures of two GPC-A antibodies presented here demonstrate that they bind the side of the pre-fusion GP trimer, bridging the GP1 and GP2 subunits. Complementary biochemical analyses indicate that antibody 25.10C, which is broadly specific, neutralizes by inhibiting binding of the endosomal receptor LAMP1 and also by blocking membrane fusion. The other GPC-A antibody, 36.1F, which is lineage-specific, prevents LAMP1 association only. These data illuminate a site of vulnerability on LASV GP and will guide efforts to elicit broadly reactive therapeutics and vaccines.


Assuntos
Febre Lassa , Vírus Lassa , Anticorpos Neutralizantes , Epitopos , Glicoproteínas/metabolismo , Humanos , Febre Lassa/prevenção & controle , Vírus Lassa/metabolismo , Proteínas do Envelope Viral
10.
mBio ; 13(4): e0127822, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35730904

RESUMO

Lassa virus (LASV) is the causative agent of the deadly Lassa fever (LF). Seven distinct LASV lineages circulate through western Africa, among which lineage I (LI), the first to be identified, is particularly resistant to antibody neutralization. Lineage I LASV evades neutralization by half of known antibodies in the GPC-A antibody competition group and all but one of the antibodies in the GPC-B competition group. Here, we solve two cryo-electron microscopy (cryo-EM) structures of LI GP in complex with a GPC-A and a GPC-B antibody. We used complementary structural and biochemical techniques to identify single-amino-acid substitutions in LI that are responsible for immune evasion by each antibody group. Further, we show that LI infection is more dependent on the endosomal receptor lysosome-associated membrane protein 1 (LAMP1) for viral entry relative to LIV. In the absence of LAMP1, LI requires a more acidic fusion pH to initiate membrane fusion with the host cell relative to LIV. IMPORTANCE No vaccine or therapeutics are approved to prevent LASV infection or treat LF. All vaccine platforms currently under development present only the LIV GP sequence. However, our data suggest that the high genetic diversity of LASV may be problematic for designing both a broadly reactive immunogen and therapeutic. Here, we examine antibodies that are highly potent against LIV yet are ineffective against LI. By pinpointing LI mutations responsible for this decrease in antibody efficacy, we suggest that future vaccine platforms may need to incorporate specific LI-like mutations in order to generate a broadly neutralizing antibody response against all LASV lineages.


Assuntos
Febre Lassa , Vírus Lassa , Anticorpos Neutralizantes , Microscopia Crioeletrônica , Humanos , Vírus Lassa/genética , Internalização do Vírus
11.
Sci Transl Med ; 14(668): eabq0991, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36288283

RESUMO

Developing potent therapeutics and effective vaccines are the ultimate goals in controlling infectious diseases. Lassa virus (LASV), the causative pathogen of Lassa fever (LF), infects hundreds of thousands annually, but effective antivirals or vaccines against LASV infection are still lacking. Furthermore, neutralizing antibodies against LASV are rare. Here, we describe biochemical analyses and high-resolution cryo-electron microscopy structures of a therapeutic cocktail of three broadly protective antibodies that target the LASV glycoprotein complex (GPC), previously identified from survivors of multiple LASV infections. Structural and mechanistic analyses reveal compatible neutralizing epitopes and complementary neutralization mechanisms that offer high potency, broad range, and resistance to escape. These antibodies either circumvent or exploit specific glycans comprising the extensive glycan shield of GPC. Further, they require mammalian glycosylation, native GPC cleavage, and proper GPC trimerization. These findings guided engineering of a next-generation GPC antigen suitable for future neutralizing antibody and vaccine discovery. Together, these results explain protective mechanisms of rare, broad, and potent antibodies and identify a strategy for the rational design of therapeutic modalities against LF and related infectious diseases.


Assuntos
Febre Lassa , Vacinas Virais , Animais , Humanos , Vírus Lassa , Microscopia Crioeletrônica , Anticorpos Neutralizantes , Epitopos , Glicoproteínas , Polissacarídeos , Antivirais , Mamíferos
12.
Am J Pathol ; 177(1): 355-61, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20489154

RESUMO

Methamphetamine (Meth) abuse increases risky behaviors that contribute to the spread of HIV infection. In addition, because HIV and Meth independently affect physiological systems including the central nervous system, HIV-induced disease may be more severe in drug users. We investigated changes in blood and brain viral load as well as differences in immune cells in chronically simian immunodeficiency virus-infected rhesus macaques that were either administered Meth or used as controls. Although Meth administration did not alter levels of virus in the plasma, viral load in the brain was significantly increased in Meth-treated animals compared with control animals. Meth treatment also resulted in an activation of natural killer cells. Given the prevalence of Meth use in HIV-infected and HIV at-risk populations, these findings reveal the likely untoward effects of Meth abuse in such individuals.


Assuntos
Encéfalo , Células Matadoras Naturais , Macaca mulatta , Metanfetamina/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia/imunologia , Carga Viral , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , HIV/efeitos dos fármacos , Infecções por HIV/virologia , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Macaca mulatta/imunologia , Macaca mulatta/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Transtornos Relacionados ao Uso de Substâncias
13.
Science ; 374(6566): 472-478, 2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34554826

RESUMO

Antibody-based therapeutics and vaccines are essential to combat COVID-19 morbidity and mortality after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Multiple mutations in SARS-CoV-2 that could impair antibody defenses propagated in human-to-human transmission and spillover or spillback events between humans and animals. To develop prevention and therapeutic strategies, we formed an international consortium to map the epitope landscape on the SARS-CoV-2 spike protein, defining and structurally illustrating seven receptor binding domain (RBD)­directed antibody communities with distinct footprints and competition profiles. Pseudovirion-based neutralization assays reveal spike mutations, individually and clustered together in variants, that affect antibody function among the communities. Key classes of RBD-targeted antibodies maintain neutralization activity against these emerging SARS-CoV-2 variants. These results provide a framework for selecting antibody treatment cocktails and understanding how viral variants might affect antibody therapeutic efficacy.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Mapeamento de Epitopos , Epitopos Imunodominantes/imunologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Antígenos Virais/química , Antígenos Virais/imunologia , COVID-19/terapia , Humanos , Epitopos Imunodominantes/química , Ligação Proteica , Domínios Proteicos , Glicoproteína da Espícula de Coronavírus/química
14.
J Virol ; 82(3): 1615-21, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18045942

RESUMO

Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infect and productively replicate in macrophages and T lymphocytes. Here, we show that SIV virions derived from macrophages have higher levels of infectivity than those derived from T cells. The lower infectivity of T-cell-derived viruses is influenced by the quantity or type of mannose residues on the virion. Our results demonstrate that the cellular origin of a virus is a major factor in viral infectivity. Cell-type-specific factors in viral infectivity, and organ-specific or disease stage-specific differences in cellular derivation of virions, can be critical in the pathogenesis of HIV and AIDS.


Assuntos
Linfócitos T CD4-Positivos/virologia , Macrófagos/virologia , Vírus da Imunodeficiência Símia/patogenicidade , Vírion/química , Animais , Células Cultivadas , Macaca mulatta , Manose/análise , Vírus da Imunodeficiência Símia/química , Virulência
15.
Alcohol Clin Exp Res ; 32(9): 1583-92, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18616669

RESUMO

BACKGROUND: Alcohol consumption is a common problem in HIV-infected individuals, and the effects of alcohol may alter the efficiency of the immune response, potentially aggravating the disease as well as affecting end organs, such as the brain. However, the elements of the virus-host interaction that are modulated by ethanol are poorly dissected. METHODS: Ethanol intake was conditioned in rhesus macaques prior to SIV infection, in order to mimic this common human behavior, and allow the evaluation of aspects of the virus-immune system interactions during acute time-points, when important facets of the infection are set up and when virus reproducibly enters the brain. RESULTS: Although ethanol had a limited effect on the acute plasma viral load, it resulted in reduced circulating memory CD4(+) T cells and increased levels of monocytes expressing the viral coreceptor CCR5. In organs, ethanol consumption impacted immune cells in the liver as well as lymphoid and other nonlymphoid tissues, where CD4(+) T cells were predominantly affected. CONCLUSION: Overall, the consumption of alcohol causes immune cell alterations that can contribute to the generation of a disease susceptible environment upon SIV infection.


