Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 238
Filter
Add more filters

Publication year range
1.
Cell ; 184(17): 4380-4391.e14, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34147139

ABSTRACT

Despite the discovery of animal coronaviruses related to SARS-CoV-2, the evolutionary origins of this virus are elusive. We describe a meta-transcriptomic study of 411 bat samples collected from a small geographical region in Yunnan province, China, between May 2019 and November 2020. We identified 24 full-length coronavirus genomes, including four novel SARS-CoV-2-related and three SARS-CoV-related viruses. Rhinolophus pusillus virus RpYN06 was the closest relative of SARS-CoV-2 in most of the genome, although it possessed a more divergent spike gene. The other three SARS-CoV-2-related coronaviruses carried a genetically distinct spike gene that could weakly bind to the hACE2 receptor in vitro. Ecological modeling predicted the co-existence of up to 23 Rhinolophus bat species, with the largest contiguous hotspots extending from South Laos and Vietnam to southern China. Our study highlights the remarkable diversity of bat coronaviruses at the local scale, including close relatives of both SARS-CoV-2 and SARS-CoV.


Subject(s)
COVID-19/virology , Chiroptera/virology , Coronavirus/genetics , Evolution, Molecular , SARS-CoV-2/genetics , Amino Acid Sequence , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/metabolism , Animals , Asia, Southeastern , China , Coronavirus/classification , Coronavirus/isolation & purification , Ecological and Environmental Phenomena , Genome, Viral , Humans , Models, Molecular , Phylogeny , SARS-CoV-2/physiology , Sequence Alignment , Sequence Analysis, RNA , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Viral Zoonoses
2.
Cell ; 177(6): 1553-1565.e16, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31104841

ABSTRACT

Enterovirus B (EV-B), a major proportion of the genus Enterovirus in the family Picornaviridae, is the causative agent of severe human infectious diseases. Although cellular receptors for coxsackievirus B in EV-B have been identified, receptors mediating virus entry, especially the uncoating process of echovirus and other EV-B remain obscure. Here, we found that human neonatal Fc receptor (FcRn) is the uncoating receptor for major EV-B. FcRn binds to the virus particles in the "canyon" through its FCGRT subunit. By obtaining multiple cryo-electron microscopy structures at different stages of virus entry at atomic or near-atomic resolution, we deciphered the underlying mechanisms of enterovirus attachment and uncoating. These structures revealed that different from the attachment receptor CD55, binding of FcRn to the virions induces efficient release of "pocket factor" under acidic conditions and initiates the conformational changes in viral particle, providing a structural basis for understanding the mechanisms of enterovirus entry.


Subject(s)
Enterovirus B, Human/metabolism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/ultrastructure , Receptors, Fc/metabolism , Receptors, Fc/ultrastructure , Capsid/metabolism , Cryoelectron Microscopy , Enterovirus , Enterovirus B, Human/pathogenicity , Enterovirus Infections/metabolism , Histocompatibility Antigens Class I/physiology , Humans , Models, Molecular , Phylogeny , Receptors, Fc/physiology , Virion , Virus Internalization
3.
Immunity ; 53(3): 685-696.e3, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32783921

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic poses a current world-wide public health threat. However, little is known about its hallmarks compared to other infectious diseases. Here, we report the single-cell transcriptional landscape of longitudinally collected peripheral blood mononuclear cells (PBMCs) in both COVID-19- and influenza A virus (IAV)-infected patients. We observed increase of plasma cells in both COVID-19 and IAV patients and XIAP associated factor 1 (XAF1)-, tumor necrosis factor (TNF)-, and FAS-induced T cell apoptosis in COVID-19 patients. Further analyses revealed distinct signaling pathways activated in COVID-19 (STAT1 and IRF3) versus IAV (STAT3 and NFκB) patients and substantial differences in the expression of key factors. These factors include relatively increase of interleukin (IL)6R and IL6ST expression in COVID-19 patients but similarly increased IL-6 concentrations compared to IAV patients, supporting the clinical observations of increased proinflammatory cytokines in COVID-19 patients. Thus, we provide the landscape of PBMCs and unveil distinct immune response pathways in COVID-19 and IAV patients.


Subject(s)
Coronavirus Infections/immunology , Cytokines/immunology , Influenza, Human/immunology , Leukocytes, Mononuclear/immunology , Pneumonia, Viral/immunology , Signal Transduction/immunology , Betacoronavirus/immunology , COVID-19 , Humans , Influenza A Virus, H1N1 Subtype/immunology , Pandemics , SARS-CoV-2
4.
Nature ; 2023 Apr 05.
Article in English | MEDLINE | ID: mdl-37019149

ABSTRACT

SARS-CoV-2, the causative agent of COVID-19, emerged in December 2019. Its origins remain uncertain. It has been reported that a number of the early human cases had a history of contact with the Huanan Seafood Market. Here we present the results of surveillance for SARS-CoV-2 within the market. From January 1st 2020, after closure of the market, 923 samples were collected from the environment. From 18th January, 457 samples were collected from 18 species of animals, comprising of unsold contents of refrigerators and freezers, swabs from stray animals, and the contents of a fish tank. Using RT-qPCR, SARS-CoV-2 was detected in 73 environmental samples, but none of the animal samples. Three live viruses were successfully isolated. The viruses from the market shared nucleotide identity of 99.99% to 100% with the human isolate HCoV-19/Wuhan/IVDC-HB-01/2019. SARS-CoV-2 lineage A (8782T and 28144C) was found in an environmental sample. RNA-seq analysis of SARS-CoV-2 positive and negative environmental samples showed an abundance of different vertebrate genera at the market. In summary, this study provides information about the distribution and prevalence of SARS-CoV-2 in the Huanan Seafood Market during the early stages of the COVID-19 outbreak.

5.
PLoS Pathog ; 19(8): e1011577, 2023 08.
Article in English | MEDLINE | ID: mdl-37603540

ABSTRACT

Circular RNAs (circRNAs) are involved in various biological roles, including viral infection and antiviral immune responses. To identify influenza A virus (IAV) infection-related circRNAs, we compared the circRNA profiles of A549 cells upon IAV infection. We found that circVAMP3 is substantially upregulated after IAV infection or interferon (IFN) stimulation. Furthermore, IAV and IFN-ß induced the expression of QKI-5, which promoted the biogenesis of circVAMP3. Overexpression of circVAMP3 inhibited IAV replication, while circVAMP3 knockdown promoted viral replication, suggesting that circVAMP3 restricts IAV replication. We verified the effect of circVAMP3 on viral infection in mice and found that circVAMP3 restricted IAV replication and pathogenesis in vivo. We also found that circVAMP3 functions as a decoy to the viral proteins nucleoprotein (NP) and nonstructural protein 1 (NS1). Mechanistically, circVAMP3 interfered with viral ribonucleoprotein complex activity by reducing the interaction of NP with polymerase basic 1, polymerase basic 2, or vRNA and restored the activation of IFN-ß by alleviating the inhibitory effect of NS1 to RIG-I or TRIM25. Our study provides new insights into the roles of circRNAs, both in directly inhibiting virus replication and in restoring innate immunity against IAV infection.


Subject(s)
Influenza, Human , RNA, Circular , Vesicle-Associated Membrane Protein 3 , Animals , Humans , Mice , Influenza, Human/genetics , Interferons , Nucleoproteins , Nucleotidyltransferases , RNA, Circular/genetics , Vesicle-Associated Membrane Protein 3/genetics
6.
Mol Ther ; 32(5): 1510-1525, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38454605

ABSTRACT

The acute respiratory virus infection can induce uncontrolled inflammatory responses, such as cytokine storm and viral pneumonia, which are the major causes of death in clinical cases. Cyclophilin A (CypA) is mainly distributed in the cytoplasm of resting cells and released into the extracellular space in response to inflammatory stimuli. Extracellular CypA (eCypA) is upregulated and promotes inflammatory response in severe COVID-19 patients. However, how eCypA promotes virus-induced inflammatory response remains elusive. Here, we observe that eCypA is induced by influenza A and B viruses and SARS-CoV-2 in cells, mice, or patients. Anti-CypA mAb reduces pro-inflammatory cytokines production, leukocytes infiltration, and lung injury in virus-infected mice. Mechanistically, eCypA binding to integrin ß2 triggers integrin activation, thereby facilitating leukocyte trafficking and cytokines production via the focal adhesion kinase (FAK)/GTPase and FAK/ERK/P65 pathways, respectively. These functions are suppressed by the anti-CypA mAb that specifically blocks eCypA-integrin ß2 interaction. Overall, our findings reveal that eCypA-integrin ß2 signaling mediates virus-induced inflammatory response, indicating that eCypA is a potential target for antibody therapy against viral pneumonia.


Subject(s)
COVID-19 , Cyclophilin A , Cyclophilin A/metabolism , Animals , Humans , Mice , COVID-19/metabolism , COVID-19/virology , COVID-19/immunology , CD18 Antigens/metabolism , SARS-CoV-2 , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Pneumonia, Viral/metabolism , Pneumonia, Viral/immunology , Cytokines/metabolism , Antibodies, Monoclonal/pharmacology , Signal Transduction , Influenza A virus , Disease Models, Animal
7.
Proc Natl Acad Sci U S A ; 119(19): e2201288119, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35507870

ABSTRACT

African swine fever virus (ASFV) is the causative agent of African swine fever, a highly contagious and usually fatal disease in pigs. The pathogenesis of ASFV infection has not been clearly elucidated. Here, we used single-cell RNA-sequencing technology to survey the transcriptomic landscape of ASFV-infected primary porcine alveolar macrophages. The temporal dynamic analysis of viral genes revealed increased expression of viral transmembrane genes. Molecular characteristics in the ASFV-exposed cells exhibited the activation of antiviral signaling pathways with increased expression levels of interferon-stimulated genes and inflammatory- and cytokine-related genes. By comparing infected cells with unexposed cells, we showed that the unfolded protein response (UPR) pathway was activated in low viral load cells, while the expression level of UPR-related genes in high viral load cells was less than that in unexposed cells. Cells infected with various viral loads showed signature transcriptomic changes at the median progression of infection. Within the infected cells, differential expression analysis and coregulated virus­host analysis both demonstrated that ASFV promoted metabolic pathways but inhibited interferon and UPR signaling, implying the regulation pathway of viral replication in host cells. Furthermore, our results revealed that the cell apoptosis pathway was activated upon ASFV infection. Mechanistically, the production of tumor necrosis factor alpha (TNF-α) induced by ASFV infection is necessary for cell apoptosis, highlighting the importance of TNF-α in ASFV pathogenesis. Collectively, the data provide insights into the comprehensive host responses and complex virus­host interactions during ASFV infection, which may instruct future research on antiviral strategies.


Subject(s)
African Swine Fever Virus , African Swine Fever , African Swine Fever Virus/genetics , Animals , Antiviral Agents/metabolism , Gene Expression Profiling , Macrophages/metabolism , Swine , Virus Replication/physiology
8.
Brief Bioinform ; 23(2)2022 03 10.
Article in English | MEDLINE | ID: mdl-35062017

ABSTRACT

Since the start of the SARS-CoV-2 pandemic in late 2019, several variants of concern (VOC) have been reported to have increased transmissibility. In addition, despite the progress of vaccination against SARS-CoV-2 worldwide, all vaccines currently in used are known to protect only partially from infection and onward transmission. We combined phylogenetic analysis with Bayesian inference under an epidemiological model to infer the reproduction number (Rt) and also trace person-to-person transmission. We examined the impact of phylogenetic uncertainty and sampling bias on the estimation. Our result indicated that lineage B had a significantly higher transmissibility than lineage A and contributed to the global pandemic to a large extent. In addition, although the transmissibility of VOCs is higher than other exponentially growing lineages, this difference is not very high. The probability of detecting onward transmission from patients infected with SARS-CoV-2 VOCs who had received at least one dose of vaccine was approximate 1.06% (3/284), which was slightly lower but not statistically significantly different from a probability of 1.21% (10/828) for unvaccinated individuals. In addition to VOCs, exponentially growing lineages in each country should also be account for when tailoring prevention and control strategies. One dose of vaccination could not efficiently prevent the onward transmission of SARS-CoV-2 VOCs. Consequently, nonpharmaceutical interventions (such as wearing masks and social distancing) should still be implemented in each country during the vaccination period.


Subject(s)
COVID-19/transmission , COVID-19/virology , SARS-CoV-2/classification , SARS-CoV-2/genetics , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Vaccines , Evolution, Molecular , Genome, Viral , Global Health , Humans , Phylogeny , Public Health Surveillance , SARS-CoV-2/immunology , Vaccination
9.
Nucleic Acids Res ; 50(D1): D888-D897, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34634813

ABSTRACT

The genomic variations of SARS-CoV-2 continue to emerge and spread worldwide. Some mutant strains show increased transmissibility and virulence, which may cause reduced protection provided by vaccines. Thus, it is necessary to continuously monitor and analyze the genomic variations of SARS-COV-2 genomes. We established an evaluation and prewarning system, SARS-CoV-2 variations evaluation and prewarning system (VarEPS), including known and virtual mutations of SARS-CoV-2 genomes to achieve rapid evaluation of the risks posed by mutant strains. From the perspective of genomics and structural biology, the database comprehensively analyzes the effects of known variations and virtual variations on physicochemical properties, translation efficiency, secondary structure, and binding capacity of ACE2 and neutralizing antibodies. An AI-based algorithm was used to verify the effectiveness of these genomics and structural biology characteristic quantities for risk prediction. This classifier could be further used to group viral strains by their transmissibility and affinity to neutralizing antibodies. This unique resource makes it possible to quickly evaluate the variation risks of key sites, and guide the research and development of vaccines and drugs. The database is freely accessible at www.nmdc.cn/ncovn.


Subject(s)
COVID-19/virology , Databases, Factual , Mutation , SARS-CoV-2/genetics , Algorithms , Angiotensin-Converting Enzyme 2/metabolism , Antibodies, Neutralizing/metabolism , Artificial Intelligence , DNA Primers , Genome, Viral , Humans
10.
Emerg Infect Dis ; 29(6): 1244-1249, 2023 06.
Article in English | MEDLINE | ID: mdl-37209677

ABSTRACT

Two novel reassortant highly pathogenic avian influenza viruses (H5N1) clade 2.3.4.4b.2 were identified in dead migratory birds in China in November 2021. The viruses probably evolved among wild birds through different flyways connecting Europe and Asia. Their low antigenic reaction to vaccine antiserum indicates high risks to poultry and to public health.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus , Influenza in Birds , Animals , Influenza in Birds/epidemiology , Phylogeny , Birds , Animals, Wild , Poultry , China/epidemiology , Influenza A virus/genetics
11.
J Med Virol ; 95(3): e28683, 2023 03.
Article in English | MEDLINE | ID: mdl-36929727

ABSTRACT

An ongoing outbreak of monkeypox virus (MPXV) was first reported in the United Kingdom on 6 May 2022. As of 17 November, there had been a total of 80 221 confirmed MPXV cases in over 110 countries. Based on data reported between 6 May and 30 June 2022 in the United Kingdom, Spain, and Germany, we applied a deep learning approach using convolutional neural networks to evaluate the parameters of the 2022 MPXV outbreak. The basic reproduction number (R0 ) of MPXV was estimated to be 2.32 in the United Kingdom, which indicates the active diffusion of MPXV since the beginning of the outbreak. The data from Spain and Germany produced higher median R0 values of 2.42 and 2.88, respectively. Importantly, the estimated R0 of MPXV in the three countries tends to the previously calculated R0 of smallpox (3.50 to 6.00). Furthermore, the incubation (1/ε) and infectious (1/γ) period was predicted between 9 and 10 days and 4-5 days, respectively. The R0 value derived from MPXV is consistent with the significantly increasing number of cases, indicating the risk of a rapid spread of MPXV worldwide, which would provide important insights for the prevention and control of MPXV epidemic.


Subject(s)
Epidemics , Mpox (monkeypox) , Humans , Mpox (monkeypox)/epidemiology , Disease Outbreaks , Basic Reproduction Number , Germany/epidemiology , Monkeypox virus
12.
J Med Virol ; 95(7): e28948, 2023 07.
Article in English | MEDLINE | ID: mdl-37436839

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic posed great impacts on public health. To fight against the pandemic, robust immune responses induced by vaccination are indispensable. Previously, we developed a subunit vaccine adjuvanted by aluminum hydroxide, ZF2001, based on the dimeric tandem-repeat RBD immunogen, which has been approved for clinical use. This dimeric RBD design was also explored as an mRNA vaccine. Both showed potent immunogenicity. In this study, a DNA vaccine candidate encoding RBD-dimer was designed. The humoral and cellular immune responses induced by homologous and heterologous prime-boost approaches with DNA-RBD-dimer and ZF2001 were assessed in mice. Protection efficacy was studied by the SARS-CoV-2 challenge. We found that the DNA-RBD-dimer vaccine was robustly immunogenic. Priming with DNA-RBD-dimer followed by ZF2001 boosting induced higher levels of neutralizing antibodies than homologous vaccination with either DNA-RBD-dimer or ZF2001, elicited polyfunctional cellular immunity with a TH 1-biased polarization, and efficiently protected mice against SARS-CoV-2 infection in the lung. This study demonstrated the robust and protective immune responses induced by the DNA-RBD-dimer candidate and provided a heterologous prime-boost approach with DNA-RBD-dimer and ZF2001.


Subject(s)
COVID-19 , Vaccines, DNA , Viral Vaccines , Humans , Animals , Mice , COVID-19 Vaccines , COVID-19/prevention & control , SARS-CoV-2 , Vaccination , Antibodies, Neutralizing , Immunity, Cellular , Antibodies, Viral
13.
Proc Natl Acad Sci U S A ; 117(11): 5949-5954, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123088

ABSTRACT

The live poultry trade is thought to play an important role in the spread and maintenance of highly pathogenic avian influenza A viruses (HP AIVs) in Asia. Despite an abundance of small-scale observational studies, the role of the poultry trade in disseminating AIV over large geographic areas is still unclear, especially for developing countries with complex poultry production systems. Here we combine virus genomes and reconstructed poultry transportation data to measure and compare the spatial spread in China of three key subtypes of AIV: H5N1, H7N9, and H5N6. Although it is difficult to disentangle the contribution of confounding factors, such as bird migration and spatial distance, we find evidence that the dissemination of these subtypes among domestic poultry is geographically continuous and likely associated with the intensity of the live poultry trade in China. Using two independent data sources and network analysis methods, we report a regional-scale community structure in China that might explain the spread of AIV subtypes in the country. The identification of this structure has the potential to inform more targeted strategies for the prevention and control of AIV in China.


Subject(s)
Influenza in Birds/epidemiology , Influenza in Birds/transmission , Influenza in Birds/virology , Poultry/virology , Animals , China/epidemiology , Genome, Viral , Humans , Influenza A Virus, H5N1 Subtype , Influenza A Virus, H7N9 Subtype , Phylogeography , Transportation
14.
Proc Natl Acad Sci U S A ; 117(38): 23807-23814, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32873642

ABSTRACT

Avian-origin influenza viruses overcome the bottleneck of the interspecies barrier and infect humans through the evolution of variants toward more efficient replication in mammals. The dynamic adaptation of the genetic substitutions and the correlation with the virulence of avian-origin influenza virus in patients remain largely elusive. Here, based on the one-health approach, we retrieved the original virus-positive samples from patients with H7N9 and their surrounding poultry/environment. The specimens were directly deep sequenced, and the subsequent big data were integrated with the clinical manifestations. Unlike poultry/environment-derived samples with the consistent dominance of avian signature 627E of H7N9 polymerase basic protein 2 (PB2), patient specimens had diverse ratios of mammalian signature 627K, indicating the rapid dynamics of H7N9 adaptation in patients during the infection process. In contrast, both human- and poultry/environment-related viruses had constant dominance of avian signature PB2-701D. The intrahost dynamic adaptation was confirmed by the gradual replacement of 627E by 627K in H7N9 in the longitudinally collected specimens from one patient. These results suggest that host adaptation for better virus replication to new hosts, termed "genetic tuning," actually occurred in H7N9-infected patients in vivo. Notably, our findings also demonstrate the correlation between rapid host adaptation of H7N9 PB2-E627K and the fatal outcome and disease severity in humans. The feature of H7N9 genetic tuning in vivo and its correlation with the disease severity emphasize the importance of testing for the evolution of this avian-origin virus during the course of infection.


Subject(s)
Adaptation, Biological/genetics , Amino Acid Substitution/genetics , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza, Human/virology , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Humans , RNA, Viral/genetics , Sequence Analysis, RNA , Virus Replication/genetics
15.
Proc Natl Acad Sci U S A ; 117(29): 17204-17210, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32601207

ABSTRACT

Pigs are considered as important hosts or "mixing vessels" for the generation of pandemic influenza viruses. Systematic surveillance of influenza viruses in pigs is essential for early warning and preparedness for the next potential pandemic. Here, we report on an influenza virus surveillance of pigs from 2011 to 2018 in China, and identify a recently emerged genotype 4 (G4) reassortant Eurasian avian-like (EA) H1N1 virus, which bears 2009 pandemic (pdm/09) and triple-reassortant (TR)-derived internal genes and has been predominant in swine populations since 2016. Similar to pdm/09 virus, G4 viruses bind to human-type receptors, produce much higher progeny virus in human airway epithelial cells, and show efficient infectivity and aerosol transmission in ferrets. Moreover, low antigenic cross-reactivity of human influenza vaccine strains with G4 reassortant EA H1N1 virus indicates that preexisting population immunity does not provide protection against G4 viruses. Further serological surveillance among occupational exposure population showed that 10.4% (35/338) of swine workers were positive for G4 EA H1N1 virus, especially for participants 18 y to 35 y old, who had 20.5% (9/44) seropositive rates, indicating that the predominant G4 EA H1N1 virus has acquired increased human infectivity. Such infectivity greatly enhances the opportunity for virus adaptation in humans and raises concerns for the possible generation of pandemic viruses.


Subject(s)
Genes, Viral , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/epidemiology , Influenza, Human/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/virology , Swine Diseases/epidemiology , Swine Diseases/virology , Animals , China , Cross Reactions , Epithelial Cells/virology , Genetic Variation , Genotype , Humans , Influenza A Virus, H1N1 Subtype/classification , Influenza, Human/immunology , Influenza, Human/transmission , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/transmission , Pandemics , Phylogeny , Prevalence , Reassortant Viruses/genetics , Seroepidemiologic Studies , Swine
16.
Proc Natl Acad Sci U S A ; 117(42): 26151-26157, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32989148

ABSTRACT

Emerging evidence suggests a resurgence of COVID-19 in the coming years. It is thus critical to optimize emergency response planning from a broad, integrated perspective. We developed a mathematical model incorporating climate-driven variation in community transmissions and movement-modulated spatial diffusions of COVID-19 into various intervention scenarios. We find that an intensive 8-wk intervention targeting the reduction of local transmissibility and international travel is efficient and effective. Practically, we suggest a tiered implementation of this strategy where interventions are first implemented at locations in what we call the Global Intervention Hub, followed by timely interventions in secondary high-risk locations. We argue that thinking globally, categorizing locations in a hub-and-spoke intervention network, and acting locally, applying interventions at high-risk areas, is a functional strategy to avert the tremendous burden that would otherwise be placed on public health and society.


Subject(s)
Communicable Disease Control/methods , Communicable Diseases, Emerging/prevention & control , Coronavirus Infections/prevention & control , Disease Transmission, Infectious/prevention & control , Global Health/trends , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Betacoronavirus , COVID-19 , Climate , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/transmission , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Forecasting , Humans , International Cooperation , Models, Theoretical , Pneumonia, Viral/epidemiology , Pneumonia, Viral/transmission , SARS-CoV-2 , Travel
17.
J Virol ; 95(18): e0087821, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34190596

ABSTRACT

The influenza A virus genome is comprised of eight single-stranded negative-sense viral RNA (vRNA) segments. Each of the eight vRNA segments contains segment-specific nonconserved noncoding regions (NCRs) of similar sequence and length in different influenza A virus strains. However, in the subtype-determinant segments, encoding hemagglutinin (HA) and neuraminidase (NA), the segment-specific noncoding regions are subtype specific, varying significantly in sequence and length at both the 3' and 5' termini among different subtypes. The significance of these subtype-specific noncoding regions (ssNCR) in the influenza virus replication cycle is not fully understood. In this study, we show that truncations of the 3'-end H1-subtype-specific noncoding region (H1-ssNCR) resulted in recombinant viruses with decreased HA vRNA replication and attenuated growth phenotype, although the vRNA replication was not affected in single-template RNP reconstitution assays. The attenuated viruses were unstable, and point mutations at nucleotide position 76 or 56 in the adjacent coding region of HA vRNA were found after serial passage. The mutations restored the HA vRNA replication and reversed the attenuated virus growth phenotype. We propose that the terminal noncoding and adjacent coding regions act synergistically to ensure optimal levels of HA vRNA replication in a multisegment environment. These results provide novel insights into the role of the 3'-end nonconserved noncoding regions and adjacent coding regions on template preference in multiple-segmented negative-strand RNA viruses. IMPORTANCE While most influenza A virus vRNA segments contain segment-specific nonconserved noncoding regions of similar length and sequence, these regions vary considerably both in length and sequence in the segments encoding HA and NA, the two major antigenic determinants of influenza A viruses. In this study, we investigated the function of the 3'-end H1-ssNCR and observed a synergistic effect between the 3'-end H1-ssNCR nucleotides and adjacent coding nucleotide(s) of the HA segment on template preference in a multisegment environment. The results unravel an additional level of complexity in the regulation of RNA replication in multiple-segmented negative-strand RNA viruses.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A virus/growth & development , Influenza, Human/virology , Open Reading Frames/genetics , RNA, Viral/metabolism , Untranslated Regions/genetics , Viral Proteins/metabolism , Virus Replication , A549 Cells , Base Sequence , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza, Human/genetics , Influenza, Human/metabolism , RNA, Viral/genetics , Viral Proteins/genetics , Virus Assembly
18.
J Virol ; 95(11)2021 05 10.
Article in English | MEDLINE | ID: mdl-33731452

ABSTRACT

H9N2 Avian influenza virus (AIV) is regarded as a principal donor of viral genes through reassortment to co-circulating influenza viruses that can result in zoonotic reassortants. Whether H9N2 virus can maintain sustained evolutionary impact on such reassortants is unclear. Since 2013, avian H7N9 virus had caused five sequential human epidemics in China; the fifth wave in 2016-2017 was by far the largest but the mechanistic explanation behind the scale of infection is not clear. Here, we found that, just prior to the fifth H7N9 virus epidemic, H9N2 viruses had phylogenetically mutated into new sub-clades, changed antigenicity and increased its prevalence in chickens vaccinated with existing H9N2 vaccines. In turn, the new H9N2 virus sub-clades of PB2 and PA genes, housing mammalian adaptive mutations, were reassorted into co-circulating H7N9 virus to create a novel dominant H7N9 virus genotype that was responsible for the fifth H7N9 virus epidemic. H9N2-derived PB2 and PA genes in H7N9 virus conferred enhanced polymerase activity in human cells at 33°C and 37°C, and increased viral replication in the upper and lower respiratory tracts of infected mice which could account for the sharp increase in human cases of H7N9 virus infection in the 2016-2017 epidemic. The role of H9N2 virus in the continual mutation of H7N9 virus highlights the public health significance of H9N2 virus in the generation of variant reassortants of increasing zoonotic potential.IMPORTANCEAvian H9N2 influenza virus, although primarily restricted to chicken populations, is a major threat to human public health by acting as a donor of variant viral genes through reassortment to co-circulating influenza viruses. We established that the high prevalence of evolving H9N2 virus in vaccinated flocks played a key role, as donor of new sub-clade PB2 and PA genes in the generation of a dominant H7N9 virus genotype (G72) with enhanced infectivity in humans during the 2016-2017 N7N9 virus epidemic. Our findings emphasize that the ongoing evolution of prevalent H9N2 virus in chickens is an important source, via reassortment, of mammalian adaptive genes for other influenza virus subtypes. Thus, close monitoring of prevalence and variants of H9N2 virus in chicken flocks is necessary in the detection of zoonotic mutations.

20.
Pharmacol Res ; 185: 106509, 2022 11.
Article in English | MEDLINE | ID: mdl-36243330

ABSTRACT

Influenza A virus is globally widespread, causing a large number of infections and deaths due to acute lung injury (ALI) every year. The destruction and impairment of alveolar epithelial and microvascular endothelial cell barrier functions are the key inducers of ALI and acute respiratory distress syndrome caused by influenza virus infection. Although noncoding ribonucleic acids (ncRNAs) do not encode proteins in host cells, they possess the ability of protein regulation and signal transduction. Moreover, studies have shown that ncRNAs are significantly and differentially expressed following influenza virus infection, and these ncRNAs play vital roles in the pathogenesis of influenza virus infection. By analyzing the recently published literature, we found that ncRNA could regulate alveolar epithelial and microvascular endothelial cell barrier functions in different ways, which include influencing the innate and acquired immune responses of host cells, affecting apoptosis and autophagy, regulating tight and adherent junctions, etc. In the present paper, we reviewed the roles and regulatory mechanisms of these ncRNAs and discussed the effects of these ncRNAs on pulmonary epithelial and endothelial cell barriers. Further, by sorting and analyzing available research data, we proposed the possibility of applying these ncRNAs for treating ALI in influenza cases, thereby alleviating the permeability of pulmonary epithelial and endothelial cell barriers. Moreover, we discussed future research and development prospects. Our review suggests that targeted therapy and drug research based on ncRNAs would provide an important direction for the molecular therapy of influenza-induced ALI.


Subject(s)
Acute Lung Injury , Influenza A virus , Influenza, Human , Humans , Influenza, Human/complications , Influenza, Human/drug therapy , Lung , Acute Lung Injury/drug therapy , Acute Lung Injury/etiology , Endothelial Cells
SELECTION OF CITATIONS
SEARCH DETAIL