Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Science ; 373(6559): 1109-1116, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34344823

RESUMEN

The spillovers of betacoronaviruses in humans and the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants highlight the need for broad coronavirus countermeasures. We describe five monoclonal antibodies (mAbs) cross-reacting with the stem helix of multiple betacoronavirus spike glycoproteins isolated from COVID-19 convalescent individuals. Using structural and functional studies, we show that the mAb with the greatest breadth (S2P6) neutralizes pseudotyped viruses from three different subgenera through the inhibition of membrane fusion, and we delineate the molecular basis for its cross-reactivity. S2P6 reduces viral burden in hamsters challenged with SARS-CoV-2 through viral neutralization and Fc-mediated effector functions. Stem helix antibodies are rare, oftentimes of narrow specificity, and can acquire neutralization breadth through somatic mutations. These data provide a framework for structure-guided design of pan-betacoronavirus vaccines eliciting broad protection.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Betacoronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas Virales/inmunología , Internalización del Virus , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/aislamiento & purificación , Convalecencia , Cricetinae , Reacciones Cruzadas , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Células Jurkat , Pulmón/inmunología , Fusión de Membrana/inmunología , Pruebas de Neutralización , Mapeo Peptídico , Conformación Proteica en Hélice alfa , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Carga Viral/inmunología
2.
Artículo en Inglés | MEDLINE | ID: mdl-32513673

RESUMEN

Hemagglutinins (HAs) are the receptor-binding and membrane fusion glycoproteins of influenza viruses. They recognize sialic acid-containing, cell-surface glycoconjugates as receptors but have limited affinity for them, and, as a consequence, virus attachment to cells requires their interaction with several virus HAs. Receptor-bound virus is transferred into endosomes where membrane fusion by HAs is activated at pH between 5 and 6.5, depending on the strain of virus. Fusion activity requires extensive rearrangements in HA conformation that include extrusion of a buried "fusion peptide" to connect with the endosomal membrane, form a bridge to the virus membrane, and eventually bring both membranes close together. In this review, we give an overview of the structures of the 16 genetically and antigenically distinct subtypes of influenza A HA in relation to these two functions in virus replication and in relation to recognition of HA by antibodies that neutralize infection.


Asunto(s)
Hemaglutininas/inmunología , Orthomyxoviridae/inmunología , Hemaglutininas/fisiología , Humanos , Concentración de Iones de Hidrógeno , Fusión de Membrana/inmunología
3.
J Virol ; 94(19)2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32699087

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Fiebre Chikungunya/inmunología , Virus Chikungunya/inmunología , Proteínas del Envoltorio Viral/inmunología , Internalización del Virus/efectos de los fármacos , Liberación del Virus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , Anticuerpos Antivirales/farmacología , Virus Chikungunya/genética , Virus Chikungunya/fisiología , Chlorocebus aethiops , Epítopos/inmunología , Femenino , Fusión de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Dominios y Motivos de Interacción de Proteínas , Células Vero , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/efectos de los fármacos , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/inmunología , Liberación del Virus/efectos de los fármacos
4.
J Immunol ; 201(5): 1421-1433, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30037848

RESUMEN

Phagosome maturation is an important innate defense mechanism of macrophages against bacterial infections. The mycobacterial secretory protein kinase G (PknG), a serine/threonine kinase, is known to block phagosome-lysosome (P-L) fusion, and the kinase activity of PknG appears to be crucial for this. However, the detail mechanisms are not well understood. In the current study, we demonstrate that PknG of Mycobacterium sp interacts with the human Rab GTPase protein, Rab7l1, but not with other Rab proteins as well as factors like Rabaptin, Rabex5, PI3K3, Mon1a, Mon1b, early endosome autoantigen 1, and LAMP2 that are known to play crucial roles in P-L fusion. The Rab7l1 protein is shown to play a role in P-L fusion during mycobacterial infection, and its absence promotes survival of bacilli inside macrophages. PknG was found to be translocated to the Golgi complex where it interacted with GDP-bound Rab7l1 and blocked transition of inactive Rab7l1-GDP to active Rab7l1-GTP, resulting in inhibition of recruitment of Rab7l1-GTP to bacilli-containing phagosomes, and these processes are dependent on the kinase activity of PknG. Localization of Rab7l1-GTP to phagosomes was found to be critical for the subsequent recruitment of other phago-lysosomal markers like early endosome autoantigen 1, Rab7, and LAMP2 during infection. Thus, by interfering with the Rab7l1 signaling process, PknG prevents P-L fusion and favors bacterial survival inside human macrophages. This study highlights a novel role of Rab7l1 in the phagosomal maturation process and hints at unique strategies of mycobacteria used to interfere with Rab7l1 function to favor its survival inside human macrophages.


Asunto(s)
Proteínas Bacterianas/inmunología , Proteínas Quinasas Dependientes de GMP Cíclico/inmunología , Lisosomas/inmunología , Macrófagos/inmunología , Mycobacterium/inmunología , Fagocitosis , Fagosomas/inmunología , Transducción de Señal/inmunología , Proteínas Bacterianas/genética , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Células HEK293 , Humanos , Lisosomas/genética , Lisosomas/microbiología , Macrófagos/microbiología , Macrófagos/patología , Fusión de Membrana/inmunología , Mycobacterium/genética , Fagosomas/genética , Fagosomas/microbiología , Fagosomas/patología , Células THP-1 , Proteínas de Unión al GTP rab , Proteínas de Unión al GTP rab1
5.
J Leukoc Biol ; 102(5): 1249-1259, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28877954

RESUMEN

Microorganisms are known to devise various strategies to thwart protective responses by the host. One such strategy is to incorporate sequences and domains in their genes/proteins that have similarity to various domains of the host proteins. In this study, we report that Mycobacterium tuberculosis protein Rv3529c exhibits significant similarity to the death domain of the TLR pathway adaptor protein MyD88. Incubation of macrophages with Rv3529c specifically inhibited TLR2-mediated proinflammatory responses. This included attenuated oxidative burst, reduced phosphorylation of MAPK-ERK, reduced activation of transcription factor NF-κB and reduced secretion of proinflammatory cytokines IFN-γ, IL-6, and IL-17A with a concomitant increased secretion of suppressor cytokines IL-10 and TGF-ß. Importantly, Rv3529c significantly inhibited TLR2-induced association of MyD88 with IRAK1 by competitively binding with IRAK1. Further, Rv3529c mediated inhibition of apoptosis and phagosome-lysosome fusion. Lastly, incubation of macrophages with Rv3529c increased bacterial burden inside macrophages. The data presented show another strategy evolved by M. tuberculosis toward immune evasion that centers on incorporating sequences in proteins that are similar to crucial proteins in the innate immune system of the host.


Asunto(s)
Proteínas Bacterianas/farmacología , Evasión Inmune , Macrófagos/microbiología , Mycobacterium tuberculosis/inmunología , Receptor Toll-Like 2/inmunología , Animales , Carga Bacteriana , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Regulación de la Expresión Génica , Interferón gamma/genética , Interferón gamma/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lisosomas/efectos de los fármacos , Lisosomas/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Fusión de Membrana/efectos de los fármacos , Fusión de Membrana/inmunología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/inmunología , Imitación Molecular , Mycobacterium tuberculosis/crecimiento & desarrollo , Mycobacterium tuberculosis/patogenicidad , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Fagosomas/efectos de los fármacos , Fagosomas/inmunología , Cultivo Primario de Células , Dominios Proteicos , Estallido Respiratorio/inmunología , Transducción de Señal , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
6.
PLoS One ; 10(8): e0135994, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26296096

RESUMEN

Killing of virally infected cells or tumor cells by cytotoxic T lymphocytes requires targeting of lytic granules to the junction between the CTL and its target. We used whole-cell patch clamp to measure the cell capacitance at fixed intracellular [Ca2+] to study fusion of lytic granules in human CTLs. Expression of a fluorescently labeled human granzyme B construct allowed identification of lytic granule fusion using total internal reflection fluorescence microscopy. In this way capacitance steps due to lytic granule fusion were identified. Our goal was to determine the size of fusing lytic granules and to describe their behavior at the plasma membrane. On average, 5.02 ± 3.09 (mean ± s.d.) lytic granules were released per CTL. The amplitude of lytic granule fusion events was ~ 3.3 fF consistent with a diameter of about 325 nm. Fusion latency was biphasic with time constants of 15.9 and 106 seconds. The dwell time of fusing lytic granules was exponentially distributed with a mean dwell time of 28.5 seconds. Fusion ended in spite of the continued presence of granules at the immune synapse. The mobility of fusing granules at the membrane was indistinguishable from that of lytic granules which failed to fuse. While dwelling at the plasma membrane lytic granules exhibit mobility consistent with docking interspersed with short periods of greater mobility. The failure of lytic granules to fuse when visible in TIRF at the membrane may indicate that a membrane-confined reaction is rate limiting.


Asunto(s)
Membrana Celular/metabolismo , Gránulos Citoplasmáticos/metabolismo , Sinapsis Inmunológicas/metabolismo , Fusión de Membrana/inmunología , Linfocitos T Citotóxicos/metabolismo , Calcio/inmunología , Calcio/metabolismo , Membrana Celular/inmunología , Membrana Celular/ultraestructura , Gránulos Citoplasmáticos/inmunología , Gránulos Citoplasmáticos/ultraestructura , Citotoxicidad Inmunológica , Capacidad Eléctrica , Electroporación , Exocitosis , Expresión Génica , Granzimas/genética , Granzimas/inmunología , Granzimas/metabolismo , Humanos , Sinapsis Inmunológicas/inmunología , Sinapsis Inmunológicas/ultraestructura , Cinética , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/inmunología , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Cultivo Primario de Células , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/ultraestructura , Factores de Tiempo , Proteína Fluorescente Roja
7.
J Biol Chem ; 290(21): 12999-3015, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25787074

RESUMEN

The membrane-proximal external region (MPER) C-terminal segment and the transmembrane domain (TMD) of gp41 are involved in HIV-1 envelope glycoprotein-mediated fusion and modulation of immune responses during viral infection. However, the atomic structure of this functional region remains unsolved. Here, based on the high resolution NMR data obtained for peptides spanning the C-terminal segment of MPER and the TMD, we report two main findings: (i) the conformational variability of the TMD helix at a membrane-buried position; and (ii) the existence of an uninterrupted α-helix spanning MPER and the N-terminal region of the TMD. Thus, our structural data provide evidence for the bipartite organization of TMD predicted by previous molecular dynamics simulations and functional studies, but they do not support the breaking of the helix at Lys-683, as was suggested by some models to mark the initiation of the TMD anchor. Antibody binding energetics examined with isothermal titration calorimetry and humoral responses elicited in rabbits by peptide-based vaccines further support the relevance of a continuous MPER-TMD helix for immune recognition. We conclude that the transmembrane anchor of HIV-1 envelope is composed of two distinct subdomains: 1) an immunogenic helix at the N terminus also involved in promoting membrane fusion; and 2) an immunosuppressive helix at the C terminus, which might also contribute to the late stages of the fusion process. The unprecedented high resolution structural data reported here may guide future vaccine and inhibitor developments.


Asunto(s)
Anticuerpos Anti-VIH/inmunología , Proteína gp41 de Envoltorio del VIH/química , Proteína gp41 de Envoltorio del VIH/inmunología , Infecciones por VIH/inmunología , Fusión de Membrana/inmunología , Fragmentos de Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Dicroismo Circular , Cristalografía por Rayos X , Proteína gp41 de Envoltorio del VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Estructura Secundaria de Proteína , Conejos , Internalización del Virus
8.
Blood ; 125(10): 1566-77, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25564401

RESUMEN

Familial hemophagocytic lymphohistiocytosis (F-HLH) and Griscelli syndrome type 2 (GS) are life-threatening immunodeficiencies characterized by impaired cytotoxic T lymphocyte (CTL) and natural killer (NK) cell lytic activity. In the majority of cases, these disorders are caused by biallelic inactivating germline mutations in genes such as RAB27A (GS) and PRF1, UNC13D, STX11, and STXBP2 (F-HLH). Although monoallelic (ie, heterozygous) mutations have been identified in certain patients, the clinical significance and molecular mechanisms by which these mutations influence CTL and NK cell function remain poorly understood. Here, we characterize 2 novel monoallelic hemophagocytic lymphohistiocytosis (HLH)-associated mutations affecting codon 65 of STXPB2, the gene encoding Munc18-2, a member of the SEC/MUNC18 family. Unlike previously described Munc18-2 mutants, Munc18-2(R65Q) and Munc18-2(R65W) retain the ability to interact with and stabilize syntaxin 11. However, presence of Munc18-2(R65Q/W) in patient-derived lymphocytes and forced expression in control CTLs and NK cells diminishes degranulation and cytotoxic activity. Mechanistic studies reveal that mutations affecting R65 hinder membrane fusion in vitro by arresting the late steps of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-complex assembly. Collectively, these results reveal a direct role for SEC/MUNC18 proteins in promoting SNARE-complex assembly in vivo and suggest that STXBP2 R65 mutations operate in a novel dominant-negative fashion to impair lytic granule fusion and contribute to HLH.


Asunto(s)
Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/inmunología , Proteínas Munc18/genética , Proteínas Mutantes/genética , Mutación Missense , Proteínas SNARE/inmunología , Adulto , Sustitución de Aminoácidos , Niño , Preescolar , Codón/genética , Femenino , Genes Dominantes , Células HeLa , Heterocigoto , Humanos , Lactante , Células Asesinas Naturales/inmunología , Linfohistiocitosis Hemofagocítica/metabolismo , Masculino , Fusión de Membrana/genética , Fusión de Membrana/inmunología , Persona de Mediana Edad , Modelos Biológicos , Modelos Moleculares , Proteínas Munc18/química , Proteínas Munc18/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Qa-SNARE/química , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas SNARE/metabolismo , Linfocitos T Citotóxicos/inmunología
9.
Proc Natl Acad Sci U S A ; 111(48): E5143-8, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25404330

RESUMEN

The ability of antibodies binding the influenza hemagglutinin (HA) protein to neutralize viral infectivity is of key importance in the design of next-generation vaccines and for prophylactic and therapeutic use. The two antibodies CR6261 and CR8020 have recently been shown to efficiently neutralize influenza A infection by binding to and inhibiting the influenza A HA protein that is responsible for membrane fusion in the early steps of viral infection. Here, we use single-particle fluorescence microscopy to correlate the number of antibodies or antibody fragments (Fab) bound to an individual virion with the capacity of the same virus particle to undergo membrane fusion. To this end, individual, infectious virus particles bound by fluorescently labeled antibodies/Fab are visualized as they fuse to a planar, supported lipid bilayer. The fluorescence intensity arising from the virus-bound antibodies/Fab is used to determine the number of molecules attached to viral HA while a fluorescent marker in the viral membrane is used to simultaneously obtain kinetic information on the fusion process. We experimentally determine that the stoichiometry required for fusion inhibition by both antibody and Fab leaves large numbers of unbound HA epitopes on the viral surface. Kinetic measurements of the fusion process reveal that those few particles capable of fusion at high antibody/Fab coverage display significantly slower hemifusion kinetics. Overall, our results support a membrane fusion mechanism requiring the stochastic, coordinated action of multiple HA trimers and a model of fusion inhibition by stem-binding antibodies through disruption of this coordinated action.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Virus de la Influenza A/inmunología , Fusión de Membrana/inmunología , Virión/inmunología , Anticuerpos Neutralizantes/farmacología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/farmacología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H1N1 del Virus de la Influenza A/ultraestructura , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/ultraestructura , Virus de la Influenza A/fisiología , Virus de la Influenza A/ultraestructura , Gripe Humana/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Cinética , Fusión de Membrana/efectos de los fármacos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Método de Montecarlo , Unión Proteica , Virión/efectos de los fármacos , Virión/ultraestructura , Internalización del Virus/efectos de los fármacos
10.
PLoS Pathog ; 9(10): e1003734, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204276

RESUMEN

Mycobacterium tuberculosis (Mtb) disrupts anti-microbial pathways of macrophages, cells that normally kill bacteria. Over 40 years ago, D'Arcy Hart showed that Mtb avoids delivery to lysosomes, but the molecular mechanisms that allow Mtb to elude lysosomal degradation are poorly understood. Specialized secretion systems are often used by bacterial pathogens to translocate effectors that target the host, and Mtb encodes type VII secretion systems (TSSSs) that enable mycobacteria to secrete proteins across their complex cell envelope; however, their cellular targets are unknown. Here, we describe a systematic strategy to identify bacterial virulence factors by looking for interactions between the Mtb secretome and host proteins using a high throughput, high stringency, yeast two-hybrid (Y2H) platform. Using this approach we identified an interaction between EsxH, which is secreted by the Esx-3 TSSS, and human hepatocyte growth factor-regulated tyrosine kinase substrate (Hgs/Hrs), a component of the endosomal sorting complex required for transport (ESCRT). ESCRT has a well-described role in directing proteins destined for lysosomal degradation into intraluminal vesicles (ILVs) of multivesicular bodies (MVBs), ensuring degradation of the sorted cargo upon MVB-lysosome fusion. Here, we show that ESCRT is required to deliver Mtb to the lysosome and to restrict intracellular bacterial growth. Further, EsxH, in complex with EsxG, disrupts ESCRT function and impairs phagosome maturation. Thus, we demonstrate a role for a TSSS and the host ESCRT machinery in one of the central features of tuberculosis pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Mycobacterium tuberculosis/patogenicidad , Fosfoproteínas/metabolismo , Tuberculosis/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Pared Celular/genética , Pared Celular/inmunología , Pared Celular/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/inmunología , Endosomas/genética , Endosomas/inmunología , Endosomas/metabolismo , Células HEK293 , Humanos , Membranas Intracelulares/inmunología , Membranas Intracelulares/metabolismo , Lisosomas/genética , Lisosomas/inmunología , Lisosomas/metabolismo , Lisosomas/microbiología , Fusión de Membrana/genética , Fusión de Membrana/inmunología , Ratones , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Tuberculosis/genética , Tuberculosis/inmunología
11.
Eur J Immunol ; 43(3): 667-78, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23280470

RESUMEN

Macrophages provide a first line of defense against bacterial infection by engulfing and killing invading bacteria, but intracellular bacteria such as Listeria monocytogenes (LM) can survive in macrophages by various mechanisms of evasion. Complement receptor of the immunoglobulin (CRIg), a C3b receptor, binds to C3b on opsonized bacteria and facilitates clearance of the bacteria by promoting their uptake. We found that CRIg signaling induced by agonistic anti-CRIg mAb enhanced the killing of intracellular LM by macrophages, and that this occurred in LM-containing phagosomes. Chloride intra-cellular channel 3 CLIC3, an intracellular chloride channel protein, was essential for CRIg-mediated LM killing by directly interacting with the cytoplasmic domain of CRIg, and the two proteins colocalized on the membranes of LM-containing vacuoles. CLIC3(-/-) mice were as susceptible to LM as CRIg(-/-) mice. These findings identify a mechanism embedded in the process by which macrophages take up opsonized bacteria that prevents the bacteria from evading cell-mediated killing.


Asunto(s)
Canales de Cloruro/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Fagosomas/inmunología , Receptores de Complemento 3b/metabolismo , Receptores de Complemento/metabolismo , Transducción de Señal , Animales , Línea Celular , Cloruros/metabolismo , Femenino , Humanos , Listeria monocytogenes/inmunología , Lisosomas/inmunología , Lisosomas/metabolismo , Macrófagos/microbiología , Masculino , Fusión de Membrana/inmunología , Ratones , Fagocitosis/genética , Fagocitosis/inmunología , Unión Proteica , Receptores de Complemento/genética , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/inmunología , Vacuolas/inmunología , Vacuolas/metabolismo , Vacuolas/microbiología
12.
Mol Immunol ; 54(2): 148-56, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23270686

RESUMEN

The crown region of the V3 loop in HIV-1 that contains the conserved amino acid sequence GPGR/G is known as the principal neutralizing determinant due to the extraordinary ability of antibodies to this region to neutralize the virus. To complement the existing peptide models of this epitope, we describe a family of 18 phage-displayed peptides, which include linear 12mer and constrained 7mer peptides that was selected by screening random libraries with serum from HIV-1 subtype B-infected patients. The 7mer constrained peptides presented two conserved amino acid sequences: PR-L in N-terminus and GPG in the C-terminus. On the basis of these peptides we propose a mimotope model of the V3 crown epitope in which the PR-L and GPG sequences represent the two known epitope binding sites. The GPG, has the same function as the V3 crown GPGR sequence but without the involvement of the "R" despite its being considered as the signature of the epitope in B-subtype viruses. The PR-L contains a proline not existing in the epitope that is postulated to induce kinks in the backbones of all peptides and create a spatial element mimicking the N-terminal conformationally variable binding site. Rabbit serum to these mimotopes recognized the V3 peptides and moderately decreased the fusion between HIV-1 Env- and CD4-expressing Jurkat cells. This study proposes the efficient generation by means of patient sera of V3 epitope mimics validated by interaction with the antibodies to contemporary viruses induced in patients. The serum antibody-selectable mimotopes are sources of novel information on the fine structure-function properties of HIV-1 principal neutralizing domain and candidate anti-HIV-1 immunogens.


Asunto(s)
Epítopos/inmunología , Proteína gp120 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Fragmentos de Péptidos/inmunología , Biblioteca de Péptidos , Secuencia de Aminoácidos , Animales , Técnicas de Visualización de Superficie Celular , Mapeo Epitopo , Epítopos/química , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/inmunología , Anticuerpos Anti-VIH/aislamiento & purificación , Proteína gp120 de Envoltorio del VIH/química , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina G/aislamiento & purificación , Fusión de Membrana/inmunología , Fragmentos de Péptidos/química , Conejos
13.
Viruses ; 4(11): 3020-43, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23202514

RESUMEN

Apoptosis of uninfected bystander cells is a key element of HIV pathogenesis and believed to be the driving force behind the selective depletion of CD4+ T cells leading to immunodeficiency. While several viral proteins have been implicated in this process the complex interaction between Env glycoprotein expressed on the surface of infected cells and the receptor and co-receptor expressing bystander cells has been proposed as a major mechanism. HIV-1 utilizes CD4 as the primary receptor for entry into cells; however, it is the viral co-receptor usage that greatly influences CD4 decline and progression to AIDS. This phenomenon is relatively simple for X4 viruses, which arise later during the course of the disease, are considered to be highly fusogenic, and cause a rapid CD4+ T cell decline. However, in contrast, R5 viruses in general have a greater transmissibility, are encountered early during the disease and have a lesser pathogenic potential than the former. The above generalization gets complicated in numerous situations where R5 viruses persist throughout the disease and are capable of causing a rigorous CD4+ T cell decline. This review will discuss the multiple factors that are reported to influence HIV induced bystander apoptosis and pathogenesis including Env glycoprotein phenotype, virus tropism, disease stage, co-receptor expression on CD4+ T cells, immune activation and therapies targeting the viral envelope.


Asunto(s)
Apoptosis/inmunología , Efecto Espectador/inmunología , Infecciones por VIH/inmunología , VIH-1/patogenicidad , Animales , Autofagia/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteína gp41 de Envoltorio del VIH/metabolismo , Humanos , Fusión de Membrana/inmunología , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo
14.
J Biol Chem ; 287(49): 40841-9, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23043104

RESUMEN

HIV entry involves binding of the trimeric viral envelope glycoprotein (Env) gp120/gp41 to cell surface receptors, which triggers conformational changes in Env that drive the membrane fusion reaction. The conformational landscape that the lipids and Env navigate en route to fusion has been examined by biophysical measurements on the microscale, whereas electron tomography, x-rays, and NMR have provided insights into the process on the nanoscale and atomic scale. However, the coupling between the lipid and protein pathways that give rise to fusion has not been resolved. Here, we discuss the known and unknown about the overall HIV Env-mediated fusion process.


Asunto(s)
Proteína gp120 de Envoltorio del VIH/metabolismo , Proteína gp41 de Envoltorio del VIH/metabolismo , Infecciones por VIH/virología , Biofisica/métodos , Cristalografía por Rayos X/métodos , Tomografía con Microscopio Electrónico/métodos , Humanos , Lípidos/química , Espectroscopía de Resonancia Magnética/métodos , Fusión de Membrana/inmunología , Modelos Moleculares , Conformación Molecular , Conformación Proteica
15.
PLoS One ; 7(1): e30152, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253913

RESUMEN

Herpesvirus entry is a complicated process involving multiple virion glycoproteins and culminating in membrane fusion. Glycoprotein conformation changes are likely to play key roles. Studies of recombinant glycoproteins have revealed some structural features of the virion fusion machinery. However, how the virion glycoproteins change during infection remains unclear. Here using conformation-specific monoclonal antibodies we show in situ that each component of the Murid Herpesvirus-4 (MuHV-4) entry machinery--gB, gH/gL and gp150--changes in antigenicity before tegument protein release begins. Further changes then occurred upon actual membrane fusion. Thus virions revealed their final fusogenic form only in late endosomes. The substantial antigenic differences between this form and that of extracellular virions suggested that antibodies have only a limited opportunity to block virion membrane fusion.


Asunto(s)
Antígenos Virales/inmunología , Fusión de Membrana/inmunología , Rhadinovirus/inmunología , Proteínas Virales de Fusión/inmunología , Cloruro de Amonio/farmacología , Animales , Línea Celular , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Cinética , Macrólidos/farmacología , Fusión de Membrana/efectos de los fármacos , Pruebas de Neutralización , Rhadinovirus/efectos de los fármacos , Virión/inmunología , Internalización del Virus/efectos de los fármacos
16.
PLoS One ; 7(12): e52740, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23285173

RESUMEN

The membrane proximal external region (MPER) of the fusogenic HIV-1 glycoprotein-41 harbors the epitope sequence recognized by 2F5, a broadly neutralizing antibody isolated from an infected individual. Structural mimicry of the conserved MPER 2F5 epitope constitutes a pursued goal in the field of anti-HIV vaccine development. It has been proposed that 2F5 epitope folding into its native state is attained in the vicinity of the membrane interface and might involve interactions with other viral structures. Here we present results indicating that oligomeric complexes established between MPER and the conserved amino-terminal fusion peptide (FP) can partition into lipid vesicles and be specifically bound by the 2F5 antibody at their surfaces. Cryo-transmission electron microscopy of liposomes doped with MPER:FP peptide mixtures provided the structural grounds for complex recognition by antibody at lipid bilayer surfaces. Supporting the immunogenicity of the membrane-bound complex, these MPER:FP peptide-vesicle formulations could trigger cross-reactive anti-MPER antibodies in rabbits. Thus, our observations suggest that contacts with N-terminal regions of gp41 may stabilize the 2F5 epitope as a membrane-surface antigen.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Anti-VIH/inmunología , Proteína gp41 de Envoltorio del VIH/química , Proteína gp41 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/metabolismo , Epítopos/química , Epítopos/inmunología , Epítopos/metabolismo , Anticuerpos Anti-VIH/metabolismo , Proteína gp41 de Envoltorio del VIH/metabolismo , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Liposomas/química , Liposomas/inmunología , Sustancias Macromoleculares/inmunología , Sustancias Macromoleculares/metabolismo , Sustancias Macromoleculares/ultraestructura , Fusión de Membrana/inmunología , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Unión Proteica/inmunología , Conejos
17.
Cell Immunol ; 271(2): 267-79, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21843881

RESUMEN

Here we imaged the exocytosis of lytic granules from human CD8(+) cytotoxic T lymphocytes using rapid total internal reflection microscopy, Lamp-1 tagged with mGFP to follow the fate of the lytic granule membrane, and granzyme A, granzyme B or serglycin tagged with mRFP to follow the fate of lytic granule cargo. Lytic granules were released by full fusion with the plasma membrane, such that the entire granule content for all three cargos visualized was released on a subsecond time scale. The behavior of GFP-Lamp-1 was, however, more complex. While it entered the plasma membrane in all cases, the extent to which it then diffused away from the site of exocytosis varied from nearly complete to highly restricted. Finally, the diffusion properties upon release of the three cargos examined put an upper limit on the size of the macromolecular complex of granzyme and serglycin that is presented to the target cell.


Asunto(s)
Exocitosis/fisiología , Fusión de Membrana/fisiología , Linfocitos T Citotóxicos/fisiología , Animales , Exocitosis/inmunología , Granzimas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Técnicas In Vitro , Proteínas de Membrana de los Lisosomas/metabolismo , Fusión de Membrana/inmunología , Ratones , Microscopía Fluorescente , Proteoglicanos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Vesículas Secretoras/inmunología , Vesículas Secretoras/fisiología , Linfocitos T Citotóxicos/inmunología , Proteínas de Transporte Vesicular/metabolismo
18.
Cell Death Differ ; 18(8): 1298-304, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21637288

RESUMEN

Almost all plant cells have large vacuoles that contain both hydrolytic enzymes and a variety of defense proteins. Plants use vacuoles and vacuolar contents for programmed cell death (PCD) in two different ways: for a destructive way and for a non-destructive way. Destruction is caused by vacuolar membrane collapse, followed by the release of vacuolar hydrolytic enzymes into the cytosol, resulting in rapid and direct cell death. The destructive way is effective in the digestion of viruses proliferating in the cytosol, in susceptible cell death induced by fungal toxins, and in developmental cell death to generate integuments (seed coats) and tracheary elements. On the other hand, the non-destructive way involves fusion of the vacuolar and the plasma membrane, which allows vacuolar defense proteins to be discharged into the extracellular space where the bacteria proliferate. Membrane fusion, which is normally suppressed, was triggered in a proteasome-dependent manner. Intriguingly, both ways use enzymes with caspase-like activity; the membrane-fusion system uses proteasome subunit PBA1 with caspase-3-like activity, and the vacuolar-collapse system uses vacuolar processing enzyme (VPE) with caspase-1-like activity. This review summarizes two different ways of vacuole-mediated PCD and discusses how plants use them to attack pathogens that invade unexpectedly.


Asunto(s)
Muerte Celular/fisiología , Células Vegetales , Plantas/enzimología , Vacuolas/enzimología , Fusión de Membrana/inmunología , Inmunidad de la Planta/inmunología , Proteínas de Plantas/metabolismo , Plantas/inmunología , Plantas/microbiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Vacuolas/ultraestructura
19.
Vaccine ; 29(32): 5250-9, 2011 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-21609746

RESUMEN

BACKGROUND: Cell-to-cell HIV spread through virological synapses proceeds in two steps, first HIV particles are rapidly transferred to target cells in a CD4-dependent manner and then coreceptor-dependent events allow for infection or death of single target cells and cell-to-cell fusion. METHODS: 293T or MOLT cells producing HIV particles were cocultured with primary CD4 T-cells or reporter cell lines. The extent of HIV transfer, cell fusion and target cell death was assessed. Inhibition by sera from 19 HIV-infected patients was evaluated and compared with cell-free HIV neutralization using different envelopes from clades A, B, C and E. RESULTS: Sera showed different abilities to protect CD4 T-cells from cell-to-cell transfer, fusion or death when cocultured with HIV producing 293T cells. Some sera were able to block all parameters (a property of IgGb12), while other showed lower activity against HIV transfer despite being able to block fusion and death (a property of antibodies blocking post-CD4 binding steps). Neutralization of cell-to-cell HIV transfer strongly correlated with IgG binding to native Env. Interestingly, sera that efficiently blocked HIV transfer showed broader neutralizing response, as they neutralized a higher percentage of the viruses tested compared with sera showing low CD4 binding site responses (P=0.01). Similar results were observed in a model of T cell-T cell HIV transmission, although this experimental model showed lower capacity to discriminate broadly neutralizing responses. CONCLUSION: Cell-to-cell HIV transfer assays identify sera with broadly neutralizing capacity and may help to characterize anti-HIV humoral responses.


Asunto(s)
Anticuerpos Neutralizantes/análisis , Anticuerpos Antivirales/sangre , Linfocitos T CD4-Positivos/virología , Infecciones por VIH/inmunología , VIH/crecimiento & desarrollo , VIH/inmunología , Inmunidad Humoral , Adulto , Anticuerpos Neutralizantes/inmunología , Sitios de Unión de Anticuerpos , Fusión Celular , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteína gp120 de Envoltorio del VIH/inmunología , Proteína gp120 de Envoltorio del VIH/metabolismo , Proteína gp41 de Envoltorio del VIH/inmunología , Proteína gp41 de Envoltorio del VIH/metabolismo , Humanos , Inmunoglobulina G , Macrófagos/virología , Masculino , Fusión de Membrana/inmunología , Fusión de Membrana/fisiología , Sinapsis/virología
20.
Mol Biol Cell ; 22(8): 1148-66, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21346189

RESUMEN

As the initial barrier to viral entry, the plasma membrane along with the membrane trafficking machinery and cytoskeleton are of fundamental importance in the viral cycle. However, little is known about the contribution of plasma membrane dynamics during early human immunodeficiency virus type 1 (HIV-1) infection. Considering that ADP ribosylation factor 6 (Arf6) regulates cellular invasion via several microorganisms by coordinating membrane trafficking, our aim was to study the function of Arf6-mediated membrane dynamics on HIV-1 entry and infection of T lymphocytes. We observed that an alteration of the Arf6-guanosine 5'-diphosphate/guanosine 5'-triphosphate (GTP/GDP) cycle, by GDP-bound or GTP-bound inactive mutants or by specific Arf6 silencing, inhibited HIV-1 envelope-induced membrane fusion, entry, and infection of T lymphocytes and permissive cells, regardless of viral tropism. Furthermore, cell-to-cell HIV-1 transmission of primary human CD4(+) T lymphocytes was inhibited by Arf6 knockdown. Total internal reflection fluorescence microscopy showed that Arf6 mutants provoked the accumulation of phosphatidylinositol-(4,5)-biphosphate-associated structures on the plasma membrane of permissive cells, without affecting CD4-viral attachment but impeding CD4-dependent HIV-1 entry. Arf6 silencing or its mutants did not affect fusion, entry, and infection of vesicular stomatitis virus G-pseudotyped viruses or ligand-induced CXCR4 or CCR5 endocytosis, both clathrin-dependent processes. Therefore we propose that efficient early HIV-1 infection of CD4(+) T lymphocytes requires Arf6-coordinated plasma membrane dynamics that promote viral fusion and entry.


Asunto(s)
Factores de Ribosilacion-ADP , Linfocitos T CD4-Positivos/metabolismo , VIH-1/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/antagonistas & inhibidores , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Endocitosis/genética , Endocitosis/inmunología , Femenino , Silenciador del Gen , Guanosina Difosfato/metabolismo , Células HEK293 , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/inmunología , Células HeLa , Humanos , Fusión de Membrana/genética , Fusión de Membrana/inmunología , Microscopía Fluorescente , Fosfatidilinositol 4,5-Difosfato/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Receptores CCR5/inmunología , Receptores CCR5/metabolismo , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Transfección , Vesiculovirus/metabolismo , Internalización del Virus , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...