Assuntos
Consumo de Bebidas Alcoólicas/imunologia , Alcoolismo/imunologia , Sistema Imunitário/fisiopatologia , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Citocinas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças/imunologia , Células Matadoras Naturais/patologia , Fígado/patologia , Fígado/virologia , Macaca mulatta , Doenças dos Macacos/epidemiologia , Fatores de Risco , Síndrome de Imunodeficiência Adquirida dos Símios/epidemiologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Baço/patologia , Baço/virologia , Linfócitos T/patologia , Carga Viral
16.
Science ; 356(6341): 923-928, 2017 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-28572385

RESUMO

The arenavirus Lassa causes severe hemorrhagic fever and a significant disease burden in West Africa every year. The glycoprotein, GPC, is the sole antigen expressed on the viral surface and the critical target for antibody-mediated neutralization. Here we present the crystal structure of the trimeric, prefusion ectodomain of Lassa GP bound to a neutralizing antibody from a human survivor at 3.2-angstrom resolution. The antibody extensively anchors two monomers together at the base of the trimer, and biochemical analysis suggests that it neutralizes by inhibiting conformational changes required for entry. This work illuminates pH-driven conformational changes in both receptor-binding and fusion subunits of Lassa virus, illustrates the unique assembly of the arenavirus glycoprotein spike, and provides a much-needed template for vaccine design against these threats to global health.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/metabolismo , Vírus Lassa/fisiologia , Modelos Moleculares , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Anticorpos Antivirais/química , Anticorpos Antivirais/metabolismo , Cristalização , Epitopos/química , Humanos , Concentração de Íons de Hidrogênio , Febre Lassa/imunologia , Febre Lassa/virologia , Vírus Lassa/química , Vírus Lassa/imunologia , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Estabilidade Proteica , Estrutura Quaternária de Proteína , Internalização do Vírus
17.
Viral Immunol ; 19(4): 679-89, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17201663

RESUMO

Facets of the immune response early after human immunodeficiency virus (HIV) infection influence the course of disease. In the simian immunodeficiency virus (SIV)-rhesus monkey system, a global dysfunction of CD4(+) T cell cytokine secretion was reported to develop early after infection [McKay PF, Barouch DH, Schmitz JE, Veazey RS, Gorgone DA, Lifton MA, Williams KC, and Letvin NL: J Virol 2003;77:4695-4702]. Because differences have been found in SIV pathogenesis depending on the origin of the monkeys, we investigated the correlation between animal background, defined by country of origin (India or China), and circulating T cell cytokine secretion as well as cycling ability within the first 3 mo of SIV infection. An early loss of CD4(+) T cells that produce interferon (IFN)-gamma and interleukin (IL)-2, those that produce IFN-gamma but not tumor necrosis factor (TNF)-alpha, as well as those that do not express IFN-gamma but can express IL-2 or TNF-alpha, was observed in animals of Indian, but not of Chinese, origin after SIV infection. After infection CD4(+) T cells in Chinese macaques developed an increased proliferating pool of T cells compared with Indian animals. These data reveal host diversity in the global effects of SIV infection on functional subsets of immune cells, which can add to a better understanding of differences observed in populations from diverse ethnic origins.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Citocinas/biossíntese , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Animais , China , Índia , Interferon gama/biossíntese , Interleucina-2/biossíntese , Ativação Linfocitária , Macaca mulatta , Especificidade da Espécie , Fator de Necrose Tumoral alfa/biossíntese
18.
Nat Struct Mol Biol ; 23(6): 513-521, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27111888

RESUMO

Arenaviruses exist worldwide and can cause hemorrhagic fever and neurologic disease. A single glycoprotein expressed on the viral surface mediates entry into target cells. This glycoprotein, termed GPC, contains a membrane-associated signal peptide, a receptor-binding subunit termed GP1 and a fusion-mediating subunit termed GP2. Although GPC is a critical target of antibodies and vaccines, the structure of the metastable GP1-GP2 prefusion complex has remained elusive for all arenaviruses. Here we describe the crystal structure of the fully glycosylated prefusion GP1-GP2 complex of the prototypic arenavirus LCMV at 3.5 Å. This structure reveals the conformational changes that the arenavirus glycoprotein must undergo to cause fusion and illustrates the fusion regions and potential oligomeric states.


Assuntos
Vírus da Coriomeningite Linfocítica/química , Glicoproteínas de Membrana/química , Proteínas do Envelope Viral/química , Animais , Linhagem Celular , Cristalografia por Raios X , Drosophila , Glicosilação , Humanos , Coriomeningite Linfocítica/metabolismo , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/fisiologia , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Sinais Direcionadores de Proteínas , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus
19.
Nat Commun ; 7: 11544, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27161536

RESUMO

Lassa fever is a severe multisystem disease that often has haemorrhagic manifestations. The epitopes of the Lassa virus (LASV) surface glycoproteins recognized by naturally infected human hosts have not been identified or characterized. Here we have cloned 113 human monoclonal antibodies (mAbs) specific for LASV glycoproteins from memory B cells of Lassa fever survivors from West Africa. One-half bind the GP2 fusion subunit, one-fourth recognize the GP1 receptor-binding subunit and the remaining fourth are specific for the assembled glycoprotein complex, requiring both GP1 and GP2 subunits for recognition. Notably, of the 16 mAbs that neutralize LASV, 13 require the assembled glycoprotein complex for binding, while the remaining 3 require GP1 only. Compared with non-neutralizing mAbs, neutralizing mAbs have higher binding affinities and greater divergence from germline progenitors. Some mAbs potently neutralize all four LASV lineages. These insights from LASV human mAb characterization will guide strategies for immunotherapeutic development and vaccine design.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vírus Lassa/imunologia , Especificidade de Anticorpos , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Arenavirus/imunologia , Reações Cruzadas , Mapeamento de Epitopos , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Humanos , Febre Lassa/imunologia , Febre Lassa/prevenção & controle , Vírus Lassa/genética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Deleção de Sequência , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia
20.
Neuropsychopharmacology ; 30(2): 350-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15483561

RESUMO

Acute high dose methamphetamine (METH) dosing regimens are frequently used in animal studies, however, these regimens can lead to considerable toxicity and even death in experimental animals. Acute high dosing regimens are quite distinct from the chronic usage patterns found in many human METH abusers. Furthermore, such doses, especially in nonhuman primates, can result in unexpected death, which is unacceptable, especially when such deaths fail to accurately model effects of human usage. As a model of chronic human METH abuse we have developed a nonlethal chronic METH administration procedure for the rhesus macaque that utilizes an escalating dose protocol. This protocol slowly increases the METH dosage from 0.1 to 0.7 mg/kg b.i.d. over a period of 4 weeks, followed by a period of chronic METH administration at 0.75 mg/kg b.i.d. (= total daily METH administration of 1.5 mg/kg). In parallel to human usage patterns, METH injections were given 20-23 times a month. This regimen produced a number of behavioral and physiological effects including decreased food intake and a significant increase in urinary cortisol excretion.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/psicologia , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Metanfetamina/farmacologia , Transtornos Relacionados ao Uso de Anfetaminas/urina , Animais , Temperatura Corporal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/administração & dosagem , Estimulantes do Sistema Nervoso Central/urina , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Hidrocortisona/urina , Macaca mulatta , Masculino , Metanfetamina/administração & dosagem , Metanfetamina/urina , Síndrome de Abstinência a Substâncias/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA