Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.352
Filter
1.
Aerosp Med Hum Perform ; 95(9): 709-715, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39169493

ABSTRACT

INTRODUCTION: The European Air Transport Command (EATC) is a seven-nation integrated command. One of its core capabilities is strategic aeromedical evacuation (AE). During the global COVID-19 pandemic and Ukrainian crisis, EATC proved that acting in concert is a valuable, effective, and reliable option.METHODS: By pooling and sharing aircraft and personnel, EATC has privileged access to a diverse fleet and pool of experts. Cooperation is based on a common set of rules and regulations, which ensures that EATC can address any problem with expertise.RESULTS: During the COVID-19 pandemic, 1060 COVID-19-positive patients were transported in 198 missions, with neither death nor disease transmission reported during those strategic AE flights. EATC transferred 986 military cases, mostly routine priority (91.4%); the other 74 cases were civilians, who were transported in 17 missions, with 81.1% categorized as urgent. During the Ukrainian crisis, 251 patients were transported, 112 military and 139 civilians, including 30 children. Among the recorded injuries were cerebrocranial, abdominal, and chest injuries, as well as fractures (180) and amputations (48) of the extremities.DISCUSSION: EATC is recognized as a center of expertise within the AE community, where interoperability and harmonization of concepts are key to safety and success. Cross-national missions, where a patient is evacuated by an aircraft and medical crew provided by another nation, offer maximum flexibility. Complex situations, such as the COVID-19 pandemic and the Ukrainian crisis, have shown that multinational cooperation is not only achievable but also provides robust, effective, and reliable solutions for AE in particular.Fiorini A, Vermeltfoort R, Dulaurent E, Hove MG, Borsch M. Cross-national strategic aeromedical evacuation at the European Air Transport Command. Aerosp Med Hum Perform. 2024; 95(9):709-715.


Subject(s)
Aerospace Medicine , Air Ambulances , COVID-19 , Military Personnel , Pandemics , Humans , COVID-19/epidemiology , COVID-19/transmission , COVID-19/prevention & control , Air Ambulances/organization & administration , Europe/epidemiology , SARS-CoV-2 , Ukraine/epidemiology , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/prevention & control , Pneumonia, Viral/epidemiology , Pneumonia, Viral/transmission , Pneumonia, Viral/prevention & control , Transportation of Patients/organization & administration , Aircraft
2.
Math Biosci Eng ; 21(7): 6425-6470, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39176403

ABSTRACT

A new mathematical model for the transmission dynamics and control of the Middle Eastern respiratory syndrome (MERS), a respiratory virus caused by MERS-CoV coronavirus (and primarily spread to humans by dromedary camels) that first emerged out of the Kingdom of Saudi Arabia (KSA) in 2012, was designed and used to study the transmission dynamics of the disease in a human-camel population within the KSA. Rigorous analysis of the model, which was fitted and cross-validated using the observed MERS-CoV data for the KSA, showed that its disease-free equilibrium was locally asymptotically stable whenever its reproduction number (denoted by $ {\mathbb R}_{0M} $) was less than unity. Using the fixed and estimated parameters of the model, the value of $ {\mathbb R}_{0M} $ for the KSA was estimated to be 0.84, suggesting that the prospects for MERS-CoV elimination are highly promising. The model was extended to allow for the assessment of public health intervention strategies, notably the potential use of vaccines for both humans and camels and the use of face masks by humans in public or when in close proximity with camels. Simulations of the extended model showed that the use of the face mask by humans who come in close proximity with camels, as a sole public health intervention strategy, significantly reduced human-to-camel and camel-to-human transmission of the disease, and this reduction depends on the efficacy and coverage of the mask type used in the community. For instance, if surgical masks are prioritized, the disease can be eliminated in both the human and camel population if at least 45% of individuals who have close contact with camels wear them consistently. The simulations further showed that while vaccinating humans as a sole intervention strategy only had marginal impact in reducing the disease burden in the human population, an intervention strategy based on vaccinating camels only resulted in a significant reduction in the disease burden in camels (and, consequently, in humans as well). Thus, this study suggests that attention should be focused on effectively combating the disease in the camel population, rather than in the human population. Furthermore, the extended model was used to simulate a hybrid strategy, which combined vaccination of both humans and camels as well as the use of face masks by humans. This simulation showed a marked reduction of the disease burden in both humans and camels, with an increasing effectiveness level of this intervention, in comparison to the baseline scenario or any of the aforementioned sole vaccination scenarios. In summary, this study showed that the prospect of the elimination of MERS-CoV-2 in the Kingdom of Saudi Arabia is promising using pharmaceutical (vaccination) and nonpharmaceutical (mask) intervention strategies, implemented in isolation or (preferably) in combination, that are focused on reducing the disease burden in the camel population.


Subject(s)
Camelus , Coronavirus Infections , Middle East Respiratory Syndrome Coronavirus , Camelus/virology , Saudi Arabia/epidemiology , Animals , Humans , Coronavirus Infections/epidemiology , Coronavirus Infections/prevention & control , Coronavirus Infections/transmission , Basic Reproduction Number/statistics & numerical data , Computer Simulation , Viral Vaccines , Models, Biological , Masks , Models, Theoretical
3.
Immun Inflamm Dis ; 12(7): e1342, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39023424

ABSTRACT

BACKGROUND: Epidemiological data on seasonal coronaviruses (sCoVs) may provide insight on transmission patterns and demographic factors that favor coronaviruses (CoVs) with greater disease severity. This study describes the incidence of CoVs in several high-risk groups in Ottawa, Canada, from October 2020 to March 2022. METHODS: Serological assays quantified IgG and IgM antibodies