Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell ; 178(4): 1004-1015.e14, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398326

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Células Germinativas/inmunología , Fiebre de Lassa/inmunología , Virus Lassa/inmunología , Glicoproteínas de Membrana/química , Proteínas del Envoltorio Viral/química , Animales , Antígenos Virales/inmunología , Chlorocebus aethiops , Drosophila/citología , Epítopos/química , Epítopos/inmunología , Células HEK293 , Humanos , Fiebre de Lassa/virología , Glicoproteínas de Membrana/inmunología , Estructura Secundaria de Proteína , Células Vero , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología
2.
PLoS Pathog ; 8(9): e1002916, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028316

RESUMEN

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.


Asunto(s)
Evasión Inmune , Interferones/antagonistas & inhibidores , Marburgvirus/química , Marburgvirus/inmunología , ARN Bicatenario/metabolismo , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/metabolismo , Línea Celular , Cristalografía por Rayos X , Ebolavirus/química , Ebolavirus/genética , Ebolavirus/inmunología , Ebolavirus/metabolismo , Células HEK293 , Humanos , Marburgvirus/genética , Marburgvirus/metabolismo , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , ARN Bicatenario/química , Proteínas de Unión al ARN/metabolismo
3.
Proc Natl Acad Sci U S A ; 108(6): 2396-401, 2011 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-21262835

RESUMEN

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.


Asunto(s)
Exorribonucleasas/química , Virus Lassa/enzimología , Nucleoproteínas/química , ARN Bicatenario/química , Proteínas Virales/química , Línea Celular , Cristalografía por Rayos X , Exorribonucleasas/inmunología , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/inmunología , Virus Lassa/inmunología , Nucleoproteínas/inmunología , Estructura Terciaria de Proteína , ARN Bicatenario/inmunología , Proteínas Virales/inmunología
4.
Proc Natl Acad Sci U S A ; 108(48): 19365-70, 2011 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-22084115

RESUMEN

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.


Asunto(s)
Virus Lassa/química , Modelos Moleculares , Conformación Proteica , ARN Viral/metabolismo , Ribonucleoproteínas/química , Proteínas Virales/química , Línea Celular , Cristalización , Ensayo de Inmunoadsorción Enzimática , Humanos , Ribonucleoproteínas/metabolismo , Proteínas Virales/metabolismo
5.
Cell Rep ; 42(5): 112421, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37083327

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes , COVID-19 , Animales , Ratones , SARS-CoV-2 , Anticuerpos Antivirales/uso terapéutico , Anticuerpos ampliamente neutralizantes
6.
bioRxiv ; 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38168261

RESUMEN

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.

7.
mBio ; 13(4): e0127822, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35730904

RESUMEN

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.


Asunto(s)
Fiebre de Lassa , Virus Lassa , Anticuerpos Neutralizantes , Microscopía por Crioelectrón , Humanos , Virus Lassa/genética , Internalización del Virus
8.
Cell Rep ; 39(8): 110841, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35613585

RESUMEN

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.


Asunto(s)
Fiebre de Lassa , Virus Lassa , Anticuerpos Neutralizantes , Epítopos , Glicoproteínas/metabolismo , Humanos , Fiebre de Lassa/prevención & control , Virus Lassa/metabolismo , Proteínas del Envoltorio Viral
9.
Science ; 356(6341): 923-928, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28572385

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/metabolismo , Virus Lassa/fisiología , Modelos Moleculares , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Anticuerpos Antivirales/química , Anticuerpos Antivirales/metabolismo , Cristalización , Epítopos/química , Humanos , Concentración de Iones de Hidrógeno , Fiebre de Lassa/inmunología , Fiebre de Lassa/virología , Virus Lassa/química , Virus Lassa/inmunología , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Internalización del Virus
10.
Nat Struct Mol Biol ; 23(6): 513-521, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27111888

RESUMEN

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.


Asunto(s)
Virus de la Coriomeningitis Linfocítica/química , Glicoproteínas de Membrana/química , Proteínas del Envoltorio Viral/química , Animales , Línea Celular , Cristalografía por Rayos X , Drosophila , Glicosilación , Humanos , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Señales de Clasificación de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
11.
Nat Commun ; 7: 11544, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27161536

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Virus Lassa/inmunología , Especificidad de Anticuerpos , Antígenos Virales/química , Antígenos Virales/genética , Antígenos Virales/inmunología , Arenavirus/inmunología , Reacciones Cruzadas , Mapeo Epitopo , Epítopos/química , Epítopos/genética , Epítopos/inmunología , Humanos , Fiebre de Lassa/inmunología , Fiebre de Lassa/prevención & control , Virus Lassa/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Eliminación de Secuencia , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
12.
Neuropsychopharmacology ; 30(2): 350-9, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15483561

RESUMEN

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.


Asunto(s)
Trastornos Relacionados con Anfetaminas/psicología , Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Metanfetamina/farmacología , Trastornos Relacionados con Anfetaminas/orina , Animales , Temperatura Corporal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/administración & dosificación , Estimulantes del Sistema Nervioso Central/orina , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Hidrocortisona/orina , Macaca mulatta , Masculino , Metanfetamina/administración & dosificación , Metanfetamina/orina , Síndrome de Abstinencia a Sustancias/psicología
13.
J Neuroimmunol ; 157(1-2): 81-92, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15579284

RESUMEN

The virus/host interactions during the acute phase of human immunodeficiency virus (HIV) infection help determine the course of disease. During this time period, virus enters the brain. Here, we report clusters of genes whose transcripts are significantly upregulated in the frontal lobe of the brain during acute simian immunodeficiency virus (SIV) infection of rhesus monkeys. Many of these genes are involved in interferon (IFN) and/or interleukin (IL)-6 pathways. Although neither IFNalpha nor IFNgamma are elevated in the brain, IL6 is increased. Both IFNalpha and IL6 are elevated in plasma during this acute phase. The upregulation of STAT1, verified by immunohistochemical staining, can be due to both central nervous system (CNS) (SIV and IL6) and peripheral (IFNalpha and IL6) causes, and can itself drive the expression of many of these genes. Examination of the levels of expression of the upregulated genes in the post-acute and long-term phases of infection, as well as in SIV encephalitis, reveals increased expression throughout SIV infection, which may serve to protect the brain, but can have untoward long-term consequences.


Asunto(s)
Encéfalo/virología , Interferones/metabolismo , Interleucina-6/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología , Análisis de Varianza , Animales , Encéfalo/metabolismo , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Inmunohistoquímica/métodos , Interferones/genética , Interleucina-6/genética , Macaca mulatta , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factor de Transcripción STAT1 , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Factores de Tiempo , Transactivadores/metabolismo , Regulación hacia Arriba , Carga Viral/métodos
14.
PLoS One ; 7(8): e44211, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22937163

RESUMEN

Lassa virus causes hemorrhagic fever characterized by immunosuppression. The nucleoprotein of Lassa virus, termed NP, binds the viral genome. It also has an additional enzymatic activity as an exonuclease that specifically digests double-stranded RNA (dsRNA). dsRNA is a strong signal to the innate immune system of viral infection. Digestion of dsRNA by the NP exonuclease activity appears to cause suppression of innate immune signaling in the infected cell. Although the fold of the NP enzyme is conserved and the active site completely conserved with other exonucleases in its DEDDh family, NP is atypical among exonucleases in its preference for dsRNA and its strict specificity for one substrate. Here, we present the crystal structure of Lassa virus NP in complex with dsRNA. We find that unlike the exonuclease in Klenow fragment, the double-stranded nucleic acid in complex with Lassa NP remains base-paired instead of splitting, and that binding of the paired complementary strand is achieved by "relocation" of a basic loop motif from its typical exonuclease position. Further, we find that just one single glycine that contacts the substrate strand and one single tyrosine that stacks with a base of the complementary, non-substrate strand are responsible for the unique substrate specificity. This work thus provides templates for development of antiviral drugs that would be specific for viral, rather than host exonucleases of similar fold and active site, and illustrates how a very few amino acid changes confer alternate specificity and biological phenotype to an enzyme.


Asunto(s)
Virus Lassa/genética , Nucleoproteínas/genética , ARN Bicatenario/genética , ARN Viral/genética , Fiebre de Lassa/genética , Fiebre de Lassa/metabolismo , Virus Lassa/metabolismo , Nucleoproteínas/metabolismo , Conformación Proteica , ARN Bicatenario/metabolismo , ARN Viral/metabolismo
15.
Am J Pathol ; 162(6): 2041-57, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12759259

RESUMEN

The etiology of the central nervous system (CNS) alterations after human immunodeficiency virus (HIV) infection, such as dementia and encephalitis, remains unknown. We have used microarray analysis in a monkey model of neuroAIDS to identify 98 genes, many previously unrecognized in lentiviral CNS pathogenesis, whose expression is significantly up-regulated in the frontal lobe of simian immunodeficiency virus-infected brains. Further, through immunohistochemical illumination, distinct classes of genes were found whose protein products localized to infiltrating macrophages, endothelial cells and resident glia, such as CD163, Glut5, and ISG15. In addition we found proteins induced in cortical neurons (ie, cyclin D3, tissue transglutaminase, alpha1-antichymotrypsin, and STAT1), which have not previously been described as participating in simian immunodeficiency virus or HIV-related CNS pathology. This molecular phenotyping in the infected brains revealed pathways promoting entry of macrophages into the brain and their subsequent detrimental effects on neurons. These data support the hypothesis that in HIV-induced CNS disease products of activated macrophages and astrocytes lead to CNS dysfunction by directly damaging neurons, as well as by induction of altered gene and protein expression profiles in neurons themselves which are deleterious to their function.


Asunto(s)
Enfermedades del Sistema Nervioso Central/patología , Perfilación de la Expresión Génica , Macrófagos/metabolismo , Neuronas/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Ciclo Celular/genética , Diferenciación Celular/genética , División Celular/genética , Movimiento Celular , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/metabolismo , Citoesqueleto/genética , Regulación de la Expresión Génica/efectos de los fármacos , Sistema Inmunológico/metabolismo , Inmunohistoquímica , Hibridación in Situ , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Interferones/farmacología , Macaca mulatta , Macrófagos/virología , Microglía/metabolismo , Microglía/virología , Monocitos/patología , Neuronas/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas/genética , ARN/genética , ARN/metabolismo , Transducción de Señal/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Transcripción Genética
16.
J Neurovirol ; 10 Suppl 1: 58-66, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14982741

RESUMEN

The duration and severity of the symptomatology present during the early phase of human immunodeficiency virus (HIV) infection (known as the acute retroviral syndrome) is associated with alterations in the clinical profile of infection, such as a shortening of duration between infection with HIV and the onset of neurocognitive impairment and acquired immunodeficiency syndrome (AIDS). Viral-specific CD8+ cytotoxic T lymphocytes (CTLs) and CD8+ natural killer (NK) cells play a key role in antiviral immunity. Loss of CD8+ cells or their functional impairment during the early period of infection is associated with a rapid progression to AIDS in nonhuman primate studies. However, no studies have determined whether CD8+ cell loss or impairment is associated with symptoms of acute retroviral illness such as fever. In this study, the authors compared the early phase of simian immunodeficiency virus (SIV) infection in animals that were treated with the anti-CD8 monoclonal antibody cM-T807 to deplete CD8+ cells during the early period of infection (SIV+ CD8- group) to those with intact CD8+ cells (SIV+ CD8+ group). The SIV+ CD8- group had an enhanced acute retroviral syndrome when compared to the SIV+ CD8+ group. The SIV+ CD8- group also had prolonged high viral loads and distinct alterations in the proinflammatory cytokines interleukin (IL)-6 and interferon (IFN)-alpha, as well as in monocyte chemoattractant protein (MCP)-1. CD8+ cell depletion, therefore, appears to enhance symptoms of the acute retroviral syndrome and alters several of the immunological factors associated with the early phase of infection.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/fisiopatología , Enfermedad Aguda , Animales , Quimiocina CCL2/sangre , Fiebre/inmunología , Fiebre/mortalidad , Fiebre/virología , Interferón-alfa/sangre , Interleucina-6/sangre , Macaca mulatta , Masculino , Actividad Motora , Síndrome de Inmunodeficiencia Adquirida del Simio/mortalidad , Análisis de Supervivencia , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA