Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Emerg Microbes Infect ; 12(2): 2256416, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37672505

ABSTRACT

The emergence of novel betacoronaviruses has posed significant financial and human health burdens, necessitating the development of appropriate tools to combat future outbreaks. In this study, we have characterized a human cell line, IGROV-1, as a robust tool to detect, propagate, and titrate betacoronaviruses SARS-CoV-2 and HCoV-OC43. IGROV-1 cells can be used for serological assays, antiviral drug testing, and isolating SARS-CoV-2 variants from patient samples. Using time-course transcriptomics, we confirmed that IGROV-1 cells exhibit a robust innate immune response upon SARS-CoV-2 infection, recapitulating the response previously observed in primary human nasal epithelial cells. We performed genome-wide CRISPR knockout genetic screens in IGROV-1 cells and identified Aryl hydrocarbon receptor (AHR) as a critical host dependency factor for both SARS-CoV-2 and HCoV-OC43. Using DiMNF, a small molecule inhibitor of AHR, we observed that the drug selectively inhibits HCoV-OC43 infection but not SARS-CoV-2. Transcriptomic analysis in primary normal human bronchial epithelial cells revealed that DiMNF blocks HCoV-OC43 infection via basal activation of innate immune responses. Our findings highlight the potential of IGROV-1 cells as a valuable diagnostic and research tool to combat betacoronavirus diseases.


Subject(s)
COVID-19 , Coronavirus OC43, Human , Humans , Coronavirus OC43, Human/genetics , SARS-CoV-2 , Receptors, Aryl Hydrocarbon/genetics , Cell Line
2.
J Gen Virol ; 104(8)2023 08.
Article in English | MEDLINE | ID: mdl-37650875

ABSTRACT

Influenza A viruses of the H2N2 subtype sparked a pandemic in 1957 and circulated in humans until 1968. Because A/H2N2 viruses still circulate in wild birds worldwide and human population immunity is low, the transmissibility of six avian A/H2N2 viruses was investigated in the ferret model. None of the avian A/H2N2 viruses was transmitted between ferrets, suggesting that their pandemic risk may be low. The transmissibility, receptor binding preference and haemagglutinin (HA) stability of human A/H2N2 viruses were also investigated. Human A/H2N2 viruses from 1957 and 1958 bound to human-type α2,6-linked sialic acid receptors, but the 1958 virus had a more stable HA, indicating adaptation to replication and spread in the new host. This increased stability was caused by a previously unknown stability substitution G205S in the 1958 H2N2 HA, which became fixed in A/H2N2 viruses after 1958. Although individual substitutions were identified that affected the HA receptor binding and stability properties, they were not found to have a substantial effect on transmissibility of A/H2N2 viruses via the air in the ferret model. Our data demonstrate that A/H2N2 viruses continued to adapt during the first year of pandemic circulation in humans, similar to what was previously shown for the A/H1N1pdm09 virus.


Subject(s)
Influenza A Virus, H2N2 Subtype , Influenza A virus , Animals , Humans , Influenza A Virus, H2N2 Subtype/genetics , Ferrets , Pandemics
3.
One Health ; 16: 100529, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37363265

ABSTRACT

Orthonairovirus is a genus of viruses in the family Nairoviridae, order Bunyavirales, with a segmented circular RNA genome. They typically infect birds and mammals and are primarily transmitted by ectoparasites such as ticks. Four of nine Orthonairovirus genogroups can infect humans, with Crimean-Congo hemorrhagic fever virus infections displaying case fatality rates up to 40%. Here, we discover and describe a novel Orthonairovirus as Cencurut virus (CENV). CENV was detected in 34 of 37 Asian house shrews (Suncus murinus) sampled in Singapore and in a nymphal Amblyomma helvolum tick collected from an infected shrew. Pairwise comparison of CENV S, M, and L segments had 95.0 to 100% nucleotide and 97.5 to 100% amino acid homology within CENV genomes, suggesting a diverse viral population. Phylogenetic analysis of the individual gene segments showed that CENV is related to Erve, Lamgora, Lamusara, and Thiafora viruses, with only 49.0 to 58.2% nucleotide and 41.7 to 61.1% amino acid homology, which has previously been detected in other shrew species from France, Gabon, and Senegal respectively. The high detection frequency suggests that CENV is endemic among S. murinus populations in Singapore. The discovery of CENV, from a virus family with known zoonotic potential, underlines the importance of surveillance of synanthropic small mammals that are widely distributed across Southeast Asia.

4.
Microbiol Spectr ; 11(3): e0348322, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37199653

ABSTRACT

Bats are the reservoir for numerous human pathogens, including coronaviruses. Despite many coronaviruses having descended from bat ancestors, little is known about virus-host interactions and broader evolutionary history involving bats. Studies have largely focused on the zoonotic potential of coronaviruses with few infection experiments conducted in bat cells. To determine genetic changes derived from replication in bat cells and possibly identify potential novel evolutionary pathways for zoonotic virus emergence, we serially passaged six human 229E isolates in a newly established Rhinolophus lepidus (horseshoe bat) kidney cell line. Here, we observed extensive deletions within the spike and open reading frame 4 (ORF4) genes of five 229E viruses after passaging in bat cells. As a result, spike protein expression and infectivity of human cells was lost in 5 of 6 viruses, but the capability to infect bat cells was maintained. Only viruses that expressed the spike protein could be neutralized by 229E spike-specific antibodies in human cells, whereas there was no neutralizing effect on viruses that did not express the spike protein inoculated on bat cells. However, one isolate acquired an early stop codon, abrogating spike expression but maintaining infection in bat cells. After passaging this isolate in human cells, spike expression was restored due to acquisition of nucleotide insertions among virus subpopulations. Spike-independent infection of human coronavirus 229E may provide an alternative mechanism for viral maintenance in bats that does not rely on the compatibility of viral surface proteins and known cellular entry receptors. IMPORTANCE Many viruses, including coronaviruses, originated from bats. Yet, we know little about how these viruses switch between hosts and enter human populations. Coronaviruses have succeeded in establishing in humans at least five times, including endemic coronaviruses and the recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In an approach to identify requirements for host switches, we established a bat cell line and adapted human coronavirus 229E viruses by serial passage. The resulting viruses lost their spike protein but maintained the ability to infect bat cells, but not human cells. Maintenance of 229E viruses in bat cells appears to be independent of a canonical spike receptor match, which in turn might facilitate cross-species transmission in bats.


Subject(s)
COVID-19 , Chiroptera , Coronavirus 229E, Human , Animals , Humans , Phylogeny , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , SARS-CoV-2/metabolism
5.
PLoS Pathog ; 18(8): e1010763, 2022 08.
Article in English | MEDLINE | ID: mdl-35939522

ABSTRACT

Transmembrane Protein 41B (TMEM41B) and Vacuole Membrane Protein 1 (VMP1) are two ER-associated lipid scramblases that play a role in autophagosome formation and cellular lipid metabolism. TMEM41B is also a recently validated host factor required by flaviviruses and coronaviruses. However, the exact underlying mechanism of TMEM41B in promoting viral infections remains an open question. Here, we validated that both TMEM41B and VMP1 are essential host dependency factors for all four serotypes of dengue virus (DENV) and human coronavirus OC43 (HCoV-OC43), but not chikungunya virus (CHIKV). While HCoV-OC43 failed to replicate entirely in both TMEM41B- and VMP1-deficient cells, we detected diminished levels of DENV infections in these cell lines, which were accompanied by upregulation of the innate immune dsRNA sensors, RIG-I and MDA5. Nonetheless, this upregulation did not correspondingly induce the downstream effector TBK1 activation and Interferon-beta expression. Despite low levels of DENV replication, classical DENV replication organelles were undetectable in the infected TMEM41B-deficient cells, suggesting that the upregulation of the dsRNA sensors is likely a consequence of aberrant viral replication rather than a causal factor for reduced DENV infection. Intriguingly, we uncovered that the inhibitory effect of TMEM41B deficiency on DENV replication, but not HCoV-OC43, can be partially reversed using exogenous fatty acid supplements. In contrast, VMP1 deficiency cannot be rescued using the metabolite treatment. In line with the observed phenotypes, we found that both TMEM41B- and VMP1-deficient cells harbor higher levels of compromised mitochondria, especially in VMP1 deficiency which results in severe dysregulations of mitochondrial beta-oxidation. Using a metabolomic profiling approach, we revealed distinctive global dysregulations of the cellular metabolome, particularly lipidome, in TMEM41B- and VMP1-deficient cells. Our findings highlight a central role for TMEM41B and VMP1 in modulating multiple cellular pathways, including lipid mobilization, mitochondrial beta-oxidation, and global metabolic regulations, to facilitate the replication of flaviviruses and coronaviruses.


Subject(s)
Coronavirus Infections , Coronavirus , Dengue , Energy Metabolism , Humans , Lipids , Membrane Proteins/genetics , Virus Replication
6.
Microbiol Spectr ; 10(3): e0044922, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35638834

ABSTRACT

Bats are considered the natural reservoir of numerous emerging viruses such as severe acute respiratory syndrome coronaviruses (SARS-CoVs). There is a need for immortalized bat cell lines to culture and investigate the pathogenicity, replication kinetics, and evolution of emerging coronaviruses. We illustrate the susceptibility and permissiveness of a spontaneously immortalized kidney cell line (Rhileki) from Blyth's horseshoe bat (R. lepidus) to SARS-CoV-2 virus, including clinical isolates, suggesting a possible virus-host relationship. We were able to observe limited SARS-CoV-2 replication in Rhileki cells compared with simian VeroE6 cells. Slower viral replication in Rhileki cells was indicated by higher ct values (RT-PCR) at later time points of the viral culture and smaller foci (foci forming assay) compared with those of VeroE6 cells. With this study we demonstrate that SARS-CoV-2 replication is not restricted to R. sinicus and could include more Rhinolophus species. The establishment of a continuous Rhinolophus lepidus kidney cell line allows further characterization of SARS-CoV-2 replication in Rhinolophus bat cells, as well as isolation attempts of other bat-borne viruses. IMPORTANCE The current COVID-19 pandemic demonstrates the significance of bats as reservoirs for severe viral diseases. However, as bats are difficult to establish as animal models, bat cell lines can be an important proxy for the investigation of bat-virus interactions and the isolation of bat-borne viruses. This study demonstrates the susceptibility and permissiveness of a continuous kidney bat cell line to SARS-CoV-2. This does not implicate the bat species Rhinolophus lepidus, where these cells originate from, as a potential reservoir, but emphasizes the usefulness of this cell line for further characterization of SARS-CoV-2. This can lead to a better understanding of emerging viruses that could cause significant disease in humans and domestic animals.


Subject(s)
COVID-19 , Chiroptera , Animals , Humans , Kidney , Pandemics , Phylogeny , SARS-CoV-2
8.
Antiviral Res ; 193: 105138, 2021 09.
Article in English | MEDLINE | ID: mdl-34246735

ABSTRACT

The global spread of SARS-CoV-2 has made millions ill with COVID-19 and even more from the economic fallout of this pandemic. Our quest to test new therapeutics and vaccines require small animal models that replicate disease phenotypes seen in COVID-19 cases. Rodent models of SARS-CoV-2 infection thus far have shown mild to moderate pulmonary disease; mortality, if any, has been associated with prominent signs of central nervous system (CNS) infection and dysfunction. Here we describe the isolation of SARS-CoV-2 variants with propensity for either pulmonary or CNS infection. Using a wild-type SARS-CoV-2 isolated from a COVID-19 patient, we first found that infection was lethal in transgenic mice expressing the human angiotensin I-converting enzyme 2 (hACE2). Fortuitously, full genome sequencing of SARS-CoV-2 from the brain and lung of these animals showed genetic differences. Likewise, SARS-CoV-2 isolates from brains and lungs of these also showed differences in plaque morphology. Inoculation of these brain and lung SARS-CoV-2 isolates into new batch of hACE2 mice intra-nasally resulted in lethal CNS and pulmonary infection, respectively. Collectively, our study suggests that genetic variants of SARS-CoV-2 could be used to replicate specific features of COVID-19 for the testing of potential vaccines or therapeutics.


Subject(s)
COVID-19/pathology , Disease Models, Animal , Lung/pathology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Animals , Brain/pathology , Brain/virology , COVID-19/metabolism , COVID-19/mortality , COVID-19/virology , Female , Humans , Lung/virology , Mice , Mice, Transgenic , Peptidyl-Dipeptidase A/metabolism
9.
Heliyon ; 7(2): e06329, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33665466

ABSTRACT

Pathogens that cause upper respiratory infections are numerous and specific preventive and therapeutic strategies are scarce. In order to ascertain the etiological agents resulting in upper respiratory tract infections (URTI) in adults in Singapore, nasal swab samples were collected from 2057 patients presenting with fever at primary healthcare clinics in Singapore from December 2007 to February 2013. Samples were tested using the Luminex NxTAG Respiratory Pathogen Panel that includes 22 respiratory pathogen targets. Patient-reported symptoms and vital signs were recorded and full blood and differential counts taken. Pathogens were detected in the following order of frequency: influenza viruses, rhino-/enteroviruses, coronaviruses, parainfluenza viruses, pneumoviruses, adenovirus, bocavirus and C. pneumoniae. Fifteen virus species were detected as part of coinfections, in which rhinoviruses were the most commonly observed pathogen. Our results suggest that influenza viruses are the main etiological agents, but multiple other respiratory viruses contribute to the total burden of URTI in adults in Singapore.

10.
Cell Rep ; 34(6): 108728, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33516277

ABSTRACT

Virus-specific humoral and cellular immunity act synergistically to protect the host from viral infection. We interrogate the dynamic changes of virological and immunological parameters in 12 patients with symptomatic acute SARS-CoV-2 infection from disease onset to convalescence or death. We quantify SARS-CoV-2 viral RNA in the respiratory tract in parallel with antibodies and circulating T cells specific for various structural (nucleoprotein [NP], membrane [M], ORF3a, and spike) and non-structural (ORF7/8, NSP7, and NSP13) proteins. Although rapid induction and quantity of humoral responses associate with an increase in disease severity, early induction of interferon (IFN)-γ-secreting SARS-CoV-2-specific T cells is present in patients with mild disease and accelerated viral clearance. These findings provide support for the prognostic value of early functional SARS-CoV-2-specific T cells with important implications in vaccine design and immune monitoring.


Subject(s)
COVID-19 , Interferon-gamma/metabolism , T-Lymphocytes , Acute-Phase Reaction , Adult , Aged , Antibodies, Viral/immunology , Antigens, Viral/immunology , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Convalescence , Humans , Immunity, Cellular , Immunity, Humoral , Longitudinal Studies , Middle Aged , SARS-CoV-2/growth & development , SARS-CoV-2/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
Nature ; 584(7821): 457-462, 2020 08.
Article in English | MEDLINE | ID: mdl-32668444

ABSTRACT

Memory T cells induced by previous pathogens can shape susceptibility to, and the clinical severity of, subsequent infections1. Little is known about the presence in humans of pre-existing memory T cells that have the potential to recognize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we studied T cell responses against the structural (nucleocapsid (N) protein) and non-structural (NSP7 and NSP13 of ORF1) regions of SARS-CoV-2 in individuals convalescing from coronavirus disease 2019 (COVID-19) (n = 36). In all of these individuals, we found CD4 and CD8 T cells that recognized multiple regions of the N protein. Next, we showed that patients (n = 23) who recovered from SARS (the disease associated with SARS-CoV infection) possess long-lasting memory T cells that are reactive to the N protein of SARS-CoV 17 years after the outbreak of SARS in 2003; these T cells displayed robust cross-reactivity to the N protein of SARS-CoV-2. We also detected SARS-CoV-2-specific T cells in individuals with no history of SARS, COVID-19 or contact with individuals who had SARS and/or COVID-19 (n = 37). SARS-CoV-2-specific T cells in uninfected donors exhibited a different pattern of immunodominance, and frequently targeted NSP7 and NSP13 as well as the N protein. Epitope characterization of NSP7-specific T cells showed the recognition of protein fragments that are conserved among animal betacoronaviruses but have low homology to 'common cold' human-associated coronaviruses. Thus, infection with betacoronaviruses induces multi-specific and long-lasting T cell immunity against the structural N protein. Understanding how pre-existing N- and ORF1-specific T cells that are present in the general population affect the susceptibility to and pathogenesis of SARS-CoV-2 infection is important for the management of the current COVID-19 pandemic.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Severe Acute Respiratory Syndrome/immunology , T-Lymphocytes/immunology , Betacoronavirus/chemistry , COVID-19 , Case-Control Studies , Coronavirus Infections/virology , Coronavirus Nucleocapsid Proteins , Cross Reactions/immunology , Humans , Immunodominant Epitopes/immunology , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/immunology , Pandemics , Phosphoproteins , Pneumonia, Viral/virology , SARS-CoV-2
12.
mBio ; 11(4)2020 07 21.
Article in English | MEDLINE | ID: mdl-32694143

ABSTRACT

To date, limited genetic changes in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome have been described. Here, we report a 382-nucleotide (nt) deletion in SARS-CoV-2 that truncates open reading frame 7b (ORF7b) and ORF8, removing the ORF8 transcription regulatory sequence (TRS) and eliminating ORF8 transcription. The earliest 382-nt deletion variant was detected in Singapore on 29 January 2020, with the deletion viruses circulating in the country and accounting for 23.6% (45/191) of SARS-CoV-2 samples screened in this study. SARS-CoV-2 with the same deletion has since been detected in Taiwan, and other ORF7b/8 deletions of various lengths, ranging from 62 nt to 345 nt, have been observed in other geographic locations, including Australia, Bangladesh, and Spain. Mutations or deletions in ORF8 of SARS-CoV have been associated with reduced replicative fitness and virus attenuation. In contrast, the SARS-CoV-2 382-nt deletion viruses showed significantly higher replicative fitness in vitro than the wild type, while no difference was observed in patient viral load, indicating that the deletion variant viruses retained their replicative fitness. A robust antibody response to ORF8 has been observed in SARS-CoV-2 infection, suggesting that the emergence of ORF8 deletions may be due to immune-driven selection and that further deletion variants may emerge during the sustained transmission of SARS-CoV-2 in humans.IMPORTANCE During the SARS epidemic in 2003/2004, a number of deletions were observed in ORF8 of SARS-CoV, and eventually deletion variants became predominant, leading to the hypothesis that ORF8 was an evolutionary hot spot for adaptation of SARS-CoV to humans. However, due to the successful control of the SARS epidemic, the importance of these deletions for the epidemiological fitness of SARS-CoV in humans could not be established. The emergence of multiple SARS-CoV-2 strains with ORF8 deletions, combined with evidence of a robust immune response to ORF8, suggests that the lack of ORF8 may assist with host immune evasion. In addition to providing a key insight into the evolutionary behavior of SARS-CoV-2 as the virus adapts to its new human hosts, the emergence of ORF8 deletion variants may also impact vaccination strategies.


Subject(s)
Betacoronavirus/genetics , Genome, Viral , Open Reading Frames , Base Sequence , Betacoronavirus/immunology , Betacoronavirus/physiology , Humans , SARS-CoV-2 , Sequence Deletion , Virus Replication
13.
Emerg Microbes Infect ; 9(1): 900-902, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32380903

ABSTRACT

Despite initial findings indicating that SARS-CoV and SARS-CoV-2 are genetically related belonging to the same virus species and that the two viruses used the same entry receptor, angiotensin-converting enzyme 2 (ACE2), our data demonstrated that there is no detectable cross-neutralization by SARS patient sera against SARS-CoV-2. We also found that there are significant levels of neutralizing antibodies in recovered SARS patients 9-17 years after initial infection. These findings will be of significant use in guiding the development of serologic tests, formulating convalescent plasma therapy strategies, and assessing the longevity of protective immunity for SARS-related coronaviruses in general as well as vaccine efficacy.


Subject(s)
Antibodies, Viral/blood , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Betacoronavirus/immunology , Broadly Neutralizing Antibodies/immunology , COVID-19 , Coronavirus Infections/therapy , Humans , Immunization, Passive/standards , Pandemics , SARS-CoV-2 , Time Factors , Viral Vaccines/standards , COVID-19 Serotherapy
14.
Appl Biosaf ; 25(3): 169-173, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-36035761

ABSTRACT

Introduction: An infectious disease outbreak like the current COVID-19 pandemic can lead to particularly high infection rates in areas where diagnostic laboratory support is limited. The deployment of mobile laboratories can help to detect pathogens, monitor the presence in a population, and inform public health authorities to take measures aimed at reducing pathogen spread. Materials and Methods: Available layouts and operational descriptions of mobile laboratories were analyzed for their suitability for the envisioned purpose and to assure high standards of biosafety and biosecurity. Recent media coverage on creative solutions for the diagnostics of SARS-CoV-2 (drive-through test centers, self-swab, inverse gloveboxes to protect health care workers) from various countries were considered. Results: A minimalistic and optimized design to construct a multifunctional laboratory on the chassis of a regular-sized box truck is proposed and can serve as a blueprint to rapidly develop additional diagnostic capacities. Discussion: For acute health threats including the current COVID-19 outbreak, rapid diagnosis of infection is key to recommend measures aimed at preventing the spread of the pathogen. Laboratory layouts that are similar to the one proposed here are used in stationary setups, and mobile laboratories have been built on varying platforms (trailers, shipping containers, etc).

15.
Influenza Other Respir Viruses ; 14(1): 67-71, 2020 01.
Article in English | MEDLINE | ID: mdl-31568667

ABSTRACT

To understand the genetic diversity and patterns of circulation of rhinoviruses (RV) and enteroviruses (EV) in Singapore, we retrospectively screened 2950 nasal swab samples collected from adults presenting to primary care services with signs of febrile illness in Singapore during 2007-2013 using sequencing and phylogenetic methods. Through sequencing and phylogenetic analysis, our results show the year-round circulation of the three rhinovirus species, A, B, and C. A diverse set of RV/EV serotypes were detected in Singapore with a predominance of RV-A in all years, whereas serotypes EV-C A21 and EV-D68 were only sporadically detected. This study highlights the previously unrecognized diversity and burden in the adult population in Singapore.


Subject(s)
Enterovirus Infections/virology , Enterovirus/genetics , Fever/virology , Genetic Variation , Picornaviridae Infections/virology , Rhinovirus/genetics , Adolescent , Adult , Aged , Enterovirus/classification , Enterovirus/isolation & purification , Enterovirus Infections/epidemiology , Female , Humans , Male , Middle Aged , Phylogeny , Picornaviridae Infections/epidemiology , Retrospective Studies , Rhinovirus/classification , Rhinovirus/isolation & purification , Seasons , Singapore/epidemiology , Young Adult
16.
Proc Natl Acad Sci U S A ; 117(1): 619-628, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31843889

ABSTRACT

Influenza B viruses have circulated in humans for over 80 y, causing a significant disease burden. Two antigenically distinct lineages ("B/Victoria/2/87-like" and "B/Yamagata/16/88-like," termed Victoria and Yamagata) emerged in the 1970s and have cocirculated since 2001. Since 2015 both lineages have shown unusually high levels of epidemic activity, the reasons for which are unclear. By analyzing over 12,000 influenza B virus genomes, we describe the processes enabling the long-term success and recent resurgence of epidemics due to influenza B virus. We show that following prolonged diversification, both lineages underwent selective sweeps across the genome and have subsequently taken alternate evolutionary trajectories to exhibit epidemic dominance, with no reassortment between lineages. Hemagglutinin deletion variants emerged concomitantly in multiple Victoria virus clades and persisted through epistatic mutations and interclade reassortment-a phenomenon previously only observed in the 1970s when Victoria and Yamagata lineages emerged. For Yamagata viruses, antigenic drift of neuraminidase was a major driver of epidemic activity, indicating that neuraminidase-based vaccines and cross-reactivity assays should be employed to monitor and develop robust protection against influenza B morbidity and mortality. Overall, we show that long-term diversification and infrequent selective sweeps, coupled with the reemergence of hemagglutinin deletion variants and antigenic drift of neuraminidase, are factors that contributed to successful circulation of diverse influenza B clades. Further divergence of hemagglutinin variants with poor cross-reactivity could potentially lead to circulation of 3 or more distinct influenza B viruses, further complicating influenza vaccine formulation and highlighting the urgent need for universal influenza vaccines.


Subject(s)
Communicable Diseases, Emerging/virology , Epidemics/prevention & control , Evolution, Molecular , Influenza B virus/genetics , Influenza Vaccines/therapeutic use , Influenza, Human/virology , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/immunology , Communicable Diseases, Emerging/prevention & control , Genetic Variation , Genome, Viral/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza B virus/immunology , Influenza B virus/pathogenicity , Influenza, Human/epidemiology , Influenza, Human/immunology , Influenza, Human/prevention & control , Neuraminidase/genetics , Neuraminidase/immunology , Selection, Genetic/immunology
17.
Br Med Bull ; 132(1): 81-95, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31848585

ABSTRACT

BACKGROUND: Human infections with avian influenza viruses (AIV) represent a persistent public health threat. The principal risk factor governing human infection with AIV is from direct contact with infected poultry and is primarily observed in Asia and Egypt where live-bird markets are common. AREAS OF AGREEMENT: Changing patterns of virus transmission and a lack of obvious disease manifestations in avian species hampers early detection and efficient control of potentially zoonotic AIV. AREAS OF CONTROVERSY: Despite extensive studies on biological and environmental risk factors, the exact conditions required for cross-species transmission from avian species to humans remain largely unknown. GROWING POINTS: The development of a universal ('across-subtype') influenza vaccine and effective antiviral therapeutics are a priority. AREAS TIMELY FOR DEVELOPING RESEARCH: Sustained virus surveillance and collection of ecological and physiological parameters from birds in different environments is required to better understand influenza virus ecology and identify risk factors for human infection.


Subject(s)
Influenza in Birds/epidemiology , Influenza, Human/epidemiology , Animals , Antiviral Agents/therapeutic use , Birds , Disease Outbreaks , Disease Susceptibility , Humans , Influenza A virus/classification , Influenza Vaccines , Influenza in Birds/therapy , Influenza in Birds/transmission , Influenza, Human/therapy , Influenza, Human/transmission , Risk Factors , Zoonoses/epidemiology , Zoonoses/therapy , Zoonoses/transmission
18.
PLoS Negl Trop Dis ; 13(10): e0007733, 2019 10.
Article in English | MEDLINE | ID: mdl-31671094

ABSTRACT

Bats are reservoirs for several zoonotic pathogens, including filoviruses. Recent work highlights the diversity of bat borne filoviruses in Asia. High risk activities at the bat-human interface pose the threat of zoonotic virus transmission. We present evidence for prior exposure of bat harvesters and two resident fruit bat species to filovirus surface glycoproteins by screening sera in a multiplexed serological assay. Antibodies reactive to two antigenically distinct filoviruses were detected in human sera and to three individual filoviruses in bats in remote Northeast India. Sera obtained from Eonycteris spelaea bats showed similar patterns of cross-reactivity as human samples, suggesting them as the species responsible for the spillover. In contrast, sera from Rousettus leschenaultii bats reacted to two different virus glycoproteins. Our results indicate circulation of several filoviruses in bats and the possibility for filovirus transmission from bats to humans.


Subject(s)
Antibodies, Viral/blood , Chiroptera/immunology , Chiroptera/virology , Disease Reservoirs/virology , Filoviridae Infections/epidemiology , Filoviridae Infections/veterinary , Filoviridae/immunology , Adolescent , Adult , Animals , Chiroptera/blood , Ebolavirus/immunology , Filoviridae/classification , Filoviridae/isolation & purification , Filoviridae Infections/immunology , Filoviridae Infections/virology , Geographic Mapping , Glycoproteins/immunology , Humans , India/epidemiology , Membrane Glycoproteins , Middle Aged , Phylogeny , Seroepidemiologic Studies , Young Adult
19.
Sci Rep ; 8(1): 6833, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717150

ABSTRACT

HPIVs are serologically and genetically grouped into four species that account for up to 10% of all hospitalizations due to acute respiratory infection in children under the age of five. Genetic and epidemiological data for the four HPIVs derived from two pediatric cohorts in Viet Nam are presented. Respiratory samples were screened for HPIV1-4 by real-time PCR. Demographic and clinical data of patients infected with different HPIV were compared. We used a hemi-nested PCR approach to generate viral genome sequences from HPIV-positive samples and conducted a comprehensive phylogenetic analysis. In total, 170 samples tested positive for HPIV. HPIV3 was most commonly detected in our cohort and 80 co-detections of HPIV with other respiratory viruses were found. Phylogenetic analyses suggest local endemic circulation as well as punctuated introductions of new HPIV lineages. Viral gene flow analysis revealed that Viet Nam is a net importer of viral genetic diversity. Epidemiological analyses imply similar disease severity for all HPIV species. HPIV sequences from Viet Nam formed local clusters and were interspersed with sequences from diverse geographic regions. Combined, this new knowledge will help to investigate global HPIV circulation patterns in more detail and ultimately define more suitable vaccine strains.


Subject(s)
Molecular Epidemiology/methods , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/genetics , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 4, Human/genetics , Respiratory Tract Infections/epidemiology , Respirovirus Infections/epidemiology , Acute Disease , Adolescent , Chi-Square Distribution , Child , Child, Preschool , Female , Follow-Up Studies , Genetic Variation , Genome, Viral , Humans , Infant , Infant, Newborn , Male , Phylogeny , Real-Time Polymerase Chain Reaction , Severity of Illness Index , Statistics, Nonparametric , Vietnam/epidemiology , Whole Genome Sequencing
20.
Emerg Infect Dis ; 24(1): 114-117, 2018 01.
Article in English | MEDLINE | ID: mdl-29260678

ABSTRACT

To determine whether fruit bats in Singapore have been exposed to filoviruses, we screened 409 serum samples from bats of 3 species by using a multiplex assay that detects antibodies against filoviruses. Positive samples reacted with glycoproteins from Bundibugyo, Ebola, and Sudan viruses, indicating filovirus circulation among bats in Southeast Asia.


Subject(s)
Chiroptera/blood , Chiroptera/virology , Ebolavirus , Marburgvirus , Viral Envelope Proteins/blood , Animals , Glycoproteins/blood , Glycoproteins/genetics , Glycoproteins/isolation & purification , Seroepidemiologic Studies , Singapore/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...