Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Microbiol Spectr ; 12(4): e0365523, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38415660

RESUMEN

Although the global crisis caused by the coronavirus disease 2019 (COVID-19) pandemic is over, the global epidemic of the disease continues. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the cause of COVID-19, initiates infection via the binding of the receptor-binding domain (RBD) of its spike protein to the human angiotensin-converting enzyme II (ACE2) receptor, and this interaction has been the primary target for the development of COVID-19 therapeutics. Here, we identified neutralizing antibodies against SARS-CoV-2 by screening mouse monoclonal antibodies and characterized an antibody, CSW1-1805, that targets a narrow region at the RBD ridge of the spike protein. CSW1-1805 neutralized several variants in vitro and completely protected mice from SARS-CoV-2 infection. Cryo-EM and biochemical analyses revealed that this antibody recognizes the loop region adjacent to the ACE2-binding interface with the RBD in both a receptor-inaccessible "down" state and a receptor-accessible "up" state and could stabilize the RBD conformation in the up-state. CSW1-1805 also showed different binding orientations and complementarity determining region properties compared to other RBD ridge-targeting antibodies with similar binding epitopes. It is important to continuously characterize neutralizing antibodies to address new variants that continue to emerge. Our characterization of this antibody that recognizes the RBD ridge of the spike protein will aid in the development of future neutralizing antibodies.IMPORTANCESARS-CoV-2 cell entry is initiated by the interaction of the viral spike protein with the host cell receptor. Therefore, mechanistic findings regarding receptor recognition by the spike protein help uncover the molecular mechanism of SARS-CoV-2 infection and guide neutralizing antibody development. Here, we characterized a SARS-CoV-2 neutralizing antibody that recognizes an epitope, a loop region adjacent to the receptor-binding interface, that may be involved in the conformational transition of the receptor-binding domain (RBD) of the spike protein from a receptor-inaccessible "down" state into a receptor-accessible "up" state, and also stabilizes the RBD in the up-state. Our mechanistic findings provide new insights into SARS-CoV-2 receptor recognition and guidance for neutralizing antibody development.


Asunto(s)
Anticuerpos Neutralizantes , COVID-19 , Humanos , Animales , Ratones , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2 , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Antivirales , Epítopos
2.
Vaccines (Basel) ; 11(8)2023 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-37631959

RESUMEN

The numerous influenza infections that occur every year present a major public health problem. Influenza vaccines are important for the prevention of the disease; however, their effectiveness against infection can be suboptimal. Particularly in the elderly, immune induction can be insufficient, and the vaccine efficacy against infection is usually lower than that in young adults. Vaccine efficacy can be improved by the addition of adjuvants, and an influenza vaccine with an oil-in-water adjuvant MF59, FLUAD, has been recently licensed in the United States and other countries for persons aged 65 years and older. Although the adverse effects of adjuvanted vaccines have been a concern, many adverse effects of currently approved adjuvanted influenza vaccines are mild and acceptable, given the overriding benefits of the vaccine. Since sufficient immunity can be induced with a small amount of vaccine antigen in the presence of an adjuvant, adjuvanted vaccines promote dose sparing and the prompt preparation of vaccines for pandemic influenza. Adjuvants not only enhance the immune response to antigens but can also be effective against antigenically different viruses. In this narrative review, we provide an overview of influenza vaccines, both past and present, before presenting a discussion of adjuvanted influenza vaccines and their future.

3.
iScience ; 26(3): 106210, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36811085

RESUMEN

Coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), encode a proofreading exonuclease, nonstructural protein 14 (nsp14), that helps ensure replication competence at a low evolutionary rate compared with other RNA viruses. In the current pandemic, SARS-CoV-2 has accumulated diverse genomic mutations including in nsp14. Here, to clarify whether amino acid substitutions in nsp14 affect the genomic diversity and evolution of SARS-CoV-2, we searched for amino acid substitutions in nature that may interfere with nsp14 function. We found that viruses carrying a proline-to-leucine change at position 203 (P203L) have a high evolutionary rate and that a recombinant SARS-CoV-2 virus with the P203L mutation acquired more diverse genomic mutations than wild-type virus during its replication in hamsters. Our findings suggest that substitutions, such as P203L, in nsp14 may accelerate the genomic diversity of SARS-CoV-2, contributing to virus evolution during the pandemic.

4.
Jpn J Infect Dis ; 76(2): 162-166, 2023 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-36575023

RESUMEN

Biosafety level 4 (BSL-4) laboratories are necessary to study microorganisms that are highly pathogenic to humans and have no prevention or therapeutic measures. Currently, most BSL-4 facilities have suit-type laboratories to conduct experiments on highly pathogenic microorganisms. In 2021, the first Japanese suit-type BSL-4 laboratory was constructed at Nagasaki University. Positive pressure protection suit (PPPS) is a primary barrier that protects and isolates laboratory workers from pathogens and the laboratory environment. Here, we developed a novel PPPS originally designed to be used in the Nagasaki BSL-4 laboratory. We modified several parts of a domestic chemical protective suit, including its front face shield, cuff, and air supply hose, for safe handling of microbiological agents. The improved suit, PS-790BSL4-AL, showed resistance to several chemicals, including quaternary ammonium disinfectant, and did not show any permeation against blood and phages. To validate the suit's integrity, we also established an airtight test that eliminated individual differences for quantitative testing. In conclusion, our developed suit performs sufficiently as a primary barrier and allows for the safe handling of pathogens in our new BSL-4 laboratory.


Asunto(s)
Contención de Riesgos Biológicos , Laboratorios , Humanos , Japón
5.
Vaccine ; 40(30): 4026-4037, 2022 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-35641357

RESUMEN

The All-Japan Influenza Vaccine Study Group has been developing a more effective vaccine than the current split vaccines for seasonal influenza virus infection. In the present study, the efficacy of formalin- and/or ß-propiolactone-inactivated whole virus particle vaccines for seasonal influenza was compared to that of the current ether-treated split vaccines in a nonhuman primate model. The monovalent whole virus particle vaccines or split vaccines of influenza A virus (H1N1) and influenza B virus (Victoria lineage) were injected subcutaneously into naïve cynomolgus macaques twice. The whole virus particle vaccines induced higher titers of neutralizing antibodies against H1N1 influenza A virus and influenza B virus in the plasma of macaques than did the split vaccines. At challenge with H1N1 influenza A virus or influenza B virus, the virus titers in nasal swabs and the increases in body temperatures were lower in the macaques immunized with the whole virus particle vaccine than in those immunized with the split vaccine. Repertoire analyses of immunoglobulin heavy chain genes demonstrated that the number of B-lymphocyte subclones was increased in macaques after the 1st vaccination with the whole virus particle vaccine, but not with the split vaccine, indicating that the whole virus particle vaccine induced the activation of vaccine antigen-specific B-lymphocytes more vigorously than did the split vaccine at priming. Thus, the present findings suggest that the superior antibody induction ability of the whole virus particle vaccine as compared to the split vaccine is attributable to its stimulatory properties on the subclonal differentiation of antigen-specific B-lymphocytes.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Linfocitos B , Genes de Inmunoglobulinas , Humanos , Gripe Humana/prevención & control , Macaca fascicularis , Vacunación , Vacunas de Productos Inactivados , Virión
6.
Nat Aging ; 2(2): 115-124, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-37117754

RESUMEN

Reports of post-acute COVID-19 syndrome, in which the inflammatory response persists even after SARS-CoV-2 has disappeared, are increasing1, but the underlying mechanisms of post-acute COVID-19 syndrome remain unknown. Here, we show that SARS-CoV-2-infected cells trigger senescence-like cell-cycle arrest2,3 in neighboring uninfected cells in a paracrine manner via virus-induced cytokine production. In cultured human cells or bronchial organoids, these SASR-CoV-2 infection-induced senescent cells express high levels of a series of inflammatory factors known as senescence-associated secretory phenotypes (SASPs)4 in a sustained manner, even after SARS-CoV-2 is no longer detectable. We also show that the expression of the senescence marker CDKN2A (refs. 5,6) and various SASP factor4 genes is increased in the pulmonary cells of patients with severe post-acute COVID-19 syndrome. Furthermore, we find that mice exposed to a mouse-adapted strain of SARS-CoV-2 exhibit prolonged signs of cellular senescence and SASP in the lung at 14 days after infection when the virus was undetectable, which could be substantially reduced by the administration of senolytic drugs7. The sustained infection-induced paracrine senescence described here may be involved in the long-term inflammation caused by SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Humanos , Ratones , Animales , SARS-CoV-2 , Senescencia Celular/genética , Pulmón , Inflamación
7.
Uirusu ; 72(2): 131-138, 2022.
Artículo en Japonés | MEDLINE | ID: mdl-38220157

RESUMEN

Biosafety Level 4 (BSL-4) laboratories are required for research on microorganisms that are highly pathogenic to humans and for which there are no prevention or treatment methods. Currently, the majority of BSL-4 laboratories in more than 60 around the world are suit-type laboratories using positive pressure protective suits. In 2021, the first suit-type BSL-4 laboratory in Japan was constructed at Nagasaki University. Positive pressure protective suits are important as primary barriers to protect workers from pathogens, but the selection process has been largely unexplored. Here, I describe the selection process for the positive pressure protective suits to be used at the BSL-4 laboratory of Nagasaki University, and introduce a novel positive pressure protective suit (PS-790BSL4-AL), which was originally designed and produced in Japan.

8.
Nat Commun ; 12(1): 2654, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976181

RESUMEN

Most anti-influenza drugs currently used, such as oseltamivir and zanamivir, inhibit the enzymatic activity of neuraminidase. However, neuraminidase inhibitor-resistant viruses have already been identified from various influenza virus isolates. Here, we report the development of a class of macrocyclic peptides that bind the influenza viral envelope protein hemagglutinin, named iHA. Of 28 iHAs examined, iHA-24 and iHA-100 have inhibitory effects on the in vitro replication of a wide range of Group 1 influenza viruses. In particular, iHA-100 bifunctionally inhibits hemagglutinin-mediated adsorption and membrane fusion through binding to the stalk domain of hemagglutinin. Moreover, iHA-100 shows powerful efficacy in inhibiting the growth of highly pathogenic influenza viruses and preventing severe pneumonia at later stages of infection in mouse and non-human primate cynomolgus macaque models. This study shows the potential for developing cyclic peptides that can be produced more efficiently than antibodies and have multiple functions as next-generation, mid-sized biomolecules.


Asunto(s)
Antivirales/farmacología , Modelos Animales de Enfermedad , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Péptidos/farmacología , Neumonía/prevención & control , Animales , Antivirales/química , Perros , Femenino , Células HEK293 , Humanos , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Macaca fascicularis , Células de Riñón Canino Madin Darby , Ratones Endogámicos BALB C , Estructura Molecular , Péptidos/química , Replicación Viral/efectos de los fármacos
9.
Antiviral Res ; 178: 104790, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32272175

RESUMEN

Human cases of H7N9 influenza A virus infection have been increasing since 2013. The first choice of treatment for influenza is neuraminidase (NA) inhibitors (NAIs), but there is a concern that NAI-resistant viruses are selected in the presence of NAIs. In our previous study, an H7N9 virus carrying AA substitution of threonine (T) for isoleucine (I) at residue 222 in NA (NA222T, N2 numbering) and an H7N9 virus carrying AA substitution of lysine (K) for arginine (R) at residue 292 in NA (NA292K, N2 numbering) were found in different macaques that had been infected with A/Anhui/1/2013 (H7N9) and treated with NAIs. In the present study, the variant with NA292K showed not only resistance to NAIs but also lower replication activity in MDCK cells than did the virus with wild-type NA, whereas the variant with NA222T, which was less resistant to NAIs, showed replication activity similar to that of the wild-type virus. Next, we examined the pathogenicity of these H7N9 NAI-resistant viruses in macaques. The variants caused clinical signs similar to those caused by the wild-type virus with similar replication potency. However, the virus with NA292K was replaced within 7 days by that with NA292R (same as the wild-type) in nasal samples from macaques infected with the virus with NA292K, i.e. the so-called revertant (wild-type virus) became dominant in the population in the absence of an NAI. These results suggest that the clinical signs observed in macaques infected with the NA292K virus are caused by the NA292K virus and the NA292R virus and that the virus with NA292K may not replicate continuously in the upper respiratory tract of patients without treatment as effectively as the wild-type virus.


Asunto(s)
Antivirales/farmacología , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Subtipo H7N9 del Virus de la Influenza A/genética , Neuraminidasa/antagonistas & inhibidores , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/virología , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Sustitución de Aminoácidos , Animales , Farmacorresistencia Viral , Inhibidores Enzimáticos/farmacología , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Subtipo H7N9 del Virus de la Influenza A/fisiología , Macaca fascicularis , Mutación , Neuraminidasa/química , Nariz/virología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/patología , Neumonía Viral/virología , Sistema Respiratorio/virología , Selección Genética , Proteínas Virales/química , Replicación Viral
10.
Antiviral Res ; 171: 104591, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31421167

RESUMEN

Clarithromycin (CAM), a 14-membered ring macrolide, has anti-inflammatory and immunomodulatory actions and antiviral effects in seasonal influenza virus infection. We examined the prophylactic and therapeutic efficacy of CAM against H5N1 highly pathogenic and H7N9 low pathogenic avian influenza virus infections in cynomolgus monkeys. CAM suppressed H5N1 virus-induced severe signs of disease in the treated monkeys and inhibited virus propagation in tracheal samples and the production of inflammatory cytokines in the lungs of monkeys infected with H5N1 and H7N9 viruses. The prophylactic administration of CAM showed more suppressive effects on clinical signs of disease and viral titers than did therapeutic administration. Thus, since administration of CAM alone showed a tendency to ameliorate clinical sings and to reduce levels of inflammatory cytokines, the macrolides are expected to have effects in combination with the other antiviral drugs on the prophylactic and treatment of patients with severe avian influenza virus infection, which should be further investigated.


Asunto(s)
Antivirales/farmacología , Claritromicina/farmacología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Infecciones por Orthomyxoviridae/virología , Animales , Citocinas/metabolismo , Pulmón/patología , Pulmón/virología , Macaca fascicularis , Infecciones por Orthomyxoviridae/diagnóstico , Carga Viral
11.
Virology ; 510: 252-261, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28756116

RESUMEN

Vaccination-primed immunity in poultry has been suggested for selection of antigenically drifted highly pathogenic avian influenza viruses (HPAIVs). In this study, we performed two consecutive passage studies of an H5N1 HPAIV in vaccinated chickens, namely, study-I and study-II, to select antigenic variants under immune pressure from the vaccination. In study-I, nine consecutive passages of a wild-type H5N1 HPAIV were carried out in chickens vaccinated with the homologous challenge strain. Antigenically drifted variants with mutations at position 179 in the hemagglutinin (HA) were selected after three passages. Similarly, in study-II, a vaccination-mediated antigenic variant isolated in study-I was used as the vaccine and challenge strain to confirm further antigenic drift after updating the vaccine; after the third passage, additional antigenic variants with a mutation at position 256 in the HA were selected. Thus, our study demonstrated the contribution of vaccination in the selection of antigenic variants of H5 HPAIVs in chickens.


Asunto(s)
Variación Antigénica , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Aviar/virología , Animales , Pollos , Flujo Genético , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Mutación , Selección Genética , Pase Seriado
12.
Vaccine ; 35(7): 1008-1017, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28109707

RESUMEN

The efficacy and detrimental effect of mucosal vaccination with an inactivated influenza vaccine were examined in a macaque model by intranasal administration with small amounts of inactivated whole virus particles and challenge by a human-derived H5N1 highly pathogenic avian influenza virus infection. Repeated nasal inoculation with the whole particle vaccine of an inactivated virus, A/duck/Hokkaido/Vac-3/2007 (H5N1) (Vac-3), induced antigen-specific IgA and IgG antibody production in nasal swabs and plasma. Vac-3-specific IgE production was also found in the nasal swabs. Nasal vaccination with Vac-3 induced broader cross-clade neutralization activity than did subcutaneous vaccination. After challenge infection, repeated nasal vaccination almost completely prevented the propagation of virus in the upper and lower airways and protected cynomolgus macaques from viral pneumonia by induction of IgA-producing B cells in the lungs. On the other hand, eosinophil clusters were observed in the lungs of vaccinated macaques. Although Vac-3-specific IgE antibody and IL-13 levels were decreased after infection compared to those before infection and no anaphylaxis in vaccinated macaques was detected after challenge infection, our results suggest that we have to pay attention to potential allergic responses at repeated nasal vaccination, especially in people who have an airway allergy.


Asunto(s)
Administración Intranasal/efectos adversos , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Hipersensibilidad a las Drogas/etiología , Vacunas contra la Influenza/administración & dosificación , Infecciones por Orthomyxoviridae/prevención & control , Vacunación , Animales , Hipersensibilidad a las Drogas/inmunología , Hipersensibilidad a las Drogas/patología , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Femenino , Inmunoglobulina A/biosíntesis , Inmunoglobulina E/biosíntesis , Inmunoglobulina G/biosíntesis , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/inmunología , Infiltración Neutrófila/efectos de los fármacos , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Vacunas de Productos Inactivados , Virión/química , Virión/inmunología
13.
Sci Rep ; 6: 37915, 2016 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-27892498

RESUMEN

H5N1 highly pathogenic avian influenza (H5N1 HPAI) virus causes elevated mortality compared with seasonal influenza viruses like H1N1 pandemic influenza (H1N1 pdm) virus. We identified a mechanism associated with the severe symptoms seen with H5N1 HPAI virus infection. H5N1 HPAI virus infection induced a decrease of dendritic cell number in the splenic extrafollicular T-cell zone and impaired formation of the outer layers of B-cell follicles, resulting in insufficient levels of antibody production after infection. However, in animals vaccinated with a live recombinant vaccinia virus expressing the H5 hemagglutinin, infection with H5N1 HPAI virus induced parafollicular dendritic cell accumulation and efficient antibody production. These results indicate that a recombinant vaccinia encoding H5 hemagglutinin gene does not impair dendritic cell recruitment and can be a useful vaccine candidate.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/inmunología , Virus Vaccinia/genética , Animales , Linfocitos B/patología , Linfocitos B/virología , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/patología , Células Dendríticas/virología , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Humoral , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/genética , Pulmón/patología , Pulmón/virología , Macaca fascicularis , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Neumonía Viral/patología , Neumonía Viral/virología , Bazo/inmunología , Bazo/patología , Bazo/virología , Virus Vaccinia/inmunología
14.
Virology ; 493: 31-8, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26994587

RESUMEN

The outbreak of H7N9 low pathogenic avian influenza viruses in China has attracted attention to H7 influenza virus infection in humans. Since we have shown that the pathogenicity of H1N1 and H5N1 influenza viruses in macaques was almost the same as that in humans, we compared the pathogenicities of H7 avian influenza viruses in cynomolgus macaques via intranasal and conjunctival inoculation, which mimics natural infection in humans. H7N9 virus, as well as H7N7 highly pathogenic avian influenza virus, showed more efficient replication and higher pathogenicity in macaques than did H7N1 and H7N3 highly pathogenic avian influenza viruses. These results are different from pathogenicity in chickens as reported previously. Therefore, our results obtained in macaques help to estimate the pathogenicity of H7 avian influenza viruses in humans.


Asunto(s)
Subtipo H7N1 del Virus de la Influenza A/patogenicidad , Subtipo H7N3 del Virus de la Influenza A/patogenicidad , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/virología , Administración Intranasal , Animales , Anticuerpos Antivirales/biosíntesis , Quimiocinas/biosíntesis , Conjuntiva , Citocinas/biosíntesis , Femenino , Humanos , Subtipo H7N1 del Virus de la Influenza A/inmunología , Subtipo H7N3 del Virus de la Influenza A/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Macaca fascicularis , Infecciones por Orthomyxoviridae/inmunología
15.
Vet Microbiol ; 182: 108-15, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26711036

RESUMEN

Highly pathogenic avian influenza viruses (HPAIVs) have spread in both poultry and wild birds. Determining transmission routes of these viruses during an outbreak is essential for the control of avian influenza. It has been widely postulated that migratory ducks play crucial roles in the widespread dissemination of HPAIVs in poultry by carrying viruses along with their migrations; however close contacts between wild migratory ducks and poultry are less likely in modern industrial poultry farming settings. Therefore, we conducted experimental infections of HPAIVs and low pathogenic avian influenza viruses (LPAIVs) to chickens, domestic ducks, tree sparrows, jungle crows, and black rats to evaluate their roles in virus transmission. The results showed that chickens, ducks, sparrows, and crows were highly susceptible to HPAIV infection. Significant titers of virus were recovered from the sparrows and crows infected with HPAIVs, which suggests that they potentially play roles of transmission of HPAIVs to poultry. In contrast, the growth of LPAIVs was limited in each of the animals tested compared with that of HPAIVs. The present results indicate that these common synanthropes play some roles in influenza virus transmission from wild birds to poultry.


Asunto(s)
Aves , Reservorios de Enfermedades/veterinaria , Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Infecciones por Orthomyxoviridae/veterinaria , Animales , Animales Salvajes , Virus de la Influenza A/clasificación , Gripe Aviar/mortalidad , Infecciones por Orthomyxoviridae/virología , Ratas , Virulencia
16.
Antimicrob Agents Chemother ; 59(8): 4962-73, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26055368

RESUMEN

The number of patients infected with H7N9 influenza virus has been increasing since 2013. We examined the efficacy of neuraminidase (NA) inhibitors and the efficacy of a vaccine against an H7N9 influenza virus, A/Anhui/1/2013 (H7N9), isolated from a patient in a cynomolgus macaque model. NA inhibitors (oseltamivir and peramivir) barely reduced the total virus amount because of the emergence of resistant variants with R289K or I219T in NA [residues 289 and 219 in N9 of A/Anhui/1/2013 (H7N9) correspond to 292 and 222 in N2, respectively] in three of the six treated macaques, whereas subcutaneous immunization of an inactivated vaccine derived from A/duck/Mongolia/119/2008 (H7N9) prevented propagation of A/Anhui/1/2013 (H7N9) in all vaccinated macaques. The percentage of macaques in which variant H7N9 viruses with low sensitivity to the NA inhibitors were detected was much higher than that of macaques in which variant H5N1 highly pathogenic influenza virus was detected after treatment with one of the NA inhibitors in our previous study. The virus with R289K in NA was reported in samples from human patients, whereas that with I219T in NA was identified for the first time in this study using macaques, though no variant H7N9 virus was reported in previous studies using mice. Therefore, the macaque model enables prediction of the frequency of emerging H7N9 virus resistant to NA inhibitors in vivo. Since H7N9 strains resistant to NA inhibitors might easily emerge compared to other influenza viruses, monitoring of the emergence of variants is required during treatment of H7N9 influenza virus infection with NA inhibitors.


Asunto(s)
Antivirales/farmacología , Farmacorresistencia Viral/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Subtipo H7N9 del Virus de la Influenza A/efectos de los fármacos , Neuraminidasa/antagonistas & inhibidores , Ácidos Carbocíclicos , Animales , Ciclopentanos/farmacología , Farmacorresistencia Viral/inmunología , Femenino , Guanidinas/farmacología , Humanos , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H7N9 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Macaca , Ratones , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Oseltamivir/farmacología , Primates , Vacunación/métodos , Proteínas Virales/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Replicación Viral/inmunología
17.
Virus Genes ; 51(1): 57-68, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26036326

RESUMEN

Migratory water birds are the natural reservoir of influenza A viruses. H5 and H7 influenza viruses are isolated over the world and also circulate among poultry in Asia. In 2010, two H5N1 highly pathogenic avian influenza viruses (HPAIVs) were isolated from fecal samples of water birds on the flyway of migration from Siberia, Russia to the south in Hokkaido, Japan. H7N9 viruses are sporadically isolated from humans and circulate in poultry in China. To monitor whether these viruses have spread in the wild bird population, we conducted virological surveillance of avian influenza in migratory water birds in autumn from 2010 to 2014. A total of 8103 fecal samples from migratory water birds were collected in Japan and Mongolia, and 350 influenza viruses including 13 H5 and 19 H7 influenza viruses were isolated. A phylogenetic analysis revealed that all isolates are genetically closely related to viruses circulating among wild water birds. The results of the antigenic analysis indicated that the antigenicity of viruses in wild water birds is highly stable despite their nucleotide sequence diversity but is distinct from that of HPAIVs recently isolated in Asia. The present results suggest that HPAIVs and Chinese H7N9 viruses were not predominantly circulating in migratory water birds; however, continued monitoring of H5 and H7 influenza viruses both in domestic and wild birds is recommended for the control of avian influenza.


Asunto(s)
Antígenos Virales/análisis , Antígenos Virales/genética , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/virología , Animales , Aves , Análisis por Conglomerados , Heces/virología , Variación Genética , Japón , Datos de Secuencia Molecular , Mongolia , Filogenia , ARN Viral/genética , Análisis de Secuencia de ADN , Homología de Secuencia
18.
PLoS Pathog ; 10(6): e1004192, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24945244

RESUMEN

Highly pathogenic avian influenza (HPAI) viruses of the H5N1 subtype often cause severe pneumonia and multiple organ failure in humans, with reported case fatality rates of more than 60%. To develop a clinical antibody therapy, we generated a human-mouse chimeric monoclonal antibody (MAb) ch61 that showed strong neutralizing activity against H5N1 HPAI viruses isolated from humans and evaluated its protective potential in mouse and nonhuman primate models of H5N1 HPAI virus infections. Passive immunization with MAb ch61 one day before or after challenge with a lethal dose of the virus completely protected mice, and partial protection was achieved when mice were treated 3 days after the challenge. In a cynomolgus macaque model, reduced viral loads and partial protection against lethal infection were observed in macaques treated with MAb ch61 intravenously one and three days after challenge. Protective effects were also noted in macaques under immunosuppression. Though mutant viruses escaping from neutralization by MAb ch61 were recovered from macaques treated with this MAb alone, combined treatment with MAb ch61 and peramivir reduced the emergence of escape mutants. Our results indicate that antibody therapy might be beneficial in reducing viral loads and delaying disease progression during H5N1 HPAI virus infection in clinical cases and combined treatment with other antiviral compounds should improve the protective effects of antibody therapy against H5N1 HPAI virus infection.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Inmunización Pasiva/métodos , Subtipo H5N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/terapia , Ácidos Carbocíclicos , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Antivirales/uso terapéutico , Línea Celular , Ciclopentanos/uso terapéutico , Perros , Quimioterapia Combinada , Femenino , Guanidinas/uso terapéutico , Huésped Inmunocomprometido/inmunología , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Interleucina-6/sangre , Pulmón/patología , Pulmón/virología , Macaca fascicularis , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Neuraminidasa/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Carga Viral/inmunología
19.
Antimicrob Agents Chemother ; 58(8): 4795-803, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24913156

RESUMEN

Highly pathogenic avian influenza A (H5N1) viruses cause severe and often fatal disease in humans. We evaluated the efficacy of repeated intravenous dosing of the neuraminidase inhibitor peramivir against highly pathogenic avian influenza virus A/Vietnam/UT3040/2004 (H5N1) infection in cynomolgus macaques. Repeated dosing of peramivir (30 mg/kg/day once a day for 5 days) starting immediately after infection significantly reduced viral titers in the upper respiratory tract, body weight loss, and cytokine production and resulted in a significant body temperature reduction in infected macaques compared with that of macaques administered a vehicle (P < 0.05). Repeated administration of peramivir starting at 24 h after infection also resulted in a reduction in viral titers and a reduction in the period of virus detection in the upper respiratory tract, although the body temperature change was not statistically significant. The macaque model used in the present study demonstrated that inhibition of viral replication at an early time point after infection by repeated intravenous treatment with peramivir is critical for reduction of the production of cytokines, i.e., interleukin-6 (IL-6), tumor necrosis factor α, gamma interferon, monocyte chemotactic protein 1, and IL-12p40, resulting in amelioration of symptoms caused by highly pathogenic avian influenza virus infection.


Asunto(s)
Antivirales/farmacología , Ciclopentanos/farmacología , Guanidinas/farmacología , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/veterinaria , Ácidos Carbocíclicos , Administración Intravenosa , Animales , Temperatura Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/biosíntesis , Esquema de Medicación , Femenino , Subtipo H5N1 del Virus de la Influenza A/fisiología , Interferón gamma/antagonistas & inhibidores , Interferón gamma/biosíntesis , Subunidad p40 de la Interleucina-12/antagonistas & inhibidores , Subunidad p40 de la Interleucina-12/biosíntesis , Interleucina-6/antagonistas & inhibidores , Interleucina-6/biosíntesis , Macaca fascicularis , Infecciones por Orthomyxoviridae/fisiopatología , Infecciones por Orthomyxoviridae/virología , Factores de Tiempo , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Virulencia , Replicación Viral/efectos de los fármacos
20.
Vaccine ; 32(28): 3473-9, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24793949

RESUMEN

H7N9 influenza virus infection in humans was reported in China on March 31, 2013. Humans are immunologically naïve to the H7N9 subtype, for which the seasonal influenza vaccine is not effective. Thus, the development of an H7N9 influenza virus vaccine is an urgent issue. To prepare for the emergence of an influenza pandemic, we have established a library comprising more than 1300 influenza virus strains with 144 different combinations of 16 HA and 9 NA subtypes. An H7N9 virus strain isolated from a 35-year-old woman, A/Anhui/1/2013 (H7N9), was found to be antigenically similar to H7N9 influenza viruses isolated from migratory ducks. In the present study, the potency of an inactivated whole virus particle vaccine prepared from an H7N9 low pathogenic avian influenza virus, A/duck/Mongolia/119/2008 (H7N9), selected from the library, was assessed by a challenge with A/Anhui/1/2013 (H7N9). The results indicate that the test vaccine was potent enough to induce sufficient immunity to reduce the impact of disease caused by the challenge with A/Anhui/1/2013 (H7N9) in mice. The present results indicate that an inactivated whole virus particle vaccine prepared from an influenza virus strain stored in the library could be useful as a vaccine strain in case of an influenza pandemic.


Asunto(s)
Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Antígenos Virales/inmunología , Reacciones Cruzadas , Femenino , Subtipo H7N9 del Virus de la Influenza A/genética , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Filogenia , ARN Viral/genética , Vacunas de Productos Inactivados/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...