Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 184(5): 1201-1213.e14, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33571429

RESUMEN

Memory B cells play a fundamental role in host defenses against viruses, but to date, their role has been relatively unsettled in the context of SARS-CoV-2. We report here a longitudinal single-cell and repertoire profiling of the B cell response up to 6 months in mild and severe COVID-19 patients. Distinct SARS-CoV-2 spike-specific activated B cell clones fueled an early antibody-secreting cell burst as well as a durable synchronous germinal center response. While highly mutated memory B cells, including pre-existing cross-reactive seasonal Betacoronavirus-specific clones, were recruited early in the response, neutralizing SARS-CoV-2 RBD-specific clones accumulated with time and largely contributed to the late, remarkably stable, memory B cell pool. Highlighting germinal center maturation, these cells displayed clear accumulation of somatic mutations in their variable region genes over time. Overall, these findings demonstrate that an antigen-driven activation persisted and matured up to 6 months after SARS-CoV-2 infection and may provide long-term protection.


Asunto(s)
Linfocitos B/inmunología , COVID-19/inmunología , Memoria Inmunológica , Adulto , COVID-19/fisiopatología , Citometría de Flujo , Centro Germinal/citología , Humanos , Activación de Linfocitos , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual , Glicoproteína de la Espiga del Coronavirus/química
2.
Immunity ; 56(9): 2137-2151.e7, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37543032

RESUMEN

How infection by a viral variant showing antigenic drift impacts a preformed mature human memory B cell (MBC) repertoire remains an open question. Here, we studied the MBC response up to 6 months after SARS-CoV-2 Omicron BA.1 breakthrough infection in individuals previously vaccinated with three doses of the COVID-19 mRNA vaccine. Longitudinal analysis, using single-cell multi-omics and functional analysis of monoclonal antibodies from RBD-specific MBCs, revealed that a BA.1 breakthrough infection mostly recruited pre-existing cross-reactive MBCs with limited de novo response against BA.1-restricted epitopes. Reorganization of clonal hierarchy and new rounds of germinal center reactions, however, combined to maintain diversity and induce progressive maturation of the MBC repertoire against common Hu-1 and BA.1, but not BA.5-restricted, SARS-CoV-2 Spike RBD epitopes. Such remodeling was further associated with a marked improvement in overall neutralizing breadth and potency. These findings have fundamental implications for the design of future vaccination booster strategies.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Humanos , SARS-CoV-2 , Células B de Memoria , Infección Irruptiva , Epítopos , Anticuerpos Antivirales , Anticuerpos Neutralizantes
3.
Immunity ; 55(6): 1096-1104.e4, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35483354

RESUMEN

The SARS-CoV-2 Omicron variant can escape neutralization by vaccine-elicited and convalescent antibodies. Memory B cells (MBCs) represent another layer of protection against SARS-CoV-2, as they persist after infection and vaccination and improve their affinity. Whether MBCs elicited by mRNA vaccines can recognize the Omicron variant remains unclear. We assessed the affinity and neutralization potency against the Omicron variant of several hundred naturally expressed MBC-derived monoclonal IgG antibodies from vaccinated COVID-19-recovered and -naive individuals. Compared with other variants of concern, Omicron evaded recognition by a larger proportion of MBC-derived antibodies, with only 30% retaining high affinity against the Omicron RBD, and the reduction in neutralization potency was even more pronounced. Nonetheless, neutralizing MBC clones could be found in all the analyzed individuals. Therefore, despite the strong immune escape potential of the Omicron variant, these results suggest that the MBC repertoire generated by mRNA vaccines still provides some protection against the Omicron variant in vaccinated individuals.


Asunto(s)
COVID-19 , SARS-CoV-2 , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Humanos , Células B de Memoria , ARN Mensajero/genética , Glicoproteína de la Espiga del Coronavirus/genética , Vacunación
4.
Immunity ; 54(12): 2893-2907.e5, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34614412

RESUMEN

In addition to serum immunoglobulins, memory B cell (MBC) generation against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is another layer of immune protection, but the quality of MBC responses in naive and coronavirus disease 2019 (COVID-19)-recovered individuals after vaccination remains ill defined. We studied longitudinal cohorts of naive and disease-recovered individuals for up to 2 months after SARS-CoV-2 mRNA vaccination. We assessed the quality of the memory response by analysis of antibody repertoires, affinity, and neutralization against variants of concern (VOCs) using unbiased cultures of 2,452 MBCs. Upon boosting, the MBC pool of recovered individuals expanded selectively, matured further, and harbored potent neutralizers against VOCs. Although naive individuals had weaker neutralizing serum responses, half of their RBD-specific MBCs displayed high affinity toward multiple VOCs, including delta (B.1.617.2), and one-third retained neutralizing potency against beta (B.1.351). Our data suggest that an additional challenge in naive vaccinees could recall such affinity-matured MBCs and allow them to respond efficiently to VOCs.


Asunto(s)
Vacuna BNT162/inmunología , COVID-19/inmunología , Células B de Memoria/inmunología , Células Precursoras de Linfocitos B/inmunología , ARN Mensajero/genética , SARS-CoV-2/fisiología , Animales , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Afinidad de Anticuerpos , Células Cultivadas , Convalecencia , Humanos , Inmunización Secundaria , Memoria Inmunológica , Vacunación Masiva , SARS-CoV-2/genética , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología
5.
Nature ; 621(7977): 120-128, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37558883

RESUMEN

Humans display substantial interindividual clinical variability after SARS-CoV-2 infection1-3, the genetic and immunological basis of which has begun to be deciphered4. However, the extent and drivers of population differences in immune responses to SARS-CoV-2 remain unclear. Here we report single-cell RNA-sequencing data for peripheral blood mononuclear cells-from 222 healthy donors of diverse ancestries-that were stimulated with SARS-CoV-2 or influenza A virus. We show that SARS-CoV-2 induces weaker, but more heterogeneous, interferon-stimulated gene activity compared with influenza A virus, and a unique pro-inflammatory signature in myeloid cells. Transcriptional responses to viruses display marked population differences, primarily driven by changes in cell abundance including increased lymphoid differentiation associated with latent cytomegalovirus infection. Expression quantitative trait loci and mediation analyses reveal a broad effect of cell composition on population disparities in immune responses, with genetic variants exerting a strong effect on specific loci. Furthermore, we show that natural selection has increased population differences in immune responses, particularly for variants associated with SARS-CoV-2 response in East Asians, and document the cellular and molecular mechanisms by which Neanderthal introgression has altered immune functions, such as the response of myeloid cells to viruses. Finally, colocalization and transcriptome-wide association analyses reveal an overlap between the genetic basis of immune responses to SARS-CoV-2 and COVID-19 severity, providing insights into the factors contributing to current disparities in COVID-19 risk.


Asunto(s)
COVID-19 , Genética de Población , SARS-CoV-2 , Análisis de Expresión Génica de una Sola Célula , Animales , Humanos , Diferenciación Celular , COVID-19/genética , COVID-19/inmunología , COVID-19/virología , Citomegalovirus/fisiología , Pueblos del Este de Asia/genética , Introgresión Genética , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Interferones/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Células Mieloides/inmunología , Hombre de Neandertal/genética , Hombre de Neandertal/inmunología , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Selección Genética , Latencia del Virus
6.
Nat Immunol ; 17(9): 1102-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27339099

RESUMEN

Zika virus (ZIKV) was discovered in 1947 and was thought to lead to relatively mild disease. The recent explosive outbreak of ZIKV in South America has led to widespread concern, with reports of neurological sequelae ranging from Guillain Barré syndrome to microcephaly. ZIKV infection has occurred in areas previously exposed to dengue virus (DENV), a flavivirus closely related to ZIKV. Here we investigated the serological cross-reaction between the two viruses. Plasma immune to DENV showed substantial cross-reaction to ZIKV and was able to drive antibody-dependent enhancement (ADE) of ZIKV infection. Using a panel of human monoclonal antibodies (mAbs) to DENV, we showed that most antibodies that reacted to DENV envelope protein also reacted to ZIKV. Antibodies to linear epitopes, including the immunodominant fusion-loop epitope, were able to bind ZIKV but were unable to neutralize the virus and instead promoted ADE. Our data indicate that immunity to DENV might drive greater ZIKV replication and have clear implications for disease pathogenesis and future vaccine programs for ZIKV and DENV.


Asunto(s)
Acrecentamiento Dependiente de Anticuerpo , Reacciones Cruzadas , Virus del Dengue/fisiología , Dengue/inmunología , Infección por el Virus Zika/inmunología , Virus Zika/fisiología , Adolescente , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Células Cultivadas , Niño , Preescolar , Dengue/epidemiología , Mapeo Epitopo , Femenino , Síndrome de Guillain-Barré/epidemiología , Humanos , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/metabolismo , Masculino , Microcefalia/epidemiología , Unión Proteica , América del Sur/epidemiología , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral , Infección por el Virus Zika/epidemiología
7.
Proc Natl Acad Sci U S A ; 117(12): 6675-6685, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32152119

RESUMEN

A comprehensive understanding of the development and evolution of human B cell responses induced by pathogen exposure will facilitate the design of next-generation vaccines. Here, we utilized a high-throughput single B cell cloning technology to longitudinally track the human B cell response to the yellow fever virus 17D (YFV-17D) vaccine. The early memory B cell (MBC) response was mediated by both classical immunoglobulin M (IgM) (IgM+CD27+) and switched immunoglobulin (swIg+) MBC populations; however, classical IgM MBCs waned rapidly, whereas swIg+ and atypical IgM+ and IgD+ MBCs were stable over time. Affinity maturation continued for 6 to 9 mo following vaccination, providing evidence for the persistence of germinal center activity long after the period of active viral replication in peripheral blood. Finally, a substantial fraction of the neutralizing antibody response was mediated by public clones that recognize a fusion loop-proximal antigenic site within domain II of the viral envelope glycoprotein. Overall, our findings provide a framework for understanding the dynamics and complexity of human B cell responses elicited by infection and vaccination.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Linfocitos B/inmunología , Memoria Inmunológica/inmunología , Vacuna contra la Fiebre Amarilla/inmunología , Fiebre Amarilla/prevención & control , Virus de la Fiebre Amarilla/inmunología , Adulto , Humanos , Vacunación , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/inmunología , Replicación Viral , Fiebre Amarilla/inmunología , Fiebre Amarilla/virología , Vacuna contra la Fiebre Amarilla/administración & dosificación
8.
Nature ; 536(7614): 48-53, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27338953

RESUMEN

Zika virus is a member of the Flavivirus genus that had not been associated with severe disease in humans until the recent outbreaks, when it was linked to microcephaly in newborns in Brazil and to Guillain-Barré syndrome in adults in French Polynesia. Zika virus is related to dengue virus, and here we report that a subset of antibodies targeting a conformational epitope isolated from patients with dengue virus also potently neutralize Zika virus. The crystal structure of two of these antibodies in complex with the envelope protein of Zika virus reveals the details of a conserved epitope, which is also the site of interaction of the envelope protein dimer with the precursor membrane (prM) protein during virus maturation. Comparison of the Zika and dengue virus immunocomplexes provides a lead for rational, epitope-focused design of a universal vaccine capable of eliciting potent cross-neutralizing antibodies to protect simultaneously against both Zika and dengue virus infections.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Reacciones Cruzadas/inmunología , Virus del Dengue/inmunología , Epítopos/química , Vacunas Virales/química , Virus Zika/inmunología , Anticuerpos Monoclonales/inmunología , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/inmunología , Brasil , Cristalografía por Rayos X , Dengue/inmunología , Vacunas contra el Dengue/química , Vacunas contra el Dengue/inmunología , Virus del Dengue/química , Epítopos/inmunología , Humanos , Modelos Moleculares , Filogenia , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Virus Zika/química , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/prevención & control
9.
Nature ; 520(7545): 109-13, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25581790

RESUMEN

Dengue disease is caused by four different flavivirus serotypes, which infect 390 million people yearly with 25% symptomatic cases and for which no licensed vaccine is available. Recent phase III vaccine trials showed partial protection, and in particular no protection for dengue virus serotype 2 (refs 3, 4). Structural studies so far have characterized only epitopes recognized by serotype-specific human antibodies. We recently isolated human antibodies potently neutralizing all four dengue virus serotypes. Here we describe the X-ray structures of four of these broadly neutralizing antibodies in complex with the envelope glycoprotein E from dengue virus serotype 2, revealing that the recognition determinants are at a serotype-invariant site at the E-dimer interface, including the exposed main chain of the E fusion loop and the two conserved glycan chains. This 'E-dimer-dependent epitope' is also the binding site for the viral glycoprotein prM during virus maturation in the secretory pathway of the infected cell, explaining its conservation across serotypes and highlighting an Achilles' heel of the virus with respect to antibody neutralization. These findings will be instrumental for devising novel immunogens to protect simultaneously against all four serotypes of dengue virus.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , Virus del Dengue/química , Virus del Dengue/inmunología , Anticuerpos Neutralizantes/genética , Anticuerpos Antivirales/genética , Reacciones Cruzadas/inmunología , Cristalografía por Rayos X , Virus del Dengue/clasificación , Epítopos/química , Epítopos/inmunología , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Conformación Proteica , Multimerización de Proteína , Solubilidad , Especificidad de la Especie , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/inmunología
10.
Nature ; 493(7433): 552-6, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23292515

RESUMEN

Little is known about the three-dimensional organization of rubella virus, which causes a relatively mild measles-like disease in children but leads to serious congenital health problems when contracted in utero. Although rubella virus belongs to the same family as the mosquito-borne alphaviruses, in many respects it is more similar to other aerosol-transmitted human viruses such as the agents of measles and mumps. Although the use of the triple MMR (measles, mumps and rubella) live vaccine has limited its incidence in western countries, congenital rubella syndrome remains an important health problem in the developing world. Here we report the 1.8 Å resolution crystal structure of envelope glycoprotein E1, the main antigen and sole target of neutralizing antibodies against rubella virus. E1 is the main player during entry into target cells owing to its receptor-binding and membrane-fusion functions. The structure reveals the epitope and the neutralization mechanism of an important category of protecting antibodies against rubella infection. It also shows that rubella virus E1 is a class II fusion protein, which had hitherto only been structurally characterized for the arthropod-borne alphaviruses and flaviviruses. In addition, rubella virus E1 has an extensive membrane-fusion surface that includes a metal site, reminiscent of the T-cell immunoglobulin and mucin family of cellular proteins that bind phosphatidylserine lipids at the plasma membrane of cells undergoing apoptosis. Such features have not been seen in any fusion protein crystallized so far. Structural comparisons show that the class II fusion proteins from alphaviruses and flaviviruses, despite belonging to different virus families, are closer to each other than they are to rubella virus E1. This suggests that the constraints on arboviruses imposed by alternating cycles between vertebrates and arthropods resulted in more conservative evolution. By contrast, in the absence of this constraint, the strictly human rubella virus seems to have drifted considerably into a unique niche as sole member of the Rubivirus genus.


Asunto(s)
Evolución Biológica , Virus de la Rubéola/química , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Animales , Sitios de Unión , Línea Celular , Cristalografía por Rayos X , Drosophila melanogaster , Evolución Molecular , Concentración de Iones de Hidrógeno , Liposomas/química , Liposomas/metabolismo , Fusión de Membrana , Metales/metabolismo , Modelos Moleculares , Multimerización de Proteína , Síndrome de Rubéola Congénita/virología , Virus de la Rubéola/fisiología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/ultraestructura
11.
J Virol ; 90(21): 9683-9692, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27535047

RESUMEN

RNA viruses present an extraordinary threat to human health, given their sudden and unpredictable appearance, the potential for rapid spread among the human population, and their ability to evolve resistance to antiviral therapies. The recent emergence of chikungunya virus, Zika virus, and Ebola virus highlights the struggles to contain outbreaks. A significant hurdle is the availability of antivirals to treat the infected or protect at-risk populations. While several compounds show promise in vitro and in vivo, these outbreaks underscore the need to accelerate drug discovery. The replication of several viruses has been described to rely on host polyamines, small and abundant positively charged molecules found in the cell. Here, we describe the antiviral effects of two molecules that alter polyamine levels: difluoromethylornithine (DFMO; also called eflornithine), which is a suicide inhibitor of ornithine decarboxylase 1 (ODC1), and diethylnorspermine (DENSpm), an activator of spermidine/spermine N1-acetyltransferase (SAT1). We show that reducing polyamine levels has a negative effect on diverse RNA viruses, including several viruses involved in recent outbreaks, in vitro and in vivo These findings highlight the importance of the polyamine biosynthetic pathway to viral replication, as well as its potential as a target in the development of further antivirals or currently available molecules, such as DFMO. IMPORTANCE: RNA viruses present a significant hazard to human health, and combatting these viruses requires the exploration of new avenues for targeting viral replication. Polyamines, small positively charged molecules within the cell, have been demonstrated to facilitate infection for a few different viruses. Our study demonstrates that diverse RNA viruses rely on the polyamine pathway for replication and highlights polyamine biosynthesis as a promising drug target.


Asunto(s)
Antivirales/farmacología , Poliaminas/metabolismo , Virus ARN/efectos de los fármacos , Acetiltransferasas/metabolismo , Animales , Línea Celular , Fiebre Chikungunya/tratamiento farmacológico , Fiebre Chikungunya/virología , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/metabolismo , Brotes de Enfermedades , Ebolavirus/efectos de los fármacos , Ebolavirus/metabolismo , Eflornitina/farmacología , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Espermina/análogos & derivados , Espermina/farmacología , Replicación Viral/efectos de los fármacos , Virus Zika/efectos de los fármacos , Infección por el Virus Zika/tratamiento farmacológico , Infección por el Virus Zika/virología
13.
Nat Commun ; 15(1): 1696, 2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38402207

RESUMEN

The yellow fever 17D vaccine (YF17D) is highly effective but is frequently administered to individuals with pre-existing cross-reactive immunity, potentially impacting their immune responses. Here, we investigate the impact of pre-existing flavivirus immunity induced by the tick-borne encephalitis virus (TBEV) vaccine on the response to YF17D vaccination in 250 individuals up to 28 days post-vaccination (pv) and 22 individuals sampled one-year pv. Our findings indicate that previous TBEV vaccination does not affect the early IgM-driven neutralizing response to YF17D. However, pre-vaccination sera enhance YF17D virus infection in vitro via antibody-dependent enhancement (ADE). Following YF17D vaccination, TBEV-pre-vaccinated individuals develop high amounts of cross-reactive IgG antibodies with poor neutralizing capacity. In contrast, TBEV-unvaccinated individuals elicit a non-cross-reacting neutralizing response. Using YF17D envelope protein mutants displaying different epitopes, we identify quaternary dimeric epitopes as the primary target of neutralizing antibodies. Additionally, TBEV-pre-vaccination skews the IgG response towards the pan-flavivirus fusion loop epitope (FLE), capable of mediating ADE of dengue and Zika virus infections in vitro. Together, we propose that YF17D vaccination conceals the FLE in individuals without prior flavivirus exposure but favors a cross-reactive IgG response in TBEV-pre-vaccinated recipients directed to the FLE with potential to enhance dengue virus infection.


Asunto(s)
Dengue , Virus de la Encefalitis Transmitidos por Garrapatas , Vacuna contra la Fiebre Amarilla , Infección por el Virus Zika , Virus Zika , Humanos , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Infección por el Virus Zika/prevención & control , Epítopos , Inmunoglobulina G , Dengue/prevención & control
14.
J Immunol ; 186(10): 5590-602, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21490153

RESUMEN

Influenza A virus (IAV) infection results in a highly contagious respiratory illness leading to substantial morbidity and occasionally death. In this report, we assessed the in vivo physiological contribution of invariant NKT (iNKT) lymphocytes, a subset of lipid-reactive αß T lymphocytes, on the host response and viral pathogenesis using a virulent, mouse-adapted, IAV H3N2 strain. Upon infection with a lethal dose of IAV, iNKT cells become activated in the lungs and bronchoalveolar space to become rapidly anergic to further restimulation. Relative to wild-type animals, C57BL/6 mice deficient in iNKT cells (Jα18(-/-) mice) developed a more severe bronchopneumonia and had an accelerated fatal outcome, a phenomenon reversed by the adoptive transfer of NKT cells prior to infection. The enhanced pathology in Jα18(-/-) animals was not associated with either reduced or delayed viral clearance in the lungs or with a defective local NK cell response. In marked contrast, Jα18(-/-) mice displayed a dramatically reduced IAV-specific CD8(+) T cell response in the lungs and in lung-draining mediastinal lymph nodes. We further show that this defective CD8(+) T cell response correlates with an altered accumulation and maturation of pulmonary CD103(+), but not CD11b(high), dendritic cells in the mediastinal lymph nodes. Taken together, these findings point to a role for iNKT cells in the control of pneumonia as well as in the development of the CD8(+) T cell response during the early stage of acute IAV H3N2 infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inflamación/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Pulmón/inmunología , Células T Asesinas Naturales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Viral/inmunología , Traslado Adoptivo , Animales , Antígenos CD , Bronconeumonía , Antígeno CD11b , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Subtipo H3N2 del Virus de la Influenza A/patogenicidad , Cadenas alfa de Integrinas , Pulmón/virología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Neumonía Viral/patología , Neumonía Viral/virología , Reacción en Cadena de la Polimerasa , Carga Viral
15.
JCI Insight ; 8(5)2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36749641

RESUMEN

Acute kidney injury is one of the most important complications in patients with COVID-19 and is considered a negative prognostic factor with respect to patient survival. The occurrence of direct infection of the kidney by SARS-CoV-2, and its contribution to the renal deterioration process, remain controversial issues. By studying 32 renal biopsies from patients with COVID-19, we verified that the major pathological feature of COVID-19 is acute tubular injury (ATI). Using single-molecule fluorescence in situ hybridization, we showed that SARS-CoV-2 infected living renal cells and that infection, which paralleled renal angiotensin-converting enzyme 2 expression levels, was associated with increased death. Mechanistically, a transcriptomic analysis uncovered specific molecular signatures in SARS-CoV-2-infected kidneys as compared with healthy kidneys and non-COVID-19 ATI kidneys. On the other hand, we demonstrated that SARS-CoV-2 and hantavirus, 2 RNA viruses, activated different genetic networks despite triggering the same pathological lesions. Finally, we identified X-linked inhibitor of apoptosis-associated factor 1 as a critical target of SARS-CoV-2 infection. In conclusion, this study demonstrated that SARS-CoV-2 can directly infect living renal cells and identified specific druggable molecular targets that can potentially aid in the design of novel therapeutic strategies to preserve renal function in patients with COVID-19.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , COVID-19/complicaciones , Hibridación Fluorescente in Situ , Riñón/patología , Biopsia
16.
J Exp Med ; 220(1)2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36342455

RESUMEN

Inborn and acquired deficits of type I interferon (IFN) immunity predispose to life-threatening COVID-19 pneumonia. We longitudinally profiled the B cell response to mRNA vaccination in SARS-CoV-2 naive patients with inherited TLR7, IRF7, or IFNAR1 deficiency, as well as young patients with autoantibodies neutralizing type I IFNs due to autoimmune polyendocrine syndrome type-1 (APS-1) and older individuals with age-associated autoantibodies to type I IFNs. The receptor-binding domain spike protein (RBD)-specific memory B cell response in all patients was quantitatively and qualitatively similar to healthy donors. Sustained germinal center responses led to accumulation of somatic hypermutations in immunoglobulin heavy chain genes. The amplitude and duration of, and viral neutralization by, RBD-specific IgG serological response were also largely unaffected by TLR7, IRF7, or IFNAR1 deficiencies up to 7 mo after vaccination in all patients. These results suggest that induction of type I IFN is not required for efficient generation of a humoral response against SARS-CoV-2 by mRNA vaccines.


Asunto(s)
Linfocitos B , Vacunas contra la COVID-19 , COVID-19 , Interferón Tipo I , Humanos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Autoanticuerpos , COVID-19/inmunología , COVID-19/prevención & control , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética , Receptor Toll-Like 7/genética , Vacunación , Vacunas de ARNm , Vacunas contra la COVID-19/inmunología , Linfocitos B/inmunología , Interferón Tipo I/deficiencia
17.
Sci Adv ; 8(29): eabo0171, 2022 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-35857849

RESUMEN

Neurological manifestations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection represent a major issue in long coronavirus disease. How SARS-CoV-2 gains access to the brain and how infection leads to neurological symptoms are not clear because the principal means of viral entry by endocytosis, the angiotensin-converting enzyme 2 receptor, are barely detectable in the brain. We report that human neuronal cells, nonpermissive to infection through the endocytic pathway, can be infected when cocultured with permissive infected epithelial cells. SARS-CoV-2 induces the formation of tunneling nanotubes (TNTs) and exploits this route to spread to uninfected cells. In cellulo correlative fluorescence and cryo-electron tomography reveal that SARS-CoV-2 is associated with TNTs between permissive cells. Furthermore, multiple vesicular structures such as double-membrane vesicles, sites of viral replication, are observed inside TNTs between permissive and nonpermissive cells. Our data highlight a previously unknown mechanism of SARS-CoV-2 spreading, likely used as a route to invade nonpermissive cells and potentiate infection in permissive cells.

18.
PLoS One ; 17(2): e0262149, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35139078

RESUMEN

There is an urgent need for better diagnostic and analytical methods for vaccine research and infection control in virology. This has been highlighted by recently emerging viral epidemics and pandemics (Zika, SARS-CoV-2), and recurring viral outbreaks like the yellow fever outbreaks in Angola and the Democratic Republic of Congo (2016) and in Brazil (2016-2018). Current assays to determine neutralising activity against viral infections in sera are costly in time and equipment and suffer from high variability. Therefore, both basic infection research and diagnostic population screenings would benefit from improved methods to determine virus-neutralising activity in patient samples. Here we describe a robust, objective, and scalable Fluorescence Reduction Neutralisation Test (FluoRNT) for yellow fever virus, relying on flow cytometric detection of cells infected with a fluorescent Venus reporter containing variant of the yellow fever vaccine strain 17D (YF-17D-Venus). It accurately measures neutralising antibody titres in human serum samples within as little as 24 h. Samples from 32 vaccinees immunised with YF-17D were tested for neutralising activity by both a conventional focus reduction neutralisation test (FRNT) and FluoRNT. Both types of tests proved to be equally reliable for the detection of neutralising activity, however, FluoRNT is significantly more precise and reproducible with a greater dynamic range than conventional FRNT. The FluoRNT assay protocol is substantially faster, easier to control, and cheaper in per-assay costs. FluoRNT additionally reduces handling time minimising exposure of personnel to patient samples. FluoRNT thus brings a range of desirable features that can accelerate and standardise the measurement of neutralising anti-yellow fever virus antibodies. It could be used in applications ranging from vaccine testing to large cohort studies in systems virology and vaccinology. We also anticipate the potential to translate the methodology and analysis of FluoRNT to other flaviviruses such as West Nile, Dengue and Zika or to RNA viruses more generally.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Chlorocebus aethiops , Fluorescencia , Humanos , Pruebas de Neutralización/economía , Pruebas de Neutralización/métodos , Células Vero , Fiebre Amarilla/sangre , Fiebre Amarilla/virología
19.
Life Sci Alliance ; 5(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34996842

RESUMEN

The current COVID-19 pandemic is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The positive-sense single-stranded RNA virus contains a single linear RNA segment that serves as a template for transcription and replication, leading to the synthesis of positive and negative-stranded viral RNA (vRNA) in infected cells. Tools to visualize vRNA directly in infected cells are critical to analyze the viral replication cycle, screen for therapeutic molecules, or study infections in human tissue. Here, we report the design, validation, and initial application of FISH probes to visualize positive or negative RNA of SARS-CoV-2 (CoronaFISH). We demonstrate sensitive visualization of vRNA in African green monkey and several human cell lines, in patient samples and human tissue. We further demonstrate the adaptation of CoronaFISH probes to electron microscopy. We provide all required oligonucleotide sequences, source code to design the probes, and a detailed protocol. We hope that CoronaFISH will complement existing techniques for research on SARS-CoV-2 biology and COVID-19 pathophysiology, drug screening, and diagnostics.


Asunto(s)
COVID-19/diagnóstico , Hibridación Fluorescente in Situ/métodos , ARN Viral/genética , SARS-CoV-2/genética , Replicación Viral/genética , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Animales , Antivirales/farmacología , COVID-19/virología , Células CACO-2 , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Hibridación in Situ/métodos , Microscopía Electrónica/métodos , ARN Viral/ultraestructura , Reproducibilidad de los Resultados , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Sensibilidad y Especificidad , Células Vero , Liberación del Virus/efectos de los fármacos , Liberación del Virus/genética , Liberación del Virus/fisiología , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología , Tratamiento Farmacológico de COVID-19
20.
J Exp Med ; 202(9): 1179-84, 2005 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-16260489

RESUMEN

The yellow fever (YF) 17D vaccine is one of the most successful live attenuated vaccines available. A single immunization induces both long-lasting neutralizing antibody and YF-specific T cell responses. Surprisingly, the mechanism for this robust immunity has not been addressed. In light of several recent reports suggesting flavivirus interaction with dendritic cells (DCs), we investigated the mechanism of YF17D interaction with DCs and the importance of this interaction in generating T cell immunity. Our results show that YF17D can infect immature and mature human DCs. Viral entry is Ca(2+) dependent, but it is independent of DC-SIGN as well as multiple integrins expressed on the DC surface. Similar to infection of cell lines, YF infection of immature DCs is cytopathic. Although infection itself does not induce DC maturation in vitro, TNF-alpha-induced maturation protects DCs from YF-induced cytopathogenicity. Furthermore, we show that DCs infected with YF17D or YF17D carrying a recombinant epitope can process and present antigens for CD8(+) T cell stimulation. These findings offer insight into the immunologic mechanisms associated with the highly capable YF17D vaccine that may guide effective vaccine design.


Asunto(s)
Presentación de Antígeno/inmunología , Células Dendríticas/inmunología , Epítopos de Linfocito T/inmunología , Linfocitos T/inmunología , Vacuna contra la Fiebre Amarilla/inmunología , Apoptosis/fisiología , Calcio/fisiología , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/virología , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/metabolismo , Humanos , Linfocitos T/metabolismo , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/metabolismo , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo , Vacuna contra la Fiebre Amarilla/genética , Vacuna contra la Fiebre Amarilla/metabolismo , Virus de la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA