Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(4): 1013-1023.e13, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-32970990

RESUMEN

Understanding how potent neutralizing antibodies (NAbs) inhibit SARS-CoV-2 is critical for effective therapeutic development. We previously described BD-368-2, a SARS-CoV-2 NAb with high potency; however, its neutralization mechanism is largely unknown. Here, we report the 3.5-Å cryo-EM structure of BD-368-2/trimeric-spike complex, revealing that BD-368-2 fully blocks ACE2 recognition by occupying all three receptor-binding domains (RBDs) simultaneously, regardless of their "up" or "down" conformations. Also, BD-368-2 treats infected adult hamsters at low dosages and at various administering windows, in contrast to placebo hamsters that manifested severe interstitial pneumonia. Moreover, BD-368-2's epitope completely avoids the common binding site of VH3-53/VH3-66 recurrent NAbs, evidenced by tripartite co-crystal structures with RBDs. Pairing BD-368-2 with a potent recurrent NAb neutralizes SARS-CoV-2 pseudovirus at pM level and rescues mutation-induced neutralization escapes. Together, our results rationalized a new RBD epitope that leads to high neutralization potency and demonstrated BD-368-2's therapeutic potential in treating COVID-19.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Betacoronavirus/inmunología , Infecciones por Coronavirus/patología , Neumonía Viral/patología , Animales , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/química , Anticuerpos Antivirales/uso terapéutico , Reacciones Antígeno-Anticuerpo , Sitios de Unión , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Cricetinae , Microscopía por Crioelectrón , Modelos Animales de Enfermedad , Epítopos/química , Epítopos/inmunología , Femenino , Pulmón/patología , Masculino , Simulación de Dinámica Molecular , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Estructura Cuaternaria de Proteína , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología
2.
Cell ; 182(3): 713-721.e9, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32778225

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) threatens global public health. The development of a vaccine is urgently needed for the prevention and control of COVID-19. Here, we report the pilot-scale production of an inactivated SARS-CoV-2 vaccine candidate (BBIBP-CorV) that induces high levels of neutralizing antibodies titers in mice, rats, guinea pigs, rabbits, and nonhuman primates (cynomolgus monkeys and rhesus macaques) to provide protection against SARS-CoV-2. Two-dose immunizations using 2 µg/dose of BBIBP-CorV provided highly efficient protection against SARS-CoV-2 intratracheal challenge in rhesus macaques, without detectable antibody-dependent enhancement of infection. In addition, BBIBP-CorV exhibits efficient productivity and good genetic stability for vaccine manufacture. These results support the further evaluation of BBIBP-CorV in a clinical trial.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Evaluación Preclínica de Medicamentos/métodos , Pandemias/prevención & control , Neumonía Viral/prevención & control , Vacunas de Productos Inactivados/uso terapéutico , Vacunas Virales/uso terapéutico , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Betacoronavirus/genética , COVID-19 , Vacunas contra la COVID-19 , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Femenino , Cobayas , Inmunogenicidad Vacunal , Macaca fascicularis , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos BALB C , Filogenia , Neumonía Viral/virología , Conejos , Ratas , Ratas Wistar , SARS-CoV-2 , Vacunas de Productos Inactivados/efectos adversos , Células Vero , Vacunas Virales/efectos adversos
3.
Cell ; 182(1): 73-84.e16, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32425270

RESUMEN

The COVID-19 pandemic urgently needs therapeutic and prophylactic interventions. Here, we report the rapid identification of SARS-CoV-2-neutralizing antibodies by high-throughput single-cell RNA and VDJ sequencing of antigen-enriched B cells from 60 convalescent patients. From 8,558 antigen-binding IgG1+ clonotypes, 14 potent neutralizing antibodies were identified, with the most potent one, BD-368-2, exhibiting an IC50 of 1.2 and 15 ng/mL against pseudotyped and authentic SARS-CoV-2, respectively. BD-368-2 also displayed strong therapeutic and prophylactic efficacy in SARS-CoV-2-infected hACE2-transgenic mice. Additionally, the 3.8 Å cryo-EM structure of a neutralizing antibody in complex with the spike-ectodomain trimer revealed the antibody's epitope overlaps with the ACE2 binding site. Moreover, we demonstrated that SARS-CoV-2-neutralizing antibodies could be directly selected based on similarities of their predicted CDR3H structures to those of SARS-CoV-neutralizing antibodies. Altogether, we showed that human neutralizing antibodies could be efficiently discovered by high-throughput single B cell sequencing in response to pandemic infectious diseases.


Asunto(s)
Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/aislamiento & purificación , Linfocitos B/inmunología , Infecciones por Coronavirus/inmunología , Neumonía Viral/inmunología , Análisis de la Célula Individual , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/metabolismo , COVID-19 , Convalecencia , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Pandemias , Análisis de Secuencia de ARN , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Exones VDJ
4.
Nature ; 583(7818): 830-833, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32380511

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of coronavirus disease 2019 (COVID-19), which has become a public health emergency of international concern1. Angiotensin-converting enzyme 2 (ACE2) is the cell-entry receptor for severe acute respiratory syndrome coronavirus (SARS-CoV)2. Here we infected transgenic mice that express human ACE2 (hereafter, hACE2 mice) with SARS-CoV-2 and studied the pathogenicity of the virus. We observed weight loss as well as virus replication in the lungs of hACE2 mice infected with SARS-CoV-2. The typical histopathology was interstitial pneumonia with infiltration of considerable numbers of macrophages and lymphocytes into the alveolar interstitium, and the accumulation of macrophages in alveolar cavities. We observed viral antigens in bronchial epithelial cells, macrophages and alveolar epithelia. These phenomena were not found in wild-type mice infected with SARS-CoV-2. Notably, we have confirmed the pathogenicity of SARS-CoV-2 in hACE2 mice. This mouse model of SARS-CoV-2 infection will be valuable for evaluating antiviral therapeutic agents and vaccines, as well as understanding the pathogenesis of COVID-19.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Pulmón/patología , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/patología , Neumonía Viral/virología , Transgenes , Enzima Convertidora de Angiotensina 2 , Animales , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Betacoronavirus/inmunología , Betacoronavirus/metabolismo , Bronquios/patología , Bronquios/virología , COVID-19 , Infecciones por Coronavirus/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Humanos , Inmunoglobulina G/inmunología , Pulmón/inmunología , Pulmón/virología , Linfocitos/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/virología , Masculino , Ratones , Ratones Transgénicos , Pandemias , Neumonía Viral/inmunología , Receptores de Complemento 3d/genética , Receptores de Complemento 3d/metabolismo , SARS-CoV-2 , Replicación Viral , Pérdida de Peso
5.
J Virol ; 98(2): e0135823, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38226810

RESUMEN

Hand, foot, and mouth disease (HFMD) is caused by more than 20 pathogenic enteroviruses belonging to the Picornaviridae family and Enterovirus genus. Since the introduction of the enterovirus-71 (EV71) vaccine in 2016, the number of HFMD cases caused by EV71 has decreased. However, cases of infections caused by other enteroviruses, such as coxsackievirus A6 (CA6) and coxsackievirus A10, have been increasing accordingly. In this study, we used a clinical isolate of CA6 to establish an intragastric infection mouse model using 7-day-old mice to mimic the natural transmission route, by which we investigated the differential gene expression profiles associated with virus infection and pathogenicity. After intragastric infection, mice exhibited hind limb paralysis symptoms and weight loss, similar to those reported for EV71 infection in mice. The skeletal muscle was identified as the main site of virus replication, with a peak viral load reaching 2.31 × 107 copies/mg at 5 dpi and increased infiltration of inflammatory cells. RNA sequencing analysis identified differentially expressed genes (DEGs) after CA6 infection. DEGs in the blood, muscle, brain, spleen, and thymus were predominantly enriched in immune system responses, including pathways such as Toll-like receptor signaling and PI3K-Akt signaling. Our study has unveiled the genes involved in the host immune response during CA6 infection, thereby enhancing our comprehension of the pathological mechanism of HFMD.IMPORTANCEThis study holds great significance for the field of hand, foot, and mouth disease (HFMD). It not only delves into the disease's etiology, transmission pathways, and severe complications but also establishes a novel mouse model that mimics the natural coxsackievirus A6 infection process, providing a pivotal platform to delve deeper into virus replication and pathogenic mechanisms. Additionally, utilizing RNA-seq technology, it unveils the dynamic gene expression changes during infection, offering valuable leads for identifying novel therapeutic drug targets. This research has the potential to enhance our understanding of HFMD, offering fresh perspectives for disease prevention and treatment and positively impacting children's health worldwide.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Enfermedad de Boca, Mano y Pie , Animales , Niño , Humanos , Ratones , Anticuerpos Antivirales , Modelos Animales de Enfermedad , Enterovirus/patogenicidad , Enterovirus/fisiología , Enterovirus Humano A , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Expresión Génica , Enfermedad de Boca, Mano y Pie/genética , Fosfatidilinositol 3-Quinasas , Virulencia
6.
Nano Lett ; 24(25): 7662-7671, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38870422

RESUMEN

Extensive investigations have proven the effectiveness of elastic binders in settling the challenge of structural damage posed by volume expansion of high-capacity anode used in nanoscale silicon. However, the sluggish ionic conductivity of polymer binder severely restricts the electrode reactions, making it unsuitable for practical applications. Inspired by the biological tissues with rapid neurotransmission and robust muscles, we propose a biomimetic binder that contains ionic conductive polymer (by polymerization reaction of poly(ethylene glycol) diglycidyl ether and polyethylenimine) and rigid polymer backbone (polyacrylic acid), which can effectively mitigate both Li-ion transport resistance and lithiation stress to stabilize the silicon nanoparticles during cycles. Consequently, the silicon anode with biomimetic binder achieves a rate capability of 1897 mAh g-1 at 8.0 A g-1 and capacity retention of 87% after 150 cycles under areal capacity upon 3.0 mAh cm-2. These results demonstrate the possibility of decoupling ionic conductivity from mechanical properties toward practical high-capacity anodes for energy-dense batteries.

7.
Proteomics ; 23(16): e2300096, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37309728

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected hundreds of millions of people all over the world and thus threatens human life. Clinical evidence shows that SARS-CoV-2 infection can cause several neurological consequences, but the existing antiviral drugs and vaccines have failed to stop its spread. Therefore, an understanding of the response to SARS-CoV-2 infection of hosts is vital to find a resultful therapy. Here, we employed a K18-hACE2 mouse infection model and LC-MS/MS to systematically evaluate the acetylomes of brain cortexes in the presence and absence of SARS-CoV-2 infection. Using a label-free strategy, 3829 lysine acetylation (Kac) sites in 1735 histone and nonhistone proteins were identified. Bioinformatics analyses indicated that SARS-CoV-2 infection might lead to neurological consequences via acetylation or deacetylation of important proteins. According to a previous study, we found 26 SARS-CoV-2 proteins interacted with 61 differentially expressed acetylated proteins with high confidence and identified one acetylated SARS-CoV-2 protein nucleocapsid phosphoprotein. We greatly expanded the known set of acetylated proteins and provide the first report of the brain cortex acetylome in this model and thus a theoretical basis for future research on the pathological mechanisms and therapies of neurological consequences after SARS-CoV-2 infection.


Asunto(s)
COVID-19 , SARS-CoV-2 , Ratones , Humanos , Animales , SARS-CoV-2/metabolismo , COVID-19/patología , Lisina/metabolismo , Acetilación , Cromatografía Liquida , Peptidil-Dipeptidasa A/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Espectrometría de Masas en Tándem , Encéfalo/metabolismo , Ratones Transgénicos , Modelos Animales de Enfermedad
8.
Virol J ; 19(1): 212, 2022 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494863

RESUMEN

The COVID-19 pandemic, caused by the SARS-CoV-2 virus and its variants, has posed unprecedented challenges worldwide. Existing vaccines have limited effectiveness against SARS-CoV-2 variants. Therefore, novel vaccines to match mutated viral lineages by providing long-term protective immunity are urgently needed. We designed a recombinant adeno-associated virus 5 (rAAV5)-based vaccine (rAAV-COVID-19) by using the SARS-CoV-2 spike protein receptor binding domain (RBD-plus) sequence with both single-stranded (ssAAV5) and self-complementary (scAAV5) delivery vectors and found that it provides excellent protection from SARS-CoV-2 infection. A single-dose vaccination in mice induced a robust immune response; induced neutralizing antibody (NA) titers were maintained at a peak level of over 1:1024 more than a year post-injection and were accompanied by functional T-cell responses. Importantly, both ssAAV- and scAAV-based RBD-plus vaccines produced high levels of serum NAs against the circulating SARS-CoV-2 variants, including Alpha, Beta, Gamma and Delta. A SARS-CoV-2 virus challenge showed that the ssAAV5-RBD-plus vaccine protected both young and old mice from SARS-CoV-2 infection in the upper and lower respiratory tracts. Whole genome sequencing demonstrated that AAV vector DNA sequences were not found in the genomes of vaccinated mice one year after vaccination, demonstrating vaccine safety. These results suggest that the rAAV5-based vaccine is safe and effective against SARS-CoV-2 and several variants as it provides long-term protective immunity. This novel vaccine has a significant potential for development into a human prophylactic vaccination to help end the global pandemic.


Asunto(s)
COVID-19 , Parvovirinae , Animales , Humanos , Ratones , SARS-CoV-2/genética , COVID-19/prevención & control , Pandemias , Vacunas Sintéticas/genética , Glicoproteína de la Espiga del Coronavirus/genética , Anticuerpos Neutralizantes , Anticuerpos Antivirales
9.
J Infect Dis ; 223(8): 1313-1321, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33605423

RESUMEN

Domestic cats, an important companion animal, can be infected with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). This has aroused concern regarding the ability of domestic cats to spread the virus that causes coronavirus disease 2019. We systematically demonstrated the pathogenesis and transmissibility of SARS-CoV-2 in cats. Serial passaging of the virus between cats dramatically attenuated the viral transmissibility, likely owing to variations of the amino acids in the receptor-binding domain sites of angiotensin-converting enzyme 2 between humans and cats. These findings provide insight into the transmissibility of SARS-CoV-2 in cats and information for protecting the health of humans and cats.


Asunto(s)
COVID-19/transmisión , COVID-19/veterinaria , SARS-CoV-2/patogenicidad , Aminoácidos/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/metabolismo , Gatos , Línea Celular , Chlorocebus aethiops , Femenino , Humanos , Masculino , Células Vero
11.
J Infect Dis ; 222(4): 551-555, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32444876

RESUMEN

We simulated 3 transmission modes, including close-contact, respiratory droplets and aerosol routes, in the laboratory. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can be highly transmitted among naive human angiotensin-converting enzyme 2 (hACE2) mice via close contact because 7 of 13 naive hACE2 mice were SARS-CoV-2 antibody seropositive 14 days after being introduced into the same cage with 3 infected-hACE2 mice. For respiratory droplets, SARS-CoV-2 antibodies from 3 of 10 naive hACE2 mice showed seropositivity 14 days after introduction into the same cage with 3 infected-hACE2 mice, separated by grids. In addition, hACE2 mice cannot be experimentally infected via aerosol inoculation until continued up to 25 minutes with high viral concentrations.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/transmisión , Neumonía Viral/transmisión , Aerosoles , Canal Anal/virología , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Antivirales/sangre , Betacoronavirus/genética , Betacoronavirus/inmunología , Betacoronavirus/aislamiento & purificación , COVID-19 , Chlorocebus aethiops , Femenino , Humanos , Inmunoglobulina G/sangre , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Transgénicos , Pandemias , Peptidil-Dipeptidasa A/genética , Faringe/virología , ARN Viral/aislamiento & purificación , Sistema Respiratorio/virología , Riesgo , SARS-CoV-2 , Organismos Libres de Patógenos Específicos , Factores de Tiempo , Células Vero , Carga Viral , Pérdida de Peso
12.
Virol J ; 17(1): 58, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32321526

RESUMEN

BACKGROUND: Prophylactic vaccines are critical in preventing hand, foot, and mouth disease (HFMD) primarily caused by human enterovirus 71 (EV71) infection. Children aged less than 5 years are especially susceptible to EV71 infections. In addition to the development of vaccines containing the inactivated virus, those containing virus-like particles (VLPs) with repeated antigens also constitute an effective preventive strategy for EV71 infections, with safety and productivity advantages. We previously developed a fusion protein composed with truncated peptides of the EV71 capsid protein, which assembled into spherical particles. This study aimed to assess the immunoprotective effects of this fusion protein as a vaccine candidate in a mouse model of EV71 infection. METHODS: To evaluate the protective effect of fusion protein vaccine candidate, neonatal mice born by immunized female mice, as well as normal neonatal mice immunized twice were infected with EV71 virus. Whereafter, the survival rates, clinical scores and viral loads were measured. RESULTS: The high dosage and booster immunization helped induce specific serum antibodies with high neutralization titers, which were transferred to neonatal mice, thereby facilitating effective resistance towards EV71 infection. An active immune response was also observed in neonatal mice which generated following immunization. CONCLUSIONS: The present results suggest that this fusion protein is a suitable vaccine candidate in treating EV71 infections.


Asunto(s)
Enterovirus Humano A/genética , Infecciones por Enterovirus/prevención & control , Péptidos/inmunología , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Línea Celular Tumoral , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/inmunología , Femenino , Humanos , Inmunización Secundaria , Ratones , Ratones Endogámicos BALB C , Péptidos/genética , Proteínas Virales de Fusión/administración & dosificación , Carga Viral , Vacunas Virales/genética
14.
J Asian Nat Prod Res ; 22(12): 1188-1196, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33176482

RESUMEN

There are no effective antiviral drugs to treat hand, foot, and mouth disease. In this study, a series of lycorine derivatives were synthesized and evaluated against enterovirus 71 and coxsackievirus A16 in vitro. Derivatives 7c-m with the phenoxyacyl group at the C-1 position showed higher efficacy and lower toxicity than lycorine. In addition, derivative 7e enhanced the survival rate to 40% in the mouse model of the lethal EV71 infection.


Asunto(s)
Antivirales , Enterovirus Humano A , Enterovirus , Enfermedad de Boca, Mano y Pie , Alcaloides de Amaryllidaceae , Animales , Antivirales/farmacología , Ratones , Estructura Molecular , Fenantridinas
16.
Bioorg Med Chem Lett ; 25(17): 3526-8, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26189894

RESUMEN

Recent outbreak of hand, foot and mouse disease caused by enterovirus 71 is a serious threat to infants and children in Asia-Pacific countries. No vaccines or antiviral therapies are available against this infection. In this study, we found that quinolizidine alkaloids could improve the levels of T cells in mice. The structure-activity relationships demonstrated that the oxidation and double bonds are important for the excellent potency. Oxysophocarpine, the most effective compound of six quinolizidine alkaloids, could also relieve symptoms and reduce mortality in lethal enterovirus 71-infected mice through compensating for the decreased levels of T cells. This work suggested that quinolizidine alkaloids have the potential against enterovirus 71 for further development of novel antiviral drugs.


Asunto(s)
Alcaloides/uso terapéutico , Antivirales/uso terapéutico , Enterovirus Humano A/efectos de los fármacos , Enfermedad de Boca, Mano y Pie/tratamiento farmacológico , Quinolizidinas/uso terapéutico , Linfocitos T/efectos de los fármacos , Alcaloides/química , Animales , Antivirales/química , Enterovirus Humano A/inmunología , Enfermedad de Boca, Mano y Pie/inmunología , Humanos , Ratones Endogámicos BALB C , Quinolizidinas/química , Relación Estructura-Actividad , Linfocitos T/inmunología , Linfocitos T/virología
17.
J Infect Dis ; 209(4): 551-6, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23990570

RESUMEN

The outbreak of human infections caused by novel avian-origin influenza A(H7N9) in China since March 2013 underscores the need to better understand the pathogenicity and transmissibility of these viruses in mammals. In a ferret model, the pathogenicity of influenza A(H7N9) was found to be less than that of an influenza A(H5N1) strain but comparable to that of 2009 pandemic influenza A(H1N1), based on the clinical signs, mortality, virus dissemination, and results of histopathologic analyses. Influenza A(H7N9) could replicate in the upper and lower respiratory tract, the heart, the liver, and the olfactory bulb. It is worth noting that influenza A(H7N9) exhibited a low level of transmission between ferrets via respiratory droplets. There were 4 mutations in the virus isolated from the contact ferret: D678Y in the gene encoding PB2, R157K in the gene encoding hemagglutinin (H3 numbering), I109T in the gene encoding nucleoprotein, and T10I in the gene encoding neuraminidase. These data emphasized that avian-origin influenza A(H7N9) can be transmitted between mammals, highlighting its potential for human-to-human transmissibility.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , Administración Intranasal , Animales , Peso Corporal , Modelos Animales de Enfermedad , Exposición a Riesgos Ambientales , Hurones/virología , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Pulmón/química , Pulmón/patología , Pulmón/virología , Cavidad Nasal/virología , Faringe/virología
18.
Virol J ; 11: 185, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25367670

RESUMEN

BACKGROUND: On 19 February 2013, the first patient infected with a novel influenza A H7N9 virus from an avian source showed symptoms of sickness. More than 349 laboratory-confirmed cases and 109 deaths have been reported in mainland China since then. Laboratory-confirmed, human-to-human H7N9 virus transmission has not been documented between individuals having close contact; however, this transmission route could not be excluded for three families. To control the spread of the avian influenza H7N9 virus, we must better understand its pathogenesis, transmissibility, and transmission routes in mammals. Studies have shown that this particular virus is transmitted by aerosols among ferrets. METHODS: To study potential transmission routes in animals with direct or close contact to other animals, we investigated these factors in a murine model. RESULTS: Viable H7N9 avian influenza virus was detected in the upper and lower respiratory tracts, intestine, and brain of model mice. The virus was transmissible between mice in close contact, with a higher concentration of virus found in pharyngeal and ocular secretions, and feces. All these biological materials were contagious for naïve mice. CONCLUSIONS: Our results suggest that the possible transmission routes for the H7N9 influenza virus were through mucosal secretions and feces.


Asunto(s)
Modelos Animales de Enfermedad , Subtipo H7N9 del Virus de la Influenza A/fisiología , Gripe Humana/transmisión , Animales , Embrión de Pollo , Femenino , Historia Antigua , Humanos , Subtipo H7N9 del Virus de la Influenza A/genética , Gripe Humana/virología , Ratones , Ratones Endogámicos BALB C
19.
Mar Drugs ; 12(7): 4086-95, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-25003792

RESUMEN

Enterovirus 71 is one of the major causative agents of hand, foot and mouth disease in children under six years of age. No vaccine or antiviral therapy is currently available. In this work, we found that the number of B cells was reduced in enterovirus 71-infected mice. Deferoxamine, a marine microbial natural product, compensated for the decreased levels of B cells caused by enterovirus 71 infection. The neutralizing antibody titer was also improved after deferoxamine treatment. Furthermore, deferoxamine relieved symptoms and reduced mortality and muscle damage caused by enterovirus 71 infection. This work suggested that deferoxamine has the potential for further development as a B cell-immunomodulator against enterovirus 71.


Asunto(s)
Linfocitos B/efectos de los fármacos , Deferoxamina/uso terapéutico , Enterovirus Humano A , Infecciones por Enterovirus/tratamiento farmacológico , Animales , Deferoxamina/farmacología , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/mortalidad , Ratones , Carga Viral
20.
J Innate Immun ; 16(1): 80-95, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38224674

RESUMEN

BACKGROUND: Neutrophils are the first line of defense against pathogens. They are divided into multiple subpopulations during development and kill pathogens through various mechanisms. Neutrophils are considered one of the markers of severe COVID-19. SUMMARY: In-depth research has revealed that neutrophil subpopulations have multiple complex functions. Different subsets of neutrophils play an important role in the progression of COVID-19. KEY MESSAGES: In this review, we provide a detailed overview of the developmental processes of neutrophils at different stages and their recruitment and activation after SARS-CoV-2 infection, aiming to elucidate the changes in neutrophil subpopulations, characteristics, and functions after infection and provide a reference for mechanistic research on neutrophil subpopulations in the context of SARS-CoV-2 infection. In addition, we have also summarized research progress on potential targeted drugs for neutrophil immunotherapy, hoping to provide information that aids the development of therapeutic drugs for the clinical treatment of critically ill COVID-19 patients.


Asunto(s)
COVID-19 , Neutrófilos , SARS-CoV-2 , Humanos , COVID-19/inmunología , Neutrófilos/inmunología , SARS-CoV-2/inmunología , Inmunoterapia/métodos , Activación Neutrófila , Animales , Infiltración Neutrófila/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA