Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Pathog ; 18(7): e1010691, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35862475

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) marks the third novel ß-coronavirus to cause significant human mortality in the last two decades. Although vaccines are available, too few have been administered worldwide to keep the virus in check and to prevent mutations leading to immune escape. To determine if antibodies could be identified with universal coronavirus activity, plasma from convalescent subjects was screened for IgG against a stabilized pre-fusion SARS-CoV-2 spike S2 domain, which is highly conserved between human ß-coronavirus. From these subjects, several S2-specific human monoclonal antibodies (hmAbs) were developed that neutralized SARS-CoV-2 with recognition of all variants of concern (VoC) tested (Beta, Gamma, Delta, Epsilon, and Omicron). The hmAb 1249A8 emerged as the most potent and broad hmAb, able to recognize all human ß-coronavirus and neutralize SARS-CoV and MERS-CoV. 1249A8 demonstrated significant prophylactic activity in K18 hACE2 mice infected with SARS-CoV-2 lineage A and lineage B Beta, and Omicron VoC. 1249A8 delivered as a single 4 mg/kg intranasal (i.n.) dose to hamsters 12 hours following infection with SARS-CoV-2 Delta protected them from weight loss, with therapeutic activity further enhanced when combined with 1213H7, an S1-specific neutralizing hmAb. As little as 2 mg/kg of 1249A8 i.n. dose 12 hours following infection with SARS-CoV Urbani strain, protected hamsters from weight loss and significantly reduced upper and lower respiratory viral burden. These results indicate in vivo cooperativity between S1 and S2 specific neutralizing hmAbs and that potent universal coronavirus neutralizing mAbs with therapeutic potential can be induced in humans and can guide universal coronavirus vaccine development.


Asunto(s)
COVID-19 , Glicoproteína de la Espiga del Coronavirus , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales , COVID-19/terapia , Vacunas contra la COVID-19 , Humanos , Ratones , SARS-CoV-2 , Pérdida de Peso
2.
J Virol ; 95(22): e0112621, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34495697

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and has been responsible for the still ongoing coronavirus disease 2019 (COVID-19) pandemic. Prophylactic vaccines have been authorized by the U.S. Food and Drug Administration (FDA) for the prevention of COVID-19. Identification of SARS-CoV-2-neutralizing antibodies (NAbs) is important to assess vaccine protection efficacy, including their ability to protect against emerging SARS-CoV-2 variants of concern (VoC). Here, we report the generation and use of a recombinant (r)SARS-CoV-2 USA/WA1/2020 (WA-1) strain expressing Venus and an rSARS-CoV-2 strain expressing mCherry and containing mutations K417N, E484K, and N501Y found in the receptor binding domain (RBD) of the spike (S) glycoprotein of the South African (SA) B.1.351 (beta [ß]) VoC in bifluorescent-based assays to rapidly and accurately identify human monoclonal antibodies (hMAbs) able to neutralize both viral infections in vitro and in vivo. Importantly, our bifluorescent-based system accurately recapitulated findings observed using individual viruses. Moreover, fluorescent-expressing rSARS-CoV-2 strain and the parental wild-type (WT) rSARS-CoV-2 WA-1 strain had similar viral fitness in vitro, as well as similar virulence and pathogenicity in vivo in the K18 human angiotensin-converting enzyme 2 (hACE2) transgenic mouse model of SARS-CoV-2 infection. We demonstrate that these new fluorescent-expressing rSARS-CoV-2 can be used in vitro and in vivo to easily identify hMAbs that simultaneously neutralize different SARS-CoV-2 strains, including VoC, for the rapid assessment of vaccine efficacy or the identification of prophylactic and/or therapeutic broadly NAbs for the treatment of SARS-CoV-2 infection. IMPORTANCE SARS-CoV-2 is responsible of the COVID-19 pandemic that has warped daily routines and socioeconomics. There is still an urgent need for prophylactics and therapeutics to treat SARS-CoV-2 infections. In this study, we demonstrate the feasibility of using bifluorescent-based assays for the rapid identification of hMAbs with neutralizing activity against SARS-CoV-2, including VoC in vitro and in vivo. Importantly, results obtained with these bifluorescent-based assays recapitulate those observed with individual viruses, demonstrating their feasibility to rapidly advance our understanding of vaccine efficacy and to identify broadly protective human NAbs for the therapeutic treatment of SARS-CoV-2.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Pruebas de Neutralización/métodos , SARS-CoV-2/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos ampliamente neutralizantes/inmunología , Anticuerpos ampliamente neutralizantes/uso terapéutico , COVID-19/terapia , COVID-19/virología , Genes Reporteros , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Pulmón/efectos de los fármacos , Pulmón/virología , Ratones , Mutación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
3.
bioRxiv ; 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38746414

RESUMEN

SARS-CoV-2 continues to be a public health burden, driven in-part by its continued antigenic diversification and resulting emergence of new variants. While increasing herd immunity, current vaccines, and therapeutics have improved outcomes for some; prophylactic and treatment interventions that are not compromised by viral evolution of the Spike protein are still needed. Using a rationally designed SARS-CoV-2 Receptor Binding Domain (RBD) - ACE2 fusion protein and differential selection process with native Omicron RBD protein, we developed a recombinant human monoclonal antibody (hmAb) from a convalescent individual following SARS-CoV-2 Omicron infection. The resulting hmAb, 1301B7 potently neutralized a wide range of SARS-CoV-2 variants including the original Wuhan and more recent Omicron JN.1 strain, as well as SARS-CoV. Structure determination of the SARS-CoV-2 EG5.1 Spike/1301B7 Fab complex by cryo-electron microscopy at 3.1Å resolution demonstrates 1301B7 contacts the ACE2 binding site of RBD exclusively through its VH1-69 heavy chain, making contacts using CDRs1-3, as well as framework region 3 (FR3). Broad specificity is achieved through 1301B7 binding to many conserved residues of Omicron variants including Y501 and H505. Consistent with its extensive binding epitope, 1301B7 is able to potently diminish viral burden in the upper and lower respiratory tract and protect mice from challenge with Omicron XBB1.5 and Omicron JN.1 viruses. These results suggest 1301B7 has broad potential to prevent or treat clinical SARS-CoV-2 infections and to guide development of RBD-based universal SARS-CoV-2 prophylactic vaccines and therapeutic approaches.

4.
Avian Dis ; 57(2 Suppl): 503-8, 2013 06.
Artículo en Inglés | MEDLINE | ID: mdl-23901768

RESUMEN

The role of pp38 in the pathogenesis of Marek's disease (MD) has not been fully elucidated. Previously, we reported the presence of two splice variants (Spl A and Spl B) for pp38. We also reported that the wild-type pp38 (WT), as well as the mutated pp38 (MUT), altered the oxidative phosphorylation pathway in infected cells. To determine whether the different forms of pp38 are important for the pathogenesis of MD, we generated RB-1B-based bacterial artificial chromosome (BAC) clones expressing pp38MUT, pp38Sp1 A, and pp38Spl B. Infectious viruses were recovered from these BAC clones in chick kidney cells (CKC). The Spl A and Spl B viruses had significantly smaller plaque sizes and replicated to a lesser degree in CKC than the WT and MUT viruses. Two in vivo experiments were conducted by inoculating 7-day-old P2a chicks with 1000 plaque-forming units of each virus. In the first experiment, chicks were sacrificed at 4, 8, 11, and 15 days postinfection (PI). WT and MUT viruses had similar viremia levels using virus isolation and quantitative real-time PCR (qPCR) assays, whereas Spl A and Spl B viruses had significantly lower viremia levels than WT and MUT viruses. In the second experiment, we showed that tumor development and MD mortality were similar in the WT- and MUT-infected chickens, with all birds MD positive at 5 wk PI. In contrast, chickens infected with Spl B and Spl A had a significantly lower MD incidence at 11 wk PI, when the experiment was terminated.


Asunto(s)
Transformación Celular Neoplásica , Pollos , Mardivirus/genética , Mardivirus/patogenicidad , Enfermedad de Marek/inmunología , Fosfoproteínas/metabolismo , Proteínas Virales/metabolismo , Animales , Transformación Celular Neoplásica/inmunología , Células Cultivadas , Embrión de Pollo , Cromosomas Artificiales Bacterianos/genética , Mardivirus/metabolismo , Enfermedad de Marek/virología , Fosfoproteínas/genética , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Recombinación Genética , Organismos Libres de Patógenos Específicos , Proteínas Virales/genética
5.
Viruses ; 15(7)2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37515226

RESUMEN

Influenza B virus (IBV) contributes to substantial influenza-mediated morbidity and mortality, particularly among children. Similar to influenza A viruses (IAV), the hemagglutinin (HA) and neuraminidase (NA) of IBV undergo antigenic drift, necessitating regular reformulation of seasonal influenza vaccines. NA inhibitors, such as oseltamivir, have reduced activity and clinical efficacy against IBV, while M2 channel inhibitors are only effective against IAV, highlighting the need for improved vaccine and therapeutics for the treatment of seasonal IBV infections. We have previously described a potent human monoclonal antibody (hMAb), 1092D4, that is specific for IBV NA and neutralizes a broad range of IBVs. The anti-viral activity of MAbs can include direct mechanisms such as through neutralization and/or Fc-mediated effector functions that are dependent on accessory cells expressing Fc receptors and that could be impacted by potential host-dependent variability. To discern if the in vivo efficacy of 1092D4 was dependent on Fc-effector function, 1092D4 hMAb with reduced ability to bind to Fc receptors (1092D4-LALAPG) was generated and tested. 1092D4-LALAPG had comparable in vitro binding, neutralization, and inhibition of NA activity to 1092D4. 1092D4-LALAPG was effective at protecting against a lethal challenge of IBV in mice. These results suggest that hMAb 1092D4 in vivo activity is minimally dependent on Fc-effector functions, a characteristic that may extend to other hMAbs that have potent NA inhibition activity.


Asunto(s)
Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Niño , Animales , Ratones , Humanos , Anticuerpos ampliamente neutralizantes , Neuraminidasa , Anticuerpos Antivirales , Virus de la Influenza B , Anticuerpos Monoclonales/farmacología , Receptores Fc , Glicoproteínas Hemaglutininas del Virus de la Influenza
6.
AIDS Res Hum Retroviruses ; 39(7): 350-366, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36762930

RESUMEN

The most potent and broad HIV envelope (Env)-specific antibodies often when reverted to their inferred germline versions representing the naive B cell receptor, fail to bind Env, suggesting that the initial responding B cell population not only exclusively comprises a naive population, but also a pre-existing cross-reactive antigen-experienced B cell pool that expands following Env exposure. Previously we isolated gp120-reactive monoclonal antibodies (mAbs) from participants in HVTN 105, an HIV vaccine trial. Using deep sequencing, focused on immunoglobulin G (IgG), IgA, and IgM, VH-lineage tracking, we identified four of these mAb lineages in pre-immune peripheral blood. We also looked through the ∼7 month postvaccination bone marrow, and interestingly, several of these lineages that were found in prevaccination blood were still persistent in the postvaccination bone marrow, including the CD138+ long-lived plasma cell compartment. The majority of the pre-immune lineage members included IgM, however, IgG and IgA members were also prevalent and exhibited somatic hypermutation. These results suggest that vaccine-induced gp120-specific antibody lineages originate from both naive and cross-reactive memory B cells. ClinicalTrials.gov NCT02207920.


Asunto(s)
Vacunas contra el SIDA , Infecciones por VIH , VIH-1 , Humanos , Infecciones por VIH/prevención & control , Anticuerpos Anti-VIH , Vacunación , Proteína gp120 de Envoltorio del VIH , Inmunoglobulina G , Anticuerpos Monoclonales , Inmunoglobulina A , Inmunoglobulina M , Anticuerpos Neutralizantes
7.
Microbiol Spectr ; 11(4): e0472822, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37318331

RESUMEN

Due to antigenic drift and shift of influenza A viruses (IAV) and the tendency to elicit predominantly strain-specific antibodies, humanity remains susceptible to new strains of seasonal IAV and is at risk from viruses with pandemic potential for which limited or no immunity may exist. The genetic drift of H3N2 IAV is specifically pronounced, resulting in two distinct clades since 2014. Here, we demonstrate that immunization with a seasonal inactivated influenza vaccine (IIV) results in increased levels of H3N2 IAV-specific serum antibodies against hemagglutinin (HA) and neuraminidase (NA). Detailed analysis of the H3N2 B cell response indicated expansion of H3N2-specific peripheral blood plasmablasts 7 days after IIV immunization which expressed monoclonal antibodies (MAbs) with broad and potent antiviral activity against many H3N2 IAV strains as well as prophylactic and therapeutic activity in mice. These H3N2-specific B cell clonal lineages persisted in CD138+ long-lived bone marrow plasma cells. These results demonstrate that IIV-induced H3N2 human MAbs can protect and treat influenza virus infection in vivo and suggest that IIV can induce a subset of IAV H3N2-specific B cells with broad protective potential, a feature that warrants further study for universal influenza vaccine development. IMPORTANCE Influenza A virus (IAV) infections continue to cause substantial morbidity and mortality despite the availability of seasonal vaccines. The extensive genetic variability in seasonal and potentially pandemic influenza strains necessitates new vaccine strategies that can induce universal protection by focusing the immune response on generating protective antibodies against conserved targets within the influenza virus hemagglutinin and neuraminidase proteins. We have demonstrated that seasonal immunization with inactivated influenza vaccine (IIV) stimulates H3N2-specific monoclonal antibodies in humans that are broad and potent in their neutralization of virus in vitro. These antibodies also provide protection from H3N2 IAV in a mouse model of infection. Furthermore, they persist in the bone marrow, where they are expressed by long-lived antibody-producing plasma cells. This significantly demonstrates that seasonal IIV can induce a subset of H3N2-specific B cells with broad protective potential, a process that if further studied and enhanced could aid in the development of a universal influenza vaccine.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Humanos , Animales , Ratones , Gripe Humana/prevención & control , Vacunas contra la Influenza/genética , Hemaglutininas , Subtipo H3N2 del Virus de la Influenza A/genética , Neuraminidasa , Anticuerpos Monoclonales , Subtipo H1N1 del Virus de la Influenza A/genética , Anticuerpos Antivirales , Virus de la Influenza A/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética
8.
bioRxiv ; 2022 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-35291292

RESUMEN

Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) marks the third novel ß-coronavirus to cause significant human mortality in the last two decades. Although vaccines are available, too few have been administered worldwide to keep the virus in check and to prevent mutations leading to immune escape. To determine if antibodies could be identified with universal coronavirus activity, plasma from convalescent subjects was screened for IgG against a stabilized pre-fusion SARS-CoV-2 spike S2 domain, which is highly conserved between human ß-coronavirus. From these subjects, several S2-specific human monoclonal antibodies (hmAbs) were developed that neutralized SARS-CoV-2 with recognition of all variants of concern (VoC) tested (Beta, Gamma, Delta, Epsilon, and Omicron). The hmAb 1249A8 emerged as the most potent and broad hmAb, able to recognize all human ß-coronavirus and neutralize SARS-CoV and MERS-CoV. 1249A8 demonstrated significant prophylactic activity in K18 hACE2 mice infected with SARS-CoV-2 lineage A and lineage B Beta, and Omicron VoC. 1249A8 delivered as a single 4 mg/kg intranasal (i.n.) dose to hamsters 12 hours following infection with SARS-CoV-2 Delta protected them from weight loss, with therapeutic activity further enhanced when combined with 1213H7, an S1-specific neutralizing hmAb. As little as 2 mg/kg of 1249A8 i.n. dose 12 hours following infection with SARS-CoV Urbani strain, protected hamsters from weight loss and significantly reduced upper and lower respiratory viral burden. These results indicate in vivo cooperativity between S1 and S2 specific neutralizing hmAbs and that potent universal coronavirus neutralizing mAbs with therapeutic potential can be induced in humans and can guide universal coronavirus vaccine development.

9.
Avian Dis ; 55(1): 90-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21500642

RESUMEN

Infection of maternal, antibody-negative chickens with chicken infectious anemia virus (CIAV) can cause clinical disease, while infection after maternal antibodies wane often results in subclinical infection and immunosuppression. Currently, vaccines are not available for vaccination in ovo or in newly hatched chickens. Development of CIAV vaccines for in ovo use depends on the ability to generate vaccines that do not cause lesions in newly hatched chicks and that can induce an immune response regardless of maternal immunity. Immune complex (IC) vaccines have been successfully used for control of infectious bursal disease, and we used a similar approach to determine if an IC vaccine is feasible for CIAV. Immune complexes were prepared that consisted of 0.1 ml containing 10(5.4) tissue culture infective dose 50% of CIA-1 and 0.1 ml containing 10 to 160 neutralizing units (IC Positive [ICP]10 to ICP160), in which one neutralizing unit is the reciprocal of the serum dilution required to protect 50% of CU147 cells from the cytopathic effects caused by CIA-1. Virus replication was delayed comparing ICP80 and ICP160 with combinations using negative serum (IC Negative [ICN]80 or ICN160). In addition, the number of birds with hematocrit values <28% were decreased with ICP80 or ICP160 compared to ICN80 or ICN160. Seroconversion was delayed in ICP80 and ICP160 groups. To determine if ICP80 or ICN 160 protected against challenge, we vaccinated maternal, antibody-free birds at 1 day of age and challenged at 2 wk or 3 wk of age with the 01-4201 strain. Both ICP80 and ICP160 protected against replication of the challenge virus, which was measured using differential quantitative PCR with primers distinguishing between the two isolates. Thus, in principle, immune complex vaccines may offer a method to protect newly hatched chicks against challenge with field virus. However, additional studies using maternal, antibody-positive chicks in combination with in ovo vaccination will be needed to determine if immune complex vaccines will be useful to protect commercial chickens.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Virus de la Anemia del Pollo/inmunología , Pollos , Infecciones por Circoviridae/veterinaria , Vacunas Virales/inmunología , Animales , Infecciones por Circoviridae/prevención & control , Organismos Libres de Patógenos Específicos
10.
Cell Rep Med ; 2(3): 100218, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33649747

RESUMEN

SARS-CoV-2 infection results in viral burden in the respiratory tract, enabling transmission and leading to substantial lung pathology. The 1212C2 fully human monoclonal antibody was derived from an IgM memory B cell of a COVID-19 patient, has high affinity for the Spike protein receptor binding domain, neutralizes SARS-CoV-2, and exhibits in vivo prophylactic and therapeutic activity in hamsters when delivered intraperitoneally, reducing upper and lower respiratory viral burden and lung pathology. Inhalation of nebulized 1212C2 at levels as low as 0.6 mg/kg, corresponding to 0.03 mg/kg lung-deposited dose, reduced the viral burden below the detection limit and mitigated lung pathology. The therapeutic efficacy of an exceedingly low dose of inhaled 1212C2 supports the rationale for local lung delivery for dose-sparing benefits, as compared to the conventional parenteral route of administration. These results suggest that the clinical development of 1212C2 formulated and delivered via inhalation for the treatment of SARS-CoV-2 infection should be considered.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Administración por Inhalación , Animales , Anticuerpos Monoclonales/clasificación , Anticuerpos Monoclonales/inmunología , COVID-19/virología , Cricetinae , Modelos Animales de Enfermedad , Mapeo Epitopo , Epítopos/inmunología , Femenino , Humanos , Inmunoglobulina M/inmunología , Masculino , Células B de Memoria/citología , Células B de Memoria/metabolismo , Persona de Mediana Edad , Pruebas de Neutralización , Filogenia , Dominios Proteicos/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo
11.
Viruses ; 12(2)2020 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-32024281

RESUMEN

Although seasonal influenza vaccines block most predominant influenza types and subtypes, humans still remain vulnerable to waves of seasonal and new potential pandemic influenza viruses for which no immunity may exist because of viral antigenic drift and/or shift. Previously, we described a human monoclonal antibody (hMAb), KPF1, which was produced in human embryonic kidney 293T cells (KPF1-HEK) with broad and potent neutralizing activity against H1N1 influenza A viruses (IAV) in vitro, and prophylactic and therapeutic activities in vivo. In this study, we produced hMAb KPF1 in tobacco plants (KPF1-Antx) and demonstrated how the plant-produced KPF1-Antx hMAb possesses similar biological activity compared with the mammalian-produced KPF1-HEK hMAb. KPF1-Antx hMAb showed broad binding to recombinant HA proteins and H1N1 IAV, including A/California/04/2009 (pH1N1) in vitro, which was comparable to that observed with KPF1-HEK hMAb. Importantly, prophylactic administration of KPF1-Antx hMAb to guinea pigs prevented pH1N1 infection and transmission in both prophylactic and therapeutic experiments, substantiating its clinical potential to prevent and treat H1N1 infections. Collectively, this study demonstrated, for the first time, a plant-produced influenza hMAb with in vitro and in vivo activity against influenza virus. Because of the many advantages of plant-produced hMAbs, such as rapid batch production, low cost, and the absence of mammalian cell products, they represent an alternative strategy for the production of immunotherapeutics for the treatment of influenza viral infections, including emerging seasonal and/or pandemic strains.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/transmisión , Planticuerpos/uso terapéutico , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Antivirales/biosíntesis , Femenino , Cobayas , Infecciones por Orthomyxoviridae/terapia , Nicotiana/metabolismo
12.
Sci Rep ; 10(1): 13031, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32747654

RESUMEN

Efficacious HIV-1 vaccination requires elicitation of long-lived antibody responses. However, our understanding of how different vaccine types elicit durable antibody responses is lacking. To assess the impact of vaccine type on antibody responses, we measured IgG isotypes against four consensus HIV antigens from 2 weeks to 10 years post HIV-1 vaccination and used mixed effects models to estimate half-life of responses in four human clinical trials. Compared to protein-boosted regimens, half-lives of gp120-specific antibodies were longer but peak magnitudes were lower in Modified Vaccinia Ankara (MVA)-boosted regimens. Furthermore, gp120-specific B cell transcriptomics from MVA-boosted and protein-boosted vaccines revealed a distinct signature at a peak (2 weeks after last vaccination) including CD19, CD40, and FCRL2-5 activation along with increased B cell receptor signaling. Additional analysis revealed contributions of RIG-I-like receptor pathway and genes such as SMAD5 and IL-32 to antibody durability. Thus, this study provides novel insights into vaccine induced antibody durability and B-cell receptor signaling.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos B/inmunología , Perfilación de la Expresión Génica , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/genética , Infecciones por VIH/inmunología , VIH-1/inmunología , Formación de Anticuerpos/inmunología , Ensayos Clínicos como Asunto , Regulación de la Expresión Génica , Semivida , Humanos , Inmunización Secundaria , Modelos Lineales , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Vacunación , Virus Vaccinia/inmunología
13.
Cell Rep Med ; 1(2)2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32577626

RESUMEN

Induction of persistent HIV-1 Envelope (Env) specific antibody (Ab) is a primary goal of HIV vaccine strategies; however, it is unclear whether HIV Env immunization in humans induces bone marrow plasma cells, the presumed source of long-lived systemic Ab. To define the features of Env-specific plasma cells after vaccination, samples were obtained from HVTN 105, a phase I trial testing the same gp120 protein immunogen, AIDSVAX B/E, used in RV144, along with a DNA immunogen in various prime and boost strategies. Boosting regimens that included AIDSVAX B/E induced robust peripheral blood plasmablast responses. The Env-specific immunoglobulin repertoire of the plasmablasts is dominated by VH1 gene usage and targeting of the V3 region. Numerous plasmablast-derived immunoglobulin lineages persisted in the bone marrow >8 months after immunization, including in the CD138+ long-lived plasma cell compartment. These findings identify a cellular linkage for the development of sustained Env-specific Abs following vaccination in humans.


Asunto(s)
Vacunas contra el SIDA/uso terapéutico , Infecciones por VIH/prevención & control , Células Plasmáticas/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Especificidad de Anticuerpos , Linaje de la Célula/inmunología , Supervivencia Celular/inmunología , Células Cultivadas , Células HEK293 , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/fisiología , Humanos , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Células Plasmáticas/virología , Células THP-1 , Vacunación
14.
Virus Genes ; 39(1): 102-12, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19472043

RESUMEN

Two splice variants of the Marek's disease virus phosphorylated polypeptide (pp)38 were previously identified in the quail cell line QTP32 expressing pp38 under the control of an inducible promoter. We developed QT35-derived cell lines expressing these splice variants or full length pp38 with the splice acceptor sites mutated to further elucidate the role of pp38. Only induction of full length pp38 resulted in an increase in mitochondrial succinate dehydrogenase activity compared to non-induced cells. Transcript copy numbers of cytochrome C oxidase subunit I and ATP synthase were reduced in induced cells. The ATP content of isolated mitochondria from induced cells was greatly reduced compared to those of non-induced cells. Mitochondrial and pp38 staining suggests that there is no direct interaction between pp38 and the mitochondria. Mitochondrial transcripts were also reduced in DF-1 cells expressing full length pp38 and in MDV-infected chick kidney cells indicating that this effect occurs independent of other viral genes and after in vitro infection with MDV.


Asunto(s)
Antígenos Virales/fisiología , Complejo IV de Transporte de Electrones/biosíntesis , Interacciones Huésped-Patógeno , Mardivirus/fisiología , Proteínas Mitocondriales/biosíntesis , Fosfoproteínas/fisiología , Succinato Deshidrogenasa/biosíntesis , Transcripción Genética , Adenosina Trifosfato/análisis , Animales , Línea Celular , Pollos , Transporte de Electrón , Perfilación de la Expresión Génica , Mitocondrias/química , Datos de Secuencia Molecular , Fosforilación Oxidativa , Codorniz , Análisis de Secuencia de ADN
15.
PLoS One ; 14(9): e0221550, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31504041

RESUMEN

HIV envelope protein (Env) is the sole target of broadly neutralizing antibodies (BNAbs) that are capable of neutralizing diverse strains of HIV. While BNAbs develop spontaneously in a subset of HIV-infected patients, efforts to design an envelope protein-based immunogen to elicit broadly neutralizing antibody responses have so far been unsuccessful. It is hypothesized that a primary barrier to eliciting BNAbs is the fact that HIV envelope proteins bind poorly to the germline-encoded unmutated common ancestor (UCA) precursors to BNAbs. To identify variant forms of Env with increased affinities for the UCA forms of BNAbs 4E10 and 10E8, which target the Membrane Proximal External Region (MPER) of Env, libraries of randomly mutated Env variants were expressed in a yeast surface display system and screened using fluorescence activated cell sorting for cells displaying variants with enhanced abilities to bind the UCA antibodies. Based on analyses of individual clones obtained from the screen and on next-generation sequencing of sorted libraries, distinct but partially overlapping sets of amino acid substitutions conferring enhanced UCA antibody binding were identified. These were particularly enriched in substitutions of arginine for highly conserved tryptophan residues. The UCA-binding variants also generally exhibited enhanced binding to the mature forms of anti-MPER antibodies. Mapping of the identified substitutions into available structures of Env suggest that they may act by destabilizing both the initial pre-fusion conformation and the six-helix bundle involved in fusion of the viral and cell membranes, as well as providing new or expanded epitopes with increased accessibility for the UCA antibodies.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Afinidad de Anticuerpos , Proteína gp41 de Envoltorio del VIH/inmunología , Mutación , Precursores de Proteínas/inmunología , Anticuerpos Antivirales/inmunología , Epítopos/química , Epítopos/genética , Epítopos/inmunología , Proteína gp41 de Envoltorio del VIH/química , Proteína gp41 de Envoltorio del VIH/genética , Unión Proteica , Precursores de Proteínas/química , Precursores de Proteínas/genética , Estabilidad Proteica
16.
mBio ; 10(2)2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862743

RESUMEN

Although most seasonal inactivated influenza vaccines (IIV) contain neuraminidase (NA), the extent and mechanisms of action of protective human NA-specific humoral responses induced by vaccination are poorly resolved. Due to the propensity of influenza virus for antigenic drift and shift and its tendency to elicit predominantly strain-specific antibodies, humanity remains susceptible to waves of new strains of seasonal viruses and is at risk from viruses with pandemic potential for which limited or no immunity may exist. Here we demonstrate that the use of IIV results in increased levels of influenza B virus (IBV) NA-specific serum antibodies. Detailed analysis of the IBV NA B cell response indicates concurrent expansion of IBV NA-specific peripheral blood plasmablasts 7 days after IIV immunization which express monoclonal antibodies with broad and potent antiviral activity against both IBV Victoria and Yamagata lineages and prophylactic and therapeutic activity in mice. These IBV NA-specific B cell clonal lineages persisted in CD138+ long-lived bone marrow plasma cells. These results represent the first demonstration that IIV-induced NA human antibodies can protect and treat influenza virus infection in vivo and suggest that IIV can induce a subset of IBV NA-specific B cells with broad protective potential, a feature that warrants further study for universal influenza vaccine development.IMPORTANCE Influenza virus infections continue to cause substantial morbidity and mortality despite the availability of seasonal vaccines. The extensive genetic variability in seasonal and potentially pandemic influenza strains necessitates new vaccine strategies that can induce universal protection by focusing the immune response on generating protective antibodies against conserved targets such as regions within the influenza neuraminidase protein. We have demonstrated that seasonal immunization stimulates neuraminidase-specific antibodies in humans that are broad and potent in their protection from influenza B virus when tested in mice. These antibodies further persist in the bone marrow, where they are expressed by long-lived antibody-producing cells, referred to here as plasma cells. The significance in our research is the demonstration that seasonal influenza immunization can induce a subset of neuraminidase-specific B cells with broad protective potential, a process that if further studied and enhanced could aid in the development of a universal influenza vaccine.


Asunto(s)
Anticuerpos Antivirales/sangre , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Neuraminidasa/inmunología , Células Plasmáticas/inmunología , Proteínas Virales/inmunología , Animales , Anticuerpos Monoclonales/sangre , Protección Cruzada , Modelos Animales de Enfermedad , Voluntarios Sanos , Humanos , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/terapia , Ratones , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/terapia , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/inmunología
17.
Vaccine ; 37(17): 2322-2330, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30926296

RESUMEN

Induction of a sustained and broad antibody (Ab) response is a major goal in developing a protective HIV-1 vaccine. DNA priming alone shows reduced levels of immunogenicity; however, when combined with protein boosting is an attractive vaccination strategy for induction of humoral responses. Using the VC10014 DNA and protein-based vaccine consisting of HIV-1 envelope (Env) gp160 plasmids and trimeric gp140 proteins derived from an HIV-1 clade B infected subject who developed broadly neutralizing serum Abs, and which has been previously demonstrated to induce Tier 2 heterologous neutralizing Abs in rhesus macaques, we evaluated whether MPLA and IL-33 when administered during the DNA priming phase enhances the humoral response in mice. The addition of IL-33 during the gp160 DNA priming phase resulted in high titer gp120-specific plasma IgG after the first immunization. The IL-33 treated mice had higher plasma IgG Ab avidity, breadth, and durability after DNA and protein co-immunization with alum adjuvant as compared to MPLA and alum only treated mice. IL-33 was also associated with a significant IgM Env-specific response and expansion of peritoneal and splenic B-1b B cells. These results indicate that DNA priming in the presence of exogenous IL-33 qualitatively alters the HIV-1 Env-specific humoral response, improving the kinetics and breadth of potentially protective Ab.


Asunto(s)
Vacunas contra el SIDA/inmunología , Formación de Anticuerpos/inmunología , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , VIH-1/inmunología , Interleucina-33/metabolismo , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA/genética , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Modelos Animales de Enfermedad , Femenino , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/virología , Humanos , Inmunización , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Ratones , Vacunas de ADN/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética
19.
Hum Exp Toxicol ; 27(2): 129-34, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18480137

RESUMEN

The developing immune system serves as a novel target for disruption by environmental chemicals and drugs, and one that can significantly influence later-life health risks. Specific immune maturational events occur during critical windows of pre- and early postnatal development that are not effectively modeled using adult exposure-assessment or general developmental toxicity screens. The range of postnatal health risks linked to developmental immunotoxicity (DIT) is influenced, in part, by the natural progression of prenatal-neonatal development. In this progression, the pregnancy itself imposes a Th2-bias in utero, and this produces a delay in the acquisition of Th1 functional capacity in the newborn. The status of Th1 regulatory and Th17 populations may also be important in immune function/dysfunction considerations. The necessary shift from a Th2 preferred capacity in late gestation to a more balance functional capacity in the neonate can be disrupted by xenobiotics leaving the child with increased vulnerability to a range of potential diseases. Knowledge of environmental factors that facilitate effective immune functional maturation as well as those xenobiotics capable of disrupting the process is important in strategies to reduce the incidence of diseases such as childhood asthma. Because hormesis has been shown to be an important factor in modulation of the adult immune system, it becomes even more important to understand potentially opposing dose-response effects for the immune system of the fetus, neonate, and juvenile. The direct linkage between immune dysfunction and chronic disease has become abundantly apparent in recent years. Therefore, a more comprehensive and effective approach for the protection of the developing immune system can help to reduce the incidence of later-life chronic diseases.


Asunto(s)
Relación Dosis-Respuesta a Droga , Exposición a Riesgos Ambientales/efectos adversos , Enfermedades del Sistema Inmune/etiología , Sistema Inmunológico , Efectos Tardíos de la Exposición Prenatal , Xenobióticos/efectos adversos , Adulto , Autoinmunidad , Femenino , Humanos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/embriología , Sistema Inmunológico/crecimiento & desarrollo , Recién Nacido , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/fisiopatología
20.
PLoS One ; 13(10): e0205756, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30335821

RESUMEN

As a step toward the development of variant forms of Env with enhanced immunogenic properties, we have expressed the glycoprotein in the yeast surface display system in a form that can be subjected to random mutagenesis followed by screening for forms with enhanced binding to germline antibodies. To optimize the expression and immunogenicity of the yeast-displayed Env protein, we tested different approaches for cell wall anchoring, expression of gp120 and gp140 Env from different viral strains, the effects of introducing mutations designed to stabilize Env, and the effects of procedures for altering N-linked glycosylation of Env. We find that diverse forms of HIV envelope glycoprotein can be efficiently expressed at the yeast cell surface and that gp140 forms of Env are effectively cleaved by Kex2p, the yeast furin protease homolog. Multiple yeast-displayed gp120 and gp140 proteins are capable of binding to antibodies directed against the V3-variable loop, CD4 binding site, and gp41 membrane-proximal regions, including some antibodies whose binding is known to depend on Env conformation and N-linked glycan. Based on antibody recognition and sensitivity to glycosidases, yeast glycosylation patterns partially mimic high mannose-type N-glycosylation in mammalian cells. However, yeast-displayed Env is not recognized by some anti-Env antibodies sensitive to quaternary structure, suggesting either that the displayed protein exists in a monomeric state or that for these antibodies, yeast glycosylation in certain regions hinders recognition or access. Consistent with studies in other systems, reconstructed predicted unmutated precursors to anti-Env antibodies exhibit little affinity for the yeast-displayed envelope protein.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Saccharomyces cerevisiae/virología , Glicosilación , Células HEK293 , Proteína gp120 de Envoltorio del VIH/genética , Proteína gp120 de Envoltorio del VIH/inmunología , Proteína gp120 de Envoltorio del VIH/metabolismo , VIH-1/genética , VIH-1/metabolismo , Humanos , Inmunogenicidad Vacunal/genética , Inmunogenicidad Vacunal/inmunología , Técnicas Inmunológicas/métodos , Mutagénesis Sitio-Dirigida , Mutación , Proproteína Convertasas/metabolismo , Unión Proteica/inmunología , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA