Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Virus Res ; 345: 199378, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38643857

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant threat to human health globally. It is crucial to develop a vaccine to reduce the effect of the virus on public health, economy, and society and regulate the transmission of SARS-CoV-2. Influenza B virus (IBV) can be used as a vector that does not rely on the current circulating influenza A strains. In this study, we constructed an IBV-based vector vaccine by inserting a receptor-binding domain (RBD) into a non-structural protein 1 (NS1)-truncated gene (rIBV-NS110-RBD). Subsequently, we assessed its safety, immunogenicity, and protective efficacy against SARS-CoV-2 in mice, and observed that it was safe in a mouse model. Intranasal administration of a recombinant rIBV-NS110-RBD vaccine induced high levels of SARS-CoV-2-specific IgA and IgG antibodies and T cell-mediated immunity in mice. Administering two doses of the intranasal rIBV-NS110-RBD vaccine significantly reduced the viral load and lung damage in mice. This novel IBV-based vaccine offers a novel approach for controlling the SARS-CoV-2 pandemic.

2.
J Med Virol ; 96(1): e29336, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38193530

RESUMEN

Based on the forefront of clinical research, there is a growing recognition that the gut microbiota, which plays a pivotal role in shaping both the innate and adaptive immune systems, may significantly contribute to the pathogenesis of coronavirus disease 2019 (COVID-19). Although an association between altered gut microbiota and COVID-19 pathogenesis has been established, the causative mechanisms remain incompletely understood. Additionally, the validation of the precise functional alterations within the gut microbiota relevant to COVID-19 pathogenesis has been limited by a scarcity of suitable animal experimental models. In the present investigation, we employed a newly developed humanized ACE2 knock-in (hACE2-KI) mouse model, capable of recapitulating critical aspects of pulmonary and intestinal infection, to explore the modifications in the gut microbiota following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Examination of fecal samples using 16S rRNA gene profiling unveiled a notable reduction in species richness and conspicuous alterations in microbiota composition at 6 days postinfection (dpi). These alterations were primarily characterized by a decline in beneficial bacterial species and an escalation in certain opportunistic pathogens. Moreover, our analysis entailed a correlation study between the gut microbiota and plasma cytokine concentrations, revealing the potential involvement of the Lachnospiraceae_NK4A136_group and unclassified_f_Lachnospiraceae genera in attenuating hyperinflammatory responses triggered by the infection. Furthermore, integration of gut microbiota data with RNA-seq analysis results suggested that the increased presence of Staphylococcus in fecal samples may signify the potential for bacterial coinfection in lung tissues via gut translocation. In summary, our hACE2-KI mouse model effectively recapitulated the observed alterations in the gut microbiota during SARS-CoV-2 infection. This model presents a valuable tool for elucidating gut microbiota-targeted strategies aimed at mitigating COVID-19.


Asunto(s)
COVID-19 , Microbioma Gastrointestinal , Animales , Ratones , SARS-CoV-2 , ARN Ribosómico 16S/genética , Modelos Animales de Enfermedad
3.
Cell Mol Immunol ; 20(12): 1457-1471, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37978243

RESUMEN

The G protein-coupled receptor ADGRE5 (CD97) binds to various metabolites that play crucial regulatory roles in metabolism. However, its function in the antiviral innate immune response remains to be determined. In this study, we report that CD97 inhibits virus-induced type-I interferon (IFN-I) release and enhances RNA virus replication in cells and mice. CD97 was identified as a new negative regulator of the innate immune receptor RIG-I, and RIG-1 degradation led to the suppression of the IFN-I signaling pathway. Furthermore, overexpression of CD97 promoted the ubiquitination of RIG-I, resulting in its degradation, but did not impact its mRNA expression. Mechanistically, CD97 upregulates RNF125 expression to induce RNF125-mediated RIG-I degradation via K48-linked ubiquitination at Lys181 after RNA virus infection. Most importantly, CD97-deficient mice are more resistant than wild-type mice to RNA virus infection. We also found that sanguinarine-mediated inhibition of CD97 effectively blocks VSV and SARS-CoV-2 replication. These findings elucidate a previously unknown mechanism through which CD97 negatively regulates RIG-I in the antiviral innate immune response and provide a molecular basis for the development of new therapeutic strategies and the design of targeted antiviral agents.


Asunto(s)
Infecciones por Virus ARN , Virus ARN , Animales , Ratones , Antivirales/farmacología , Proteína 58 DEAD Box/metabolismo , Inmunidad Innata , Receptores Acoplados a Proteínas G/metabolismo , Infecciones por Virus ARN/genética , Virus ARN/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
4.
J Virol ; 97(11): e0110123, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37916835

RESUMEN

IMPORTANCE: Clade 2.3.4.4 H5Nx avian influenza viruses (AIVs) have circulated globally and caused substantial economic loss. Increasing numbers of humans have been infected with Clade 2.3.4.4 H5N6 AIVs in recent years. Only a few human influenza vaccines have been licensed to date. However, the licensed live attenuated influenza virus vaccine exhibited the potential of being recombinant with the wild-type influenza A virus (IAV). Therefore, we developed a chimeric cold-adapted attenuated influenza vaccine based on the Clade 2.3.4.4 H5 AIVs. These H5 vaccines demonstrate the advantage of being non-recombinant with circulated IAVs in the future influenza vaccine study. The findings of our current study reveal that these H5 vaccines can induce cross-reactive protective efficacy in mice and ferrets. Our H5 vaccines may provide a novel option for developing human-infected Clade 2.3.4.4 H5 AIV vaccines.


Asunto(s)
Protección Cruzada , Virus de la Influenza A , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae , Animales , Ratones , Anticuerpos Antivirales , Hurones , Gripe Aviar , Vacunas contra la Influenza/genética , Vacunas Atenuadas , Infecciones por Orthomyxoviridae/prevención & control
5.
Chin Med ; 18(1): 144, 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37919750

RESUMEN

BACKGROUND: Influenza viruses, especially Influenza A virus and Influenza B virus, are respiratory pathogens and can cause seasonal epidemics and pandemics. Severe influenza viruses infection induces strong host-defense response and excessive inflammatory response, resulting in acute lung damage, multiple organ failure and high mortality. Isoquercitrin is a Chinese medicine monomer, which was reported to have multiple biological activities, including antiviral activity against HSV, IAV, SARS-CoV-2 and so on. Aims of this study were to assess the in vitro anti-IAV and anti-IBV activity, evaluate the in vivo protective efficacy against lethal infection of the influenza virus and searched for the more optimal method of drug administration of isoquercitrin. METHODS: In vitro infection model (MDCK and A549 cells) and mouse lethal infection model of Influenza A virus and Influenza B virus were used to evaluate the antiviral activity of isoquercitrin. RESULTS: Isoquercitrin could significantly suppress the replication in vitro and in vivo and reduced the mortality of mouse lethal infection models. Compared with virus infection group, isoquercitrin mitigated lung and multiple organ damage. Moreover, isoquercitrin blocked hyperproduction of cytokines induced by virus infection via inactivating NF-κB signaling. Among these routes of isoquercitrin administration, intramuscular injection is a better drug delivery method. CONCLUSION: Isoquercitrin is a potential Chinese medicine monomer Against Influenza A Virus and Influenza B Virus infection.

6.
ACS Omega ; 8(29): 26055-26064, 2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37521655

RESUMEN

Porous Bi2O3-Bi2S3 composite sheets were constructed through a combinational methodology of chemical bath deposition and hydrothermal reaction. The Na2S precursor concentration in the hydrothermal solution was varied to understand the correlation between the vulcanization degree and structure evolution of the porous Bi2O3-Bi2S3 composite sheets. The control of the etching rate of the Bi2O3 sheet template and the regrowth rate of Bi2S3 crystallites via suitable sulfide precursor concentration during the hydrothermal reaction utilizes the formation of porous Bi2O3-Bi2S3 sheets. Due to the presence of Bi2S3 crystallites and porous structure in the Bi2O3-Bi2S3 composites, the improved visible-light absorption ability and separation efficiency of photogenerated charge carriers are achieved. Furthermore, the as-synthesized Bi2O3-Bi2S3 composite sheets obtained from vulcanization with a 0.01M Na2S precursor display highly enhanced photocatalytic degradation toward methyl orange (MO) dyes compared with the pristine Bi2O3 and Bi2S3. The porous Bi2O3-Bi2S3 sheet system shows high surface active sites, fast transfer, high-efficiency separation of photoinduced charge carriers, and enhanced redox capacity concerning their constituent counterparts. This study affords a promising approach to constructing Bi2O3-based Z-scheme composites with a suitable microstructure and Bi2O3/Bi2S3 phase ratio for photoactive device applications.

7.
Virology ; 585: 145-154, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37348143

RESUMEN

The SARS-CoV-2 pandemic has continued for about three years since emerging in late December 2019, resulting in millions of deaths. Therefore, there is an urgent need to develop a safe and effective vaccine to control SARS-CoV-2. In this study, we developed a bacterium-like particle vaccine that displays the SARS-CoV-2 receptor binding domain (RBD) (named Trim-RBD-GEM) using the GEM-PA system. We evaluated the immunogenicity and protective efficacy of the Trim-RBD-GEM vaccine with the oil-in-water adjuvant AddaVax in C57BL/6 N mice intramuscularly. We found that Trim-RBD-GEM&AddaVax induced high levels of humoral immunity in C57BL/6 N mice. Additionally, the lung virus loads in the immunized group were significantly decreased compared to the adjuvant control and mock groups. Therefore, this vaccine provides protection against lethal infection in a C57BL/6 N mouse model. Our Trim-RBD-GEM&AddaVax vaccine is potentially a promising, rapid, and safe subunit vaccine for preventing and controlling SARS-CoV-2.


Asunto(s)
COVID-19 , Vacunas , Animales , Ratones , Ratones Endogámicos C57BL , COVID-19/prevención & control , SARS-CoV-2/genética , Adyuvantes Inmunológicos , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Antivirales , Anticuerpos Neutralizantes
8.
Front Microbiol ; 14: 1175188, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37350787

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transmission is responsible for the coronavirus disease 2019 (COVID-19) pandemic. SARS-CoV-2 uses the angiotensin-converting enzyme 2 (ACE2) receptor to enter the host, and the gastrointestinal tract is a potential infection site as this receptor is expressed on it. Multiple studies have indicated that an increasing number of COVID-19 patients presented with gastrointestinal symptoms that are highly associated with disease severity. Moreover, emerging evidence has demonstrated that alterations in the gut immune microenvironment induced by intestinal SARS-CoV-2 infection can regulate respiratory symptoms. Therefore, targeting the intestines may be a candidate therapeutic strategy in patients with COVID-19; however, no mouse model can serve as an appropriate infection model for the development of fatal pneumonia while mimicking intestinal infection. In this study, a novel human ACE2 knock-in (KI) mouse model (or hACE2-KI) was systemically compared with the popular K18-hACE2 mice; it showed differences in the distribution of lung and intestinal infections and pathophysiological characteristics. These newly generated hACE2-KI mice were susceptible to intranasal infection with SARS-CoV-2, and not only developed mild to severe lung injury, but also acquired intestinal infection. Consequently, this model can be a useful tool for studying intestinal SARS-CoV-2 infection and developing effective therapeutic strategies.

9.
J Virol ; 97(3): e0001123, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36877072

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the viral pathogen responsible for the worldwide coronavirus disease 2019 (COVID-19) pandemic. The novel SARS-CoV-2 ORF8 protein is not highly homologous with known proteins, including accessory proteins of other coronaviruses. ORF8 contains a 15-amino-acid signal peptide in the N terminus that localizes the mature protein to the endoplasmic reticulum. Oligomannose-type glycosylation has been identified at the N78 site. Here, the unbiased molecular functions of ORF8 are also demonstrated. Via an immunoglobulin-like fold in a glycan-independent manner, both exogenous and endogenous ORF8 interacts with human calnexin and HSPA5. The key ORF8-binding sites of Calnexin and HSPA5 are indicated on the globular domain and the core substrate-binding domain, respectively. ORF8 induces species-dependent endoplasmic reticulum stress-like responses in human cells exclusively via the IRE1 branch, including intensive HSPA5 and PDIA4 upregulation, with increases in other stress-responding effectors, including CHOP, EDEM and DERL3. ORF8 overexpression facilitates SARS-CoV-2 replication. Both stress-like responses and viral replication induced by ORF8 have been shown to result from triggering the Calnexin switch. Thus, ORF8 serves as a key unique virulence gene of SARS-CoV-2, potentially contributing to COVID-19-specific and/or human-specific pathogenesis. IMPORTANCE Although SARS-CoV-2 is basically regarded as a homolog of SARS-CoV, with their genomic structure and the majority of their genes being highly homologous, the ORF8 genes of SARS-CoV and SARS-CoV-2 are distinct. The SARS-CoV-2 ORF8 protein also shows little homology with other viral or host proteins and is thus regarded as a novel special virulence gene of SARS-CoV-2. The molecular function of ORF8 has not been clearly known until now. Our results reveal the unbiased molecular characteristics of the SARS-CoV-2 ORF8 protein and demonstrate that it induces rapidly generated but highly controllable endoplasmic reticulum stress-like responses and facilitates virus replication by triggering Calnexin in human but not mouse cells, providing an explanation for the superficially known in vivo virulence discrepancy of ORF8 between SARS-CoV-2-infected patients and mouse.


Asunto(s)
COVID-19 , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Humanos , Calnexina/genética , SARS-CoV-2/genética , Replicación Viral
10.
Emerg Microbes Infect ; 12(1): e2184177, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36877121

RESUMEN

Wild aquatic birds are the primary hosts of H13 avian influenza viruses (AIVs). Herein, we performed a genetic analysis of two H13 AIVs isolated from wild birds in China and evaluated their infection potential in poultry to further explore the potential for transmission from wild aquatic birds to poultry. Our results showed that the two strains belong to different groups, one strain (A/mallard/Dalian/DZ-137/2013; abbreviated as DZ137) belongs to Group I, whereas the other strain (A/Eurasian Curlew/Liaoning/ZH-385/2014; abbreviated as ZH385) belongs to Group III. In vitro experiments showed that both DZ137 and ZH385 can replicate efficiently in chicken embryo fibroblast cells. We found that these H13 AIVs can also efficiently replicate in mammalian cell lines, including human embryonic kidney cells and Madin-Darby canine kidney cells. In vivo experiments showed that DZ137 and ZH385 can infect 1-day-old specific pathogen-free (SPF) chickens, and that ZH385 has a higher replication ability in chickens than DZ137. Notably, only ZH385 can replicate efficiently in 10-day-old SPF chickens. However, neither DZ137 nor ZH385 can replicate well in turkeys and quails. Both DZ137 and ZH385 can replicate in 3-week-old mice. Serological surveillance of poultry showed a 4.6%-10.4% (15/328-34/328) antibody-positive rate against H13 AIVs in farm chickens. Our findings indicate that H13 AIVs have the replication ability in chickens and mice and may have a risk of crossing the host barrier from wild aquatic birds to poultry or mammals in the future.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Embrión de Pollo , Animales , Perros , Ratones , Humanos , Aves de Corral , Pollos , Animales Salvajes , Mamíferos , Filogenia
11.
Virol Sin ; 38(1): 119-127, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36450323

RESUMEN

Taurolidine (TRD), a derivative of taurine, has anti-bacterial and anti-tumor effects by chemically reacting with cell-walls, endotoxins and exotoxins to inhibit the adhesion of microorganisms. However, its application in antiviral therapy is seldom reported. Here, we reported that TRD significantly inhibited the replication of influenza virus H5N1 in MDCK cells with the half-maximal inhibitory concentration (EC50) of 34.45 â€‹µg/mL. Furthermore, the drug inhibited the amplification of the cytokine storm effect and improved the survival rate of mice lethal challenged with H5N1 (protection rate was 86%). Moreover, TRD attenuated virus-induced lung damage and reduced virus titers in mice lungs. Administration of TRD reduced the number of neutrophils and increased the number of lymphocytes in the blood of H5N1 virus-infected mice. Importantly, the drug regulated the NF-κB signaling pathway by inhibiting the separation of NF-κB and IκBa, thereby reducing the expression of inflammatory factors. In conclusion, our findings suggested that TRD could act as a potential anti-influenza drug candidate in further clinical studies.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Infecciones por Orthomyxoviridae , Animales , Ratones , FN-kappa B/metabolismo , Antivirales/farmacología , Antivirales/uso terapéutico , Infecciones por Orthomyxoviridae/prevención & control , Virus de la Influenza A/fisiología , Transducción de Señal , Taurina/farmacología , Taurina/uso terapéutico , Ratones Endogámicos BALB C , Replicación Viral
12.
Eur J Ophthalmol ; 32(6): 3321-3327, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35196147

RESUMEN

PURPOSE: To study the effects of a bandage contact lens immersed in 0.1% diclofenac on pain management for patients undergoing transepithelial photorefractive keratectomy (TPRK). METHODS: In a prospective, comparative, contralateral, randomized, double-masked study, we assessed a total of 51 patients. The eyes of each patient were randomly divided into two groups. After TPRK, a normal soft bandage contact lens was placed on one eye as the control group, and a bandage contact lens soaked in diclofenac was placed on the other eye as the experimental group. When the bandage contact lens was not removed, postoperative pain and other ocular discomforts were compared at 2, 18, and 24 h and 2, 3, 4, and 5 postoperative days. Patients were then examined after 1 month. Visual acuity and subepithelial haze were also evaluated. RESULTS: The mean pain score was 2.69 ± 1.96 in the control group, which was significantly higher than that in the experimental group, which received the diclofenac-soaked bandage contact lens at 2 postoperative hours. The statistical difference between the two groups' mean foreign body sensation at 2 postoperative hours was detected (p = 0.035). No differences were detected between the two groups' subepithelial haze scores or visual acuity. CONCLUSION: A bandage contact lens soaked in 0.1% diclofenac solution can be used as a potential drug-delivery system to relieve early postoperative pain and foreign body sensation after TPRK.


Asunto(s)
Cuerpos Extraños , Miopía , Queratectomía Fotorrefractiva , Vendajes , Diclofenaco/uso terapéutico , Cuerpos Extraños/cirugía , Humanos , Láseres de Excímeros/uso terapéutico , Miopía/cirugía , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/prevención & control , Dolor Postoperatorio/cirugía , Estudios Prospectivos , Sensación
13.
Transbound Emerg Dis ; 69(2): 669-684, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33566453

RESUMEN

H9N2 influenza virus has been reported worldwide for several decades, and it has evolved into multiple genotypes among domestic poultry. However, the study involving ecology and evolution of low pathogenic avian influenza virus H9N2 in wild birds in China is limited. Here, we carried out surveillance of avian influenza virus H9N2 in wild birds along with the East Asian-Australian migratory flyway in China in 2017. To estimate the prevalence of H9N2 avian virus in wild birds, information on exposure of wild bird populations to H9N2 viruses using serology, in addition to virology, would greatly improve monitoring capabilities. In this study, we also present serological data of H9N2 among wild birds in China during 2013-2016. We report the identification of poultry-derived H9N2 isolates from asymptomatic infected multispecies wild birds such as Common kestrel (Falco tinnunculus), Northern goshawk (Accipiter gentilis), Little owl (Athene noctua) and Ring-necked Pheasant (Phasianus colchicus) in North China in June 2017. Phylogenetic analysis demonstrated that Tianjin H9N2 isolates belong to the G81 and carry internal genes highly homologous to human H10N8 and H7N9. The isolates could directly infect mice without adaptation but were restricted to replicate in the respiratory system. Glycan-binding preference analyses suggested that the H9N2 isolates have acquired a binding affinity for the human-like receptor. Notably, results from transmission experiment in guinea pigs and ferrets demonstrated the wild birds-derived H9N2 influenza virus exhibits efficient transmission phenotypes in mammalian models via respiratory droplets. Our results indicate that the H9N2 AIVs continued to circulate extensively in wild bird populations and migratory birds play an important role in the spread and genetic diversification of H9N2 AIVs. The pandemic potential of H9N2 viruses demonstrated by aerosol transmission in mammalian models via respiratory droplets highlights the importance of monitoring influenza viruses in these hosts.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Enfermedades de los Roedores , Animales , Australia , Aves , China/epidemiología , Hurones , Cobayas , Humanos , Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/genética , Mamíferos , Ratones , Filogenia , Aves de Corral , Aerosoles y Gotitas Respiratorias
14.
Viruses ; 13(12)2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34960689

RESUMEN

H5N1 influenza virus is a threat to public health worldwide. The virus can cause severe morbidity and mortality in humans. We constructed an H5N1 influenza candidate virus vaccine from the A/chicken/Guizhou/1153/2016 strain that was recommended by the World Health Organization. In this study, we designed an H5N1 chimeric influenza A/B vaccine based on a cold-adapted (ca) influenza B virus B/Vienna/1/99 backbone. We modified the ectodomain of H5N1 hemagglutinin (HA) protein, while retaining the packaging signals of influenza B virus, and then rescued a chimeric cold-adapted H5N1 candidate influenza vaccine through a reverse genetic system. The chimeric H5N1 vaccine replicated well in eggs and the Madin-Darby Canine Kidney cells. It maintained a temperature-sensitive and cold-adapted phenotype. The H5N1 vaccine was attenuated in mice. Hemagglutination inhibition (HAI) antibodies, micro-neutralizing (MN) antibodies, and IgG antibodies were induced in immunized mice, and the mucosal IgA antibody responses were detected in their lung lavage fluids. The IFN-γ-secretion and IL-4-secretion by the mouse splenocytes were induced after stimulation with the specific H5N1 HA protein. The chimeric H5N1 candidate vaccine protected mice against lethal challenge with a wild-type highly pathogenic avian H5N1 influenza virus. The chimeric H5 candidate vaccine is thus a potentially safe, attenuated, and reassortment-incompetent vaccine with circulating A viruses.


Asunto(s)
Inmunogenicidad Vacunal , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza , Infecciones por Orthomyxoviridae/prevención & control , Eficacia de las Vacunas , Adaptación Fisiológica , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Frío , Perros , Femenino , Pruebas de Inhibición de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Inmunidad Celular , Inmunidad Mucosa , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/fisiología , Virus de la Influenza B/genética , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/efectos adversos , Vacunas contra la Influenza/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/inmunología , Proteínas Recombinantes , Vacunas Atenuadas/inmunología , Replicación Viral
15.
Virol J ; 18(1): 230, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34809668

RESUMEN

BACKGROUND: In 2011, a new influenza virus, named Influenza D Virus (IDV), was isolated from pigs, and then cattle, presenting influenza-like symptoms. IDV is one of the causative agents of Bovine Respiratory Disease (BRD), which causes high morbidity and mortality in feedlot cattle worldwide. To date, the molecular mechanisms of IDV pathogenicity are unknown. Recent IDV outbreaks in cattle, along with serological and genetic evidence of IDV infection in humans, have raised concerns regarding the zoonotic potential of this virus. Influenza virus polymerase is a determining factor of viral pathogenicity to mammals. METHODS: Here we take a prospective approach to this question by creating a random mutation library about PB2 subunit of the IDV viral polymerase to test which amino acid point mutations will increase viral polymerase activity, leading to increased pathogenicity of the virus. RESULTS: Our work shows some exact sites that could affect polymerase activities in influenza D viruses. For example, two single-site mutations, PB2-D533S and PB2-G603Y, can independently increase polymerase activity. The PB2-D533S mutation alone can increase the polymerase activity by 9.92 times, while the PB2-G603Y mutation increments the activity by 8.22 times. CONCLUSION: Taken together, our findings provide important insight into IDV replication fitness mediated by the PB2 protein, increasing our understanding of IDV replication and pathogenicity and facilitating future studies.


Asunto(s)
Infecciones por Orthomyxoviridae , Orthomyxoviridae , Thogotovirus , Aminoácidos/genética , Animales , Bovinos , Mutación , Porcinos , Thogotovirus/genética , Replicación Viral
16.
Viruses ; 13(11)2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34835084

RESUMEN

Avian H3N2 influenza virus follows cross-host transmission and has spread among dogs in Asia since 2005. After 2015-2016, a new H3N2 subtype canine influenza epidemic occurred in dogs in North America and Asia. The disease prevalence was assessed by virological and serological surveillance in dogs in China. Herein, five H3N2 canine influenza virus (CIV) strains were isolated from 1185 Chinese canine respiratory disease samples in 2017-2018; these strains were on the evolutionary branch of the North American CIVs after 2016 and genetically far from the classical canine H3N2 strain discovered in China before 2016. Serological surveillance showed an HI antibody positive rate of 6.68%. H3N2 was prevalent in the coastal areas and northeastern regions of China. In 2018, it became the primary epidemic strain in the country. The QK01 strain of H3N2 showed high efficiency in transmission among dogs through respiratory droplets. Nevertheless, the virus only replicated in the upper respiratory tract and exhibited low pathogenicity in mice. Furthermore, highly efficient transmission by direct contact other than respiratory droplet transmission was found in a guinea pig model. The low-level replication in avian species other than ducks could not facilitate contact and airborne transmission in chickens. The current results indicated that a novel H3N2 virus has become a predominant epidemic strain in dogs in China since 2016 and acquired highly efficient transmissibility but could not be replicated in avian species. Thus, further monitoring is required for designing optimal immunoprophylactic tools for dogs and estimating the zoonotic risk of CIV in China.


Asunto(s)
Enfermedades de los Perros/epidemiología , Subtipo H3N2 del Virus de la Influenza A/genética , Infecciones por Orthomyxoviridae/veterinaria , Aerosoles y Gotitas Respiratorias/virología , Animales , Pollos , China/epidemiología , Perros , Patos , Femenino , Cobayas , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Filogenia
17.
Viruses ; 13(7)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209472

RESUMEN

West Nile virus disease (WND) is an arthropod-borne zoonosis responsible for nonspecific fever or severe encephalitis. The pathogen is West Nile virus belonging to the genus Flavivirus, family Flaviviridae. Every year, thousands of cases were reported, which poses significant public health risk. Here, we constructed a West Nile virus chimera, ChiVax-WN01, by replacing the prMΔE gene of JEV SA14-14-2 with that of the West Nile virus NY99. The ChiVax-WN01 chimera showed clear, different characters compared with that of JEV SA14-14-2 and WNV NY99 strain. An animal study indicated that the ChiVax-WN01 chimera presented moderate safety and immunogenicity for 4-week female BALB/c mice.


Asunto(s)
Quimera , Virus de la Encefalitis Japonesa (Especie)/genética , Virus del Nilo Occidental/genética , Animales , Línea Celular , Cricetinae , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Femenino , Ratones , Ratones Endogámicos BALB C , Virulencia , Virus del Nilo Occidental/patogenicidad
19.
J Virol ; 95(18): e0060021, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34106002

RESUMEN

Coronaviruses are commonly characterized by a unique discontinuous RNA transcriptional synthesis strategy guided by transcription-regulating sequences (TRSs). However, the details of RNA synthesis in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have not been fully elucidated. Here, we present a time-scaled, gene-comparable transcriptome of SARS-CoV-2, demonstrating that ACGAAC functions as a core TRS guiding the discontinuous RNA synthesis of SARS-CoV-2 from a holistic perspective. During infection, viral transcription, rather than genome replication, dominates all viral RNA synthesis activities. The most highly expressed viral gene is the nucleocapsid gene, followed by ORF7 and ORF3 genes, while the envelope gene shows the lowest expression. Host transcription dysregulation keeps exacerbating after viral RNA synthesis reaches a maximum. The most enriched host pathways are metabolism related. Two of them (cholesterol and valine metabolism) affect viral replication in reverse. Furthermore, the activation of numerous cytokines emerges before large-scale viral RNA synthesis. IMPORTANCE SARS-CoV-2 is responsible for the current severe global health emergency that began at the end of 2019. Although the universal transcriptional strategies of coronaviruses are preliminarily understood, the details of RNA synthesis, especially the time-matched transcription level of each SARS-CoV-2 gene and the principles of subgenomic mRNA synthesis, are not clear. The coterminal subgenomic mRNAs of SARS-CoV-2 present obstacles in identifying the expression of most genes by PCR-based methods, which are exacerbated by the lack of related antibodies. Moreover, SARS-CoV-2-related metabolic imbalance and cytokine storm are receiving increasing attention from both clinical and mechanistic perspectives. Our transcriptomic research provides information on both viral RNA synthesis and host responses, in which the transcription-regulating sequences and transcription levels of viral genes are demonstrated, and the metabolic dysregulation and cytokine levels identified at the host cellular level support the development of novel medical treatment strategies.


Asunto(s)
COVID-19/genética , Células Epiteliales/metabolismo , Pulmón/metabolismo , ARN Mensajero/genética , SARS-CoV-2/aislamiento & purificación , Transcriptoma , Animales , COVID-19/metabolismo , COVID-19/virología , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/virología , Humanos , Pulmón/virología , ARN Mensajero/metabolismo , Células Vero , Replicación Viral
20.
Emerg Microbes Infect ; 10(1): 1038-1051, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33840358

RESUMEN

Influenza H3N8 viruses have been recovered frequently from wild bird species, including Anseriformes (primarily from migratory ducks) and Charadriiformes (primarily from shorebirds). However, little attention has been given to the transmission ability of H3N8 avian influenza viruses among mammals. Here, we study the potential human health threat and the molecular basis of mammalian transmissibility of H3N8 avian influenza viruses isolated from wild bird reservoirs. We classified eight H3N8 viruses into seven different genotypes based on genomic diversity. Six of eight H3N8 viruses isolated naturally from wild birds have acquired the ability to bind to the human-type receptor. However, the affinity for α-2,6-linked SAs was lower than that for α-2,3-linked SAs. Experiments on guinea pigs demonstrated that three viruses transmitted efficiently to direct-contact guinea pigs without prior adaptation. Notably, one virus transmitted efficiently via respiratory droplets in guinea pigs but not in ferrets. We further found that the PB1 S524G mutation conferred T222 virus airborne transmissibility between ferrets. We also determined that the 524G mutant increased viral pathogenicity slightly in mice compared with the WT (wild type). Based on these results, we elucidated the potential human health threat and molecular basis of mammalian transmissibility of H3N8 influenza viruses. We emphasized the need for continued surveillance of the H3N8 influenza viruses circulating in birds.


Asunto(s)
Subtipo H3N8 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/transmisión , Polimorfismo de Nucleótido Simple , Proteínas Virales/genética , Animales , Modelos Animales de Enfermedad , Perros , Femenino , Aptitud Genética , Genotipo , Cobayas , Humanos , Subtipo H3N8 del Virus de la Influenza A/genética , Células de Riñón Canino Madin Darby , Ratones , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...