Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Nat Commun ; 15(1): 4629, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38821950

RESUMEN

The Paramyxoviridae family encompasses medically significant RNA viruses, including human respiroviruses 1 and 3 (RV1, RV3), and zoonotic pathogens like Nipah virus (NiV). RV3, previously known as parainfluenza type 3, for which no vaccines or antivirals have been approved, causes respiratory tract infections in vulnerable populations. The RV3 fusion (F) protein is inherently metastable and will likely require prefusion (preF) stabilization for vaccine effectiveness. Here we used structure-based design to stabilize regions involved in structural transformation to generate a preF protein vaccine antigen with high expression and stability, and which, by stabilizing the coiled-coil stem region, does not require a heterologous trimerization domain. The preF candidate induces strong neutralizing antibody responses in both female naïve and pre-exposed mice and provides protection in a cotton rat challenge model (female). Despite the evolutionary distance of paramyxovirus F proteins, their structural transformation and local regions of instability are conserved, which allows successful transfer of stabilizing substitutions to the distant preF proteins of RV1 and NiV. This work presents a successful vaccine antigen design for RV3 and provides a toolbox for future paramyxovirus vaccine design and pandemic preparedness.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Sigmodontinae , Proteínas Virales de Fusión , Vacunas Virales , Animales , Femenino , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/química , Ratones , Vacunas Virales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Humanos , Ratones Endogámicos BALB C , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/virología , Virus de la Parainfluenza 3 Humana/inmunología , Virus de la Parainfluenza 3 Humana/genética
2.
Nature ; 626(7998): 392-400, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38086420

RESUMEN

An ideal vaccine both attenuates virus growth and disease in infected individuals and reduces the spread of infections in the population, thereby generating herd immunity. Although this strategy has proved successful by generating humoral immunity to measles, yellow fever and polio, many respiratory viruses evolve to evade pre-existing antibodies1. One approach for improving the breadth of antiviral immunity against escape variants is through the generation of memory T cells in the respiratory tract, which are positioned to respond rapidly to respiratory virus infections2-6. However, it is unknown whether memory T cells alone can effectively surveil the respiratory tract to the extent that they eliminate or greatly reduce viral transmission following exposure of an individual to infection. Here we use a mouse model of natural parainfluenza virus transmission to quantify the extent to which memory CD8+ T cells resident in the respiratory tract can provide herd immunity by reducing both the susceptibility of acquiring infection and the extent of transmission, even in the absence of virus-specific antibodies. We demonstrate that protection by resident memory CD8+ T cells requires the antiviral cytokine interferon-γ (IFNγ) and leads to altered transcriptional programming of epithelial cells within the respiratory tract. These results suggest that tissue-resident CD8+ T cells in the respiratory tract can have important roles in protecting the host against viral disease and limiting viral spread throughout the population.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Células T de Memoria , Infecciones por Paramyxoviridae , Sistema Respiratorio , Animales , Ratones , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Inmunidad Colectiva/inmunología , Memoria Inmunológica/inmunología , Interferón gamma/inmunología , Células T de Memoria/inmunología , Paramyxoviridae/inmunología , Paramyxoviridae/fisiología , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/transmisión , Infecciones por Paramyxoviridae/virología , Sistema Respiratorio/citología , Sistema Respiratorio/inmunología , Sistema Respiratorio/virología , Transcripción Genética , Humanos
3.
Nat Commun ; 14(1): 798, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36781872

RESUMEN

Respiratory syncytial virus (RSV), human metapneumovirus (HMPV), and human parainfluenza virus types one (HPIV1) and three (HPIV3) can cause severe disease and death in immunocompromised patients, the elderly, and those with underlying lung disease. A protective monoclonal antibody exists for RSV, but clinical use is limited to high-risk infant populations. Hence, therapeutic options for these viruses in vulnerable patient populations are currently limited. Here, we present the discovery, in vitro characterization, and in vivo efficacy testing of two cross-neutralizing monoclonal antibodies, one targeting both HPIV3 and HPIV1 and the other targeting both RSV and HMPV. The 3 × 1 antibody is capable of targeting multiple parainfluenza viruses; the MxR antibody shares features with other previously reported monoclonal antibodies that are capable of neutralizing both RSV and HMPV. We obtained structures using cryo-electron microscopy of these antibodies in complex with their antigens at 3.62 Å resolution for 3 × 1 bound to HPIV3 and at 2.24 Å for MxR bound to RSV, providing a structural basis for in vitro binding and neutralization. Together, a cocktail of 3 × 1 and MxR could have clinical utility in providing broad protection against four of the respiratory viruses that cause significant morbidity and mortality in at-risk individuals.


Asunto(s)
Metapneumovirus , Infecciones por Paramyxoviridae , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Humanos , Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Microscopía por Crioelectrón , Infecciones por Paramyxoviridae/prevención & control , Proteínas Virales de Fusión , Protección Cruzada
4.
Proc Natl Acad Sci U S A ; 119(25): e2203326119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35696580

RESUMEN

Human metapneumovirus (hMPV) is a leading cause of morbidity and hospitalization among children worldwide, however, no vaccines or therapeutics are currently available for hMPV disease prevention and treatment. The hMPV fusion (F) protein is the sole target of neutralizing antibodies. To map the immunodominant epitopes on the hMPV F protein, we isolated a panel of human monoclonal antibodies (mAbs), and the mAbs were assessed for binding avidity, neutralization potency, and epitope specificity. We found the majority of the mAbs target diverse epitopes on the hMPV F protein, and we discovered multiple mAb binding approaches for antigenic site III. The most potent mAb, MPV467, which had picomolar potency, was examined in prophylactic and therapeutic mouse challenge studies, and MPV467 limited virus replication in mouse lungs when administered 24 h before or 72 h after viral infection. We determined the structure of MPV467 in complex with the hMPV F protein using cryo-electron microscopy to a resolution of 3.3 Å, which revealed a complex novel prefusion-specific epitope overlapping antigenic sites II and V on a single protomer. Overall, our data reveal insights into the immunodominant antigenic epitopes on the hMPV F protein, identify a mAb therapy for hMPV F disease prevention and treatment, and provide the discovery of a prefusion-specific epitope on the hMPV F protein.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Antígenos Virales , Metapneumovirus , Infecciones por Paramyxoviridae , Proteínas Virales de Fusión , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/química , Anticuerpos Antivirales/aislamiento & purificación , Anticuerpos Antivirales/uso terapéutico , Antígenos Virales/química , Antígenos Virales/inmunología , Microscopía por Crioelectrón , Epítopos/inmunología , Humanos , Metapneumovirus/inmunología , Ratones , Infecciones por Paramyxoviridae/prevención & control , Prevención Primaria , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/inmunología
5.
Sci Rep ; 11(1): 20769, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34675220

RESUMEN

Human metapneumovirus (HMPV) is a leading cause of acute lower respiratory tract illness in children and adults. Repeated infections are common and can be severe in young, elderly, and immunocompromised persons due to short-lived protective humoral immunity. In turn, few protective T cell epitopes have been identified in humans. Thus, we infected transgenic mice expressing the common human HLA MHC-I allele B*07:02 (HLA-B7) with HMPV and screened a robust library of overlapping and computationally predicted HLA-B7 binding peptides. Six HLA-B7-restricted CD8+ T cell epitopes were identified using ELISPOT screening in the F, M, and N proteins, with M195-203 (M195) eliciting the strongest responses. MHC-tetramer flow cytometric staining confirmed HLA-B7 epitope-specific CD8+ T cells migrated to lungs and spleen of HMPV-immune mice. Immunization with pooled HLA-B7-restricted peptides reduced viral titer and protected mice from virulent infection. Finally, we confirmed that CD8+ T cells from HLA-B7 positive humans also recognize the identified epitopes. These results enable identification of HMPV-specific CD8+ T cells in humans and help to inform future HMPV vaccine design.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Antígeno HLA-B7/inmunología , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Animales , Células Cultivadas , Epítopos de Linfocito T/uso terapéutico , Humanos , Interferón gamma/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Paramyxoviridae/prevención & control , Péptidos/inmunología , Péptidos/uso terapéutico , Vacunas Virales/inmunología , Vacunas Virales/uso terapéutico
6.
Vaccine ; 38(9): 2122-2127, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32007293

RESUMEN

Human metapneumovirus (hMPV) is an important respiratory pathogen especially in young children and elderly subjects. Our objective was to assess the immunogenicity and protection conferred by predominant pre- and post-fusion (F) hMPV-F constructs in Balb/C mice. Immunizations without adjuvant were not immunogenic whereas alum-adjuvanted hMPV-F proteins, regardless of their conformations, generated comparable neutralizing antibody titers with undetectable pulmonary viral titers following viral challenge. In conclusion, we found no apparent advantage for mixtures of predominant pre-fusion F proteins over post-fusion conformations for hMPV vaccination in opposite to recent data obtained with the human respiratory syncytial virus.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Metapneumovirus , Infecciones por Paramyxoviridae , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Metapneumovirus/inmunología , Ratones , Ratones Endogámicos BALB C , Infecciones por Paramyxoviridae/prevención & control , Vacunas de Subunidad/inmunología , Proteínas Virales de Fusión/administración & dosificación
7.
Sci Rep ; 8(1): 10425, 2018 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-29992955

RESUMEN

Paramyxoviridae, a large family of enveloped viruses harboring a nonsegmented negative-sense RNA genome, include important human pathogens as measles, mumps, respiratory syncytial virus (RSV), parainfluenza viruses, and henipaviruses, which cause some of the deadliest emerging zoonoses. There is no effective antiviral chemotherapy for most of these pathogens. Paramyxoviruses evolved a sophisticated membrane-fusion machine consisting of receptor-binding proteins and the fusion F-protein, critical for virus infectivity. Herein we identify the antiprotozoal/antimicrobial nitazoxanide as a potential anti-paramyxovirus drug targeting the F-protein. We show that nitazoxanide and its circulating-metabolite tizoxanide act at post-entry level by provoking Sendai virus and RSV F-protein aggregate formation, halting F-trafficking to the host plasma membrane. F-protein folding depends on ER-resident glycoprotein-specific thiol-oxidoreductase ERp57 for correct disulfide-bond architecture. We found that tizoxanide behaves as an ERp57 non-competitive inhibitor; the putative drug binding-site was located at the ERp57-b/b' non-catalytic domains interface. ERp57-silencing mimicked thiazolide-induced F-protein alterations, suggesting an important role of this foldase in thiazolides anti-paramyxovirus activity. Nitazoxanide is used in the clinic as a safe and effective antiprotozoal/antimicrobial drug; its antiviral activity was shown in patients infected with hepatitis-C virus, rotavirus and influenza viruses. Our results now suggest that nitazoxanide may be effective also against paramyxovirus infection.


Asunto(s)
Infecciones por Paramyxoviridae/tratamiento farmacológico , Paramyxoviridae/fisiología , Tiazoles/farmacología , Replicación Viral/efectos de los fármacos , Células A549 , Animales , Sitios de Unión , Humanos , Nitrocompuestos , Oxidorreductasas/metabolismo , Paramyxoviridae/efectos de los fármacos , Infecciones por Paramyxoviridae/prevención & control , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/química , Pliegue de Proteína/efectos de los fármacos , Transporte de Proteínas , Tiazoles/metabolismo , Proteínas Virales de Fusión/metabolismo
8.
Virology ; 509: 60-66, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28605636

RESUMEN

Human metapneumovirus (hMPV) infections pose a serious health risk to young children, particularly in cases of premature birth. No licensed vaccine exists and there is no standard treatment for hMPV infections apart from supportive hospital care. We describe the production of a Sendai virus (SeV) recombinant that carries a gene for a truncated hMPV fusion (F) protein (SeV-MPV-Ft). The vaccine induces binding and neutralizing antibody responses toward hMPV and protection against challenge with hMPV in a cotton rat system. Results encourage advanced development of SeV-MPV-Ft to prevent the morbidity and mortality caused by hMPV infections in young children.


Asunto(s)
Antígenos Virales/inmunología , Portadores de Fármacos , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/prevención & control , Virus Sendai/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Modelos Animales de Enfermedad , Metapneumovirus/genética , Infecciones por Paramyxoviridae/inmunología , Sigmodontinae , Resultado del Tratamiento , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas Virales de Fusión/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
9.
Sci Rep ; 7(1): 4025, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28642611

RESUMEN

Avian metapneumovirus (AMPV) infects the respiratory and reproductive tracts of domestic poultry, resulting in substantial economic losses for producers. Live attenuated vaccines appear to be the most effective in countries where the disease is prevalent. However, reversion to virulence has been demonstrated in several studies. Therefore, the development of a stable and safe next generation vaccine against the AMPV disease is needed. In the present study, we generated a recombinant Newcastle disease virus (NDV) vectoring the fusion (F) protein and glycoprotein (G) genes of AMPV subtype-C (AMPV-C) as a bivalent vaccine candidate using reverse genetics technology. The recombinant virus, rLS/AMPV-C F&G, was slightly attenuated in vivo, yet maintained similar characteristics in vitro when compared to the parental LaSota virus. Vaccination of turkeys with rLS/AMPV-C F&G induced both AMPV-C and NDV-specific antibody responses, and provided significant protection against pathogenic AMPV-C challenge and complete protection against velogenic NDV challenge. These results suggest that the rLS/AMPV-C F&G recombinant virus is a safe and effective bivalent vaccine candidate and that the expression of both F and G proteins of AMPV-C induces a protective response against the AMPV-C disease.


Asunto(s)
Vectores Genéticos/genética , Metapneumovirus/genética , Metapneumovirus/inmunología , Virus de la Enfermedad de Newcastle/genética , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/genética , Vacunas Virales/genética , Animales , Anticuerpos Antivirales/inmunología , Expresión Génica , Orden Génico , Vectores Genéticos/inmunología , Inmunidad Humoral , Inmunización , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/virología , Enfermedades de las Aves de Corral/prevención & control , Pavos , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Esparcimiento de Virus
10.
Vaccine ; 34(7): 950-6, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26772634

RESUMEN

Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) are major causes of illness among children, the elderly, and the immunocompromised. No vaccine has been licensed for protection against either of these viruses. We tested the ability of two Venezuelan equine encephalitis virus-based viral replicon particle (VEE-VRP) vaccines that express the hRSV or hMPV fusion (F) protein to confer protection against hRSV or hMPV in African green monkeys. Animals immunized with VEE-VRP vaccines developed RSV or MPV F-specific antibodies and serum neutralizing activity. Compared to control animals, immunized animals were better able to control viral load in the respiratory mucosa following challenge and had lower levels of viral genome in nasopharyngeal and bronchoalveolar lavage fluids. The high level of immunogenicity and protective efficacy induced by these vaccine candidates in nonhuman primates suggest that they hold promise for further development.


Asunto(s)
Infecciones por Paramyxoviridae/prevención & control , Replicón , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas Virales/inmunología , Alphavirus , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Líquido del Lavado Bronquioalveolar/virología , Chlorocebus aethiops , Virus de la Encefalitis Equina Venezolana , Inmunoglobulina G/sangre , Metapneumovirus , Pruebas de Neutralización , Nariz/virología , Virus Sincitial Respiratorio Humano , Proteínas Virales de Fusión/inmunología
11.
J Virol ; 89(17): 8713-26, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26063431

RESUMEN

UNLABELLED: Human metapneumovirus (HMPV) is a major cause of respiratory disease in infants, the elderly, and immunocompromised individuals worldwide. There is currently no licensed HMPV vaccine. Virus-like particles (VLPs) are an attractive vaccine candidate because they are noninfectious and elicit a neutralizing antibody response. However, studies show that serum neutralizing antibodies are insufficient for complete protection against reinfection and that adaptive T cell immunity is important for viral clearance. HMPV and other respiratory viruses induce lung CD8(+) T cell (TCD8) impairment, mediated by programmed death 1 (PD-1). In this study, we generated HMPV VLPs by expressing the fusion and matrix proteins in mammalian cells and tested whether VLP immunization induces functional HMPV-specific TCD8 responses in mice. C57BL/6 mice vaccinated twice with VLPs and subsequently challenged with HMPV were protected from lung viral replication for at least 20 weeks postimmunization. A single VLP dose elicited F- and M-specific lung TCD8s with higher function and lower expression of PD-1 and other inhibitory receptors than TCD8s from HMPV-infected mice. However, after HMPV challenge, lung TCD8s from VLP-vaccinated mice exhibited inhibitory receptor expression and functional impairment similar to those of mice experiencing secondary infection. HMPV challenge of VLP-immunized µMT mice also elicited a large percentage of impaired lung TCD8s, similar to mice experiencing secondary infection. Together, these results indicate that VLPs are a promising vaccine candidate but do not prevent lung TCD8 impairment upon HMPV challenge. IMPORTANCE: Human metapneumovirus (HMPV) is a leading cause of acute respiratory disease for which there is no licensed vaccine. Virus-like particles (VLPs) are an attractive vaccine candidate and induce antibodies, but T cell responses are less defined. Moreover, HMPV and other respiratory viruses induce lung CD8(+) T cell (TCD8) impairment mediated by programmed death 1 (PD-1). In this study, HMPV VLPs containing viral fusion and matrix proteins elicited epitope-specific TCD8s that were functional with low PD-1 expression. Two VLP doses conferred sterilizing immunity in C57BL/6 mice and facilitated HMPV clearance in antibody-deficient µMT mice without enhancing lung pathology. However, regardless of whether responding lung TCD8s had previously encountered HMPV antigens in the context of VLPs or virus, similar proportions were impaired and expressed comparable levels of PD-1 upon viral challenge. These results suggest that VLPs are a promising vaccine candidate but do not prevent lung TCD8 impairment upon HMPV challenge.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Pulmón/inmunología , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Células HEK293 , Humanos , Pulmón/citología , Depleción Linfocítica , Macaca mulatta , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/virología , Receptor de Muerte Celular Programada 1/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Infecciones del Sistema Respiratorio/virología , Vacunación , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Proteínas de la Matriz Viral/biosíntesis , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Replicación Viral/inmunología
12.
Intervirology ; 58(6): 403-12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27096202

RESUMEN

OBJECTIVES: No licensed vaccines or therapeutic agents for human metapneumovirus (hMPV) infection exist to date. We aimed to construct a multi-epitope peptide (MEP) of hMPV to show promising results for epitope-based vaccine development. METHODS: Six independent algorithms were screened to predict B-cell epitopes of hMPV, and three algorithms were used to predict cytotoxic T lymphocyte and T helper (Th) lymphocyte epitopes. Predicted epitopes were assembled in series with the spacers GPGPG and KK introduced, termed MEP. Recombinant mep genes were inserted into pET32a(+) plasmid and expressed in Escherichia coli strain BL21 (DE3). BALB/c mice were immunized with MEP with different adjuvants. Antibody titer, lymphocyte proliferation, cytotoxic T lymphocyte (CTL) activity and splenocyte cytokines were detected 2 weeks later after the last immunization. Microneutralization assay was used to detect neutralizing antibodies. RESULTS: Six B-cell epitopes, four CTL epitopes and two Th epitopes were screened to construct the mep gene. Expressed MEP induced >104 antibodies in BALB/c mice, and produced anti-MEP antibody reacting with hMPV strains specifically as detected in indirect fluorescent assay (the titer was 160). The lymphocyte proliferation index, CTL activity and splenocyte cytokines of the MEP immunization groups were higher than in the control group (p < 0.05). Both IgG1 and IgG2a antibodies could be detected in the different groups, and balanced Th1/Th2 cytokines were secreted by splenocytes in these groups. The mean neutralizing titers of the MEP+CpG ODN, MEP+Alum and MEP+Alum+ CpG ODN groups were 87 (95% CI 50-126), 93 (95% CI 67-121) and 96 (95% CI 69-147), respectively. CONCLUSION: MEP of hMPV elicited both strong humoral immunity and cell-mediated immunity in mice. The anti-MEP serum could neutralize hMPV infection in vitro. Joint use of CpG ODN and aluminum hydroxide adjuvants obtained the best immune effects. This study may contribute to hMPV epitope-based vaccine development.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Inmunidad Celular , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/prevención & control , Péptidos/inmunología , Adyuvantes Inmunológicos , Compuestos de Alumbre , Animales , Chlorocebus aethiops , Epítopos de Linfocito B/inmunología , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Humanos , Inmunización , Ratones , Ratones Endogámicos BALB C , Oligodesoxirribonucleótidos , Proteínas Recombinantes , Linfocitos T Citotóxicos , Células Vero
13.
J Infect Dis ; 211(2): 216-25, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24864121

RESUMEN

BACKGROUND: Human metapneumovirus (HMPV) is a leading cause of acute respiratory tract infection, with significant morbidity and mortality. No licensed vaccines or therapeutic agents exist. Monoclonal antibodies (mAbs) are effective at preventing other infectious diseases and could be used against HMPV in high-risk hosts. METHODS: In vitro assays were performed to assess the neutralizing activity and affinity kinetics of human mAb 54G10. A new mouse model was developed to assess prophylactic and therapeutic efficacy in vivo. The epitope of 54G10 was identified by generating mAb-resistant mutants (MARMs). RESULTS: At low concentrations, 54G10 neutralized all 4 subgroups of HMPV in vitro and had subnanomolar affinity for the fusion protein. DBA/2 mice were permissive for all 4 HMPV subgroups, and 54G10 was effective both prophylactically and therapeutically against HMPV in vivo. Sequencing of HMPV MARMs identified the 54G10 epitope, which was similar to an antigenic site on respiratory syncytial virus (RSV). 54G10 also exhibited in vitro neutralizing activity and in vivo protective and therapeutic efficacy against RSV. CONCLUSIONS: Human mAb 54G10 has broad neutralizing activity against HMPV and could have prophylactic and therapeutic utility clinically. The conserved epitope could represent a structural vaccine target for HMPV and RSV.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Metapneumovirus/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Animales , Anticuerpos Monoclonales Humanizados/aislamiento & purificación , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Antivirales/aislamiento & purificación , Quimioprevención/métodos , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoterapia/métodos , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Pruebas de Neutralización , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/terapia , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/terapia , Resultado del Tratamiento
14.
J Virol ; 88(21): 12348-63, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25122790

RESUMEN

UNLABELLED: Avian metapneumovirus (aMPV), also known as avian pneumovirus or turkey rhinotracheitis virus, is the causative agent of turkey rhinotracheitis and is associated with swollen head syndrome in chickens. Since its discovery in the 1970s, aMPV has been recognized as an economically important pathogen in the poultry industry worldwide. The conserved region VI (CR VI) of the large (L) polymerase proteins of paramyxoviruses catalyzes methyltransferase (MTase) activities that typically methylate viral mRNAs at guanine N-7 (G-N-7) and ribose 2'-O positions. In this study, we generated a panel of recombinant aMPV (raMPV) Colorado strains carrying mutations in the S-adenosyl methionine (SAM) binding site in the CR VI of L protein. These recombinant viruses were specifically defective in ribose 2'-O, but not G-N-7 methylation and were genetically stable and highly attenuated in cell culture and viral replication in the upper and lower respiratory tracts of specific-pathogen-free (SPF) young turkeys. Importantly, turkeys vaccinated with these MTase-defective raMPVs triggered a high level of neutralizing antibody and were completely protected from challenge with homologous aMPV Colorado strain and heterologous aMPV Minnesota strain. Collectively, our results indicate (i) that aMPV lacking 2'-O methylation is highly attenuated in vitro and in vivo and (ii) that inhibition of mRNA cap MTase can serve as a novel target to rationally design live attenuated vaccines for aMPV and perhaps other paramyxoviruses. IMPORTANCE: Paramyxoviruses include many economically and agriculturally important viruses such as avian metapneumovirus (aMPV), and Newcastle disease virus (NDV), human pathogens such as human respiratory syncytial virus, human metapneumovirus, human parainfluenza virus type 3, and measles virus, and highly lethal emerging pathogens such as Nipah virus and Hendra virus. For many of them, there is no effective vaccine or antiviral drug. These viruses share common strategies for viral gene expression and replication. During transcription, paramyxoviruses produce capped, methylated, and polyadenylated mRNAs. Using aMPV as a model, we found that viral ribose 2'-O methyltransferase (MTase) is a novel approach to rationally attenuate the virus for vaccine purpose. Recombinant aMPV (raMPV) lacking 2'-O MTase were not only highly attenuated in turkeys but also provided complete protection against the challenge of homologous and heterologous aMPV strains. This novel approach can be applicable to other animal and human paramyxoviruses for rationally designing live attenuated vaccines.


Asunto(s)
Protección Cruzada , Metapneumovirus/enzimología , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/veterinaria , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Pulmón/virología , Metapneumovirus/genética , Metiltransferasas/deficiencia , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/virología , Tráquea/virología , Pavos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Carga Viral , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
15.
J Virol ; 88(19): 11411-29, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25056882

RESUMEN

UNLABELLED: The paramyxoviruses human respiratory syncytial virus (hRSV), human metapneumovirus (hMPV), and human parainfluenza virus type 3 (hPIV3) are responsible for the majority of pediatric respiratory diseases and inflict significant economic loss, health care costs, and emotional burdens. Despite major efforts, there are no vaccines available for these viruses. The conserved region VI (CR VI) of the large (L) polymerase proteins of paramyxoviruses catalyzes methyltransferase (MTase) activities that typically methylate viral mRNAs at positions guanine N-7 (G-N-7) and ribose 2'-O. In this study, we generated a panel of recombinant hMPVs carrying mutations in the S-adenosylmethionine (SAM) binding site in CR VI of L protein. These recombinant viruses were specifically defective in ribose 2'-O methylation but not G-N-7 methylation and were genetically stable and highly attenuated in cell culture and viral replication in the upper and lower respiratory tracts of cotton rats. Importantly, vaccination of cotton rats with these recombinant hMPVs (rhMPVs) with defective MTases triggered a high level of neutralizing antibody, and the rats were completely protected from challenge with wild-type rhMPV. Collectively, our results indicate that (i) amino acid residues in the SAM binding site in the hMPV L protein are essential for 2'-O methylation and (ii) inhibition of mRNA cap MTase can serve as a novel target to rationally design live attenuated vaccines for hMPV and perhaps other paramyxoviruses, such as hRSV and hPIV3. IMPORTANCE: Human paramyxoviruses, including hRSV, hMPV, and hPIV3, cause the majority of acute upper and lower respiratory tract infections in humans, particularly in infants, children, the elderly, and immunocompromised individuals. Currently, there is no licensed vaccine available. A formalin-inactivated vaccine is not suitable for these viruses because it causes enhanced lung damage upon reinfection with the same virus. A live attenuated vaccine is the most promising vaccine strategy for human paramyxoviruses. However, it remains a challenge to identify an attenuated virus strain that has an optimal balance between attenuation and immunogenicity. Using reverse genetics, we generated a panel of recombinant hMPVs that were specifically defective in ribose 2'-O methyltransferase (MTase) but not G-N-7 MTase. These MTase-defective hMPVs were genetically stable and sufficiently attenuated but retained high immunogenicity. This work highlights a critical role of 2'-O MTase in paramyxovirus replication and pathogenesis and a new avenue for the development of safe and efficacious live attenuated vaccines for hMPV and other human paramyxoviruses.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Metapneumovirus/inmunología , Metiltransferasas/inmunología , Infecciones por Paramyxoviridae/prevención & control , Proteínas Virales/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/biosíntesis , Sitios de Unión , Femenino , Expresión Génica , Humanos , Inmunidad Activa , Metapneumovirus/enzimología , Metapneumovirus/genética , Metiltransferasas/química , Metiltransferasas/genética , Infecciones por Paramyxoviridae/inmunología , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Sigmodontinae , Vacunación , Vacunas Atenuadas , Proteínas Virales/química , Proteínas Virales/genética , Vacunas Virales/administración & dosificación
16.
Nature ; 501(7467): 439-43, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-23955151

RESUMEN

Broadly neutralizing antibodies reactive against most and even all variants of the same viral species have been described for influenza and HIV-1 (ref. 1). However, whether a neutralizing antibody could have the breadth of range to target different viral species was unknown. Human respiratory syncytial virus (HRSV) and human metapneumovirus (HMPV) are common pathogens that cause severe disease in premature newborns, hospitalized children and immune-compromised patients, and play a role in asthma exacerbations. Although antisera generated against either HRSV or HMPV are not cross-neutralizing, we speculated that, because of the repeated exposure to these viruses, cross-neutralizing antibodies may be selected in some individuals. Here we describe a human monoclonal antibody (MPE8) that potently cross-neutralizes HRSV and HMPV as well as two animal paramyxoviruses: bovine RSV (BRSV) and pneumonia virus of mice (PVM). In its germline configuration, MPE8 is HRSV-specific and its breadth is achieved by somatic mutations in the light chain variable region. MPE8 did not result in the selection of viral escape mutants that evaded antibody targeting and showed potent prophylactic efficacy in animal models of HRSV and HMPV infection, as well as prophylactic and therapeutic efficacy in the more relevant model of lethal PVM infection. The core epitope of MPE8 was mapped on two highly conserved anti-parallel ß-strands on the pre-fusion viral F protein, which are rearranged in the post-fusion F protein conformation. Twenty-six out of the thirty HRSV-specific neutralizing antibodies isolated were also found to be specific for the pre-fusion F protein. Taken together, these results indicate that MPE8 might be used for the prophylaxis and therapy of severe HRSV and HMPV infections and identify the pre-fusion F protein as a candidate HRSV vaccine.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Reacciones Cruzadas/inmunología , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/virología , Paramyxoviridae/clasificación , Paramyxoviridae/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/uso terapéutico , Especificidad de Anticuerpos/inmunología , Bovinos , Epítopos/inmunología , Humanos , Cadenas Ligeras de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/inmunología , Región Variable de Inmunoglobulina/química , Región Variable de Inmunoglobulina/inmunología , Metapneumovirus/inmunología , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Virus de la Neumonía Murina/inmunología , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/terapia , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/prevención & control , Infecciones por Pneumovirus/virología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/terapia , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Bovino/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/inmunología , Vacunas Virales/química , Vacunas Virales/inmunología
17.
East Mediterr Health J ; 19 Suppl 1: S39-47, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23888794

RESUMEN

Viruses account for the majority of the acute respiratory tract infections (ARIs) globally with a mortality exceeding 4 million deaths per year. The most commonly encountered viruses, in order of frequency, include influenza, respiratory syncytial virus, parainfluenza and adenovirus. Current evidence suggests that the major mode of transmission of ARls is through large droplets, but transmission through contact (including hand contamination with subsequent self-inoculation) and infectious respiratory aerosols of various sizes and at short range (coined as "opportunistic" airborne transmission) may also occur for some pathogens. Opportunistic airborne transmission may occur when conducting highrisk aerosol generating procedures and airborne precautions will be required in this setting. General infection control measures effective for all respiratory viral infections are reviewed and followed by discussion on some of the common viruses, including severe acute respiratory syndrome (SARS) coronavirus and the recently discovered novel coronavirus.


Asunto(s)
Infección Hospitalaria/prevención & control , Atención a la Salud/métodos , Control de Infecciones/métodos , Infecciones del Sistema Respiratorio/prevención & control , Enfermedad Aguda , Infecciones por Adenovirus Humanos/prevención & control , Infecciones por Adenovirus Humanos/transmisión , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/transmisión , Humanos , Gripe Humana/prevención & control , Gripe Humana/transmisión , Internacionalidad , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/transmisión , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/transmisión , Infecciones del Sistema Respiratorio/transmisión , Síndrome Respiratorio Agudo Grave/prevención & control , Síndrome Respiratorio Agudo Grave/transmisión
18.
Vaccine ; 31(22): 2565-71, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23588091

RESUMEN

The study investigates the ability of subtype A Avian metapneumovirus (AMPV) to accept foreign genes and be used as a vector for delivery of Infectious bronchitis virus (IBV) QX genes to chickens. Initially the GFP gene was added to AMPV at all gene junctions in conjunction with the development of cassetted full length DNA AMPV copies. After recombinant virus had been recovered by reverse genetics, GFP positions supporting gene expression while maintaining virus viability in vitro, were determined. Subsequently, either S1 or nucleocapsid (N) genes of IBV were positioned between AMPV M and F genes, while later a bivalent recombinant was prepared by inserting S1 and N at AMPV MF and GL junctions respectively. Immunofluorescent antibody staining showed that all recombinants expressed the inserted IBV genes in vitro and furthermore, all recombinant viruses were found to be highly stable during serial passage. Eyedrop inoculation of chickens with some AMPV-IBV recombinants at one-day-old induced protection against virulent IBV QX challenge 3 weeks later, as assessed by greater motility of tracheal cilia from chickens receiving the recombinants. Nonetheless evidence of AMPV/IBV seroconversion, or major recombinant tracheal replication, were largely absent.


Asunto(s)
Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/inmunología , Metapneumovirus/genética , Metapneumovirus/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología , Animales , Pollos , Chlorocebus aethiops , Proteínas de la Nucleocápside de Coronavirus , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Mutagénesis Insercional , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteínas de la Nucleocápside/biosíntesis , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/inmunología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/veterinaria , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunación/veterinaria , Células Vero
19.
Rev. chil. infectol ; 29(supl.1): 33-36, set. 2012. tab
Artículo en Español | LILACS | ID: lil-656324

RESUMEN

Respiratory viruses have been identified as a cause of morbidity and mortality in patients undergoing SOT and HSCT, specially in children. The most frequent are respiratory syncytial virus (RSV), influenza (FLU), parainfluenza (PI) and adenovirus (ADV). These infections are associated with progression to severe lower respiratory tract infections in up to 60% of the cases. It is advised to apply universal protection recommendations for respiratory viruses (A2) and some specific measures for FLU and AD. FLU: Annual anti-influenza vaccination (from 4-6 months post-transplantation in SOT, 6 months in HSCT (A2)); post- exposure prophylaxis in FLU (oseltamivir for 10 days (B2)). In lung transplantion, the prophylaxis should last as long as the risk period (B2). ADV: There is no vaccine nor valid chemoprophylaxis strategy to prevent ADV disease. In some specific HSCT recipients, weekly PCR monitoring is recommended until day+100 (A3).


Los virus respiratorios se han identificado como causa de morbi-mortalidad en pacientes sometidos a TOS y TPH, particularmente en pediatría. Los más frecuentes son virus respiratorio sincicial (VRS), influenza (FLU), parainfluenza (PI) y adenovirus (ADV). La fuente de contagio está en la comunidad y en el hospital afectando al paciente en cualquier período post-trasplante. Se describe progresión a infecciones graves del tracto respiratorio bajo hasta en 60 % de los casos. Se recomienda aplicar medidas de aislamiento de precaución universal para todos los virus respiratorios (A2) y se describen algunas medidas específicas para FLU y AlDV. Vacunación anti-influenza anual con vacuna inactivada (en TOS a partir de 4-6 meses post-trasplante (A2), en TPH a partir de 6 meses (A2)); profilaxis post exposición a virus FLU (oseltamivir durante 10 días (B2)). En trasplante de pulmón, la duración de la profilaxis se extenderá mientras dure el período de riesgo (B2). Con respecto a ADV, no se dispone de una vacuna adecuada y no existe a la fecha una estrategia validada de quimioprofilaxis para prevenir enfermedad por ADV; en casos específicos de TPH pediátrico, se recomienda vigilancia semanal con RPC en sangre periférica hasta el día +100 post-TPH (A3).


Asunto(s)
Adulto , Niño , Humanos , Antivirales , Gripe Humana/prevención & control , Trasplante de Órganos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Trasplante de Células Madre , Infecciones por Adenoviridae/prevención & control , Antivirales/administración & dosificación , Esquema de Medicación , Incidencia , Gripe Humana/epidemiología , Gripe Humana/virología , Oseltamivir/administración & dosificación , Oseltamivir/uso terapéutico , Guías de Práctica Clínica como Asunto , Infecciones por Paramyxoviridae/prevención & control , Complicaciones Posoperatorias/prevención & control , Factores de Riesgo , Infecciones por Virus Sincitial Respiratorio/epidemiología
20.
Infect Control Hosp Epidemiol ; 33(6): 545-50, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22561708

RESUMEN

BACKGROUND: Viral respiratory pathogens cause outbreaks in pediatric long-term care facilities (LTCFs), but few studies have used viral diagnostic testing to identify the causative pathogens. We describe the use of such testing during a prolonged period of respiratory illness and elucidate the epidemiology of human metapneumovirus (hMPV) at our LTCF. DESIGN: Retrospective study of influenza-like illness (ILI). SETTING: A 136-bed pediatric LTCF from January 1 through April 30, 2010. METHODS: The ILI case definition included fever, cough, change in oropharyngeal secretions, increase in oxygen requirement, and/or wheezing. RESULTS: During the study period, 69 episodes of ILI occurred in 61 (41%) of 150 residents. A viral pathogen was detected in 27 (39%) of the episodes, including respiratory syncytial virus (RSV) (n = 3), influenza A virus (not typed; n = 2), parainfluenza virus (n =2), adenovirus (n = 1), and hMPV (n = 19). Twenty-seven of the residents with ILI (44%) required transfer to acute care hospitals (mean length of hospitalization, 12 days; range, 3-47 days). Residents with tracheostomies were more likely to have ILI (adjusted odds ratio [OR], 3.99 [95% confidence interval {CI}, 1.87-8.53]; P = .0004). The mortality rate for residents with ILI was 1.6%. Residents with hMPV were younger (P = .03), more likely to be transferred to an acute care facility (OR, 3.73 [95% CI, 1.17-11.95]; P = .02), and less likely to have a tracheostomy (adjusted OR, 0.19 [95% CI, 0.047-0.757]; P = .02 ). DISCUSSION: Diverse pathogens, most notably hMPV, caused ILI in our pediatric LTCF during a prolonged period of time. Viral testing was helpful in characterizing the epidemiology of ILI in this population.


Asunto(s)
Infección Hospitalaria/epidemiología , Brotes de Enfermedades , Metapneumovirus/aislamiento & purificación , Infecciones por Paramyxoviridae/epidemiología , Instituciones Residenciales , Infecciones por Adenoviridae/diagnóstico , Infecciones por Adenoviridae/epidemiología , Infecciones por Adenoviridae/prevención & control , Adolescente , Niño , Preescolar , Infección Hospitalaria/diagnóstico , Infección Hospitalaria/prevención & control , Brotes de Enfermedades/prevención & control , Brotes de Enfermedades/estadística & datos numéricos , Femenino , Humanos , Lactante , Control de Infecciones , Virus de la Influenza A/aislamiento & purificación , Gripe Humana/diagnóstico , Gripe Humana/epidemiología , Gripe Humana/prevención & control , Masculino , Ciudad de Nueva York , Infecciones por Paramyxoviridae/diagnóstico , Infecciones por Paramyxoviridae/prevención & control , Transferencia de Pacientes/estadística & datos numéricos , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Estudios Retrospectivos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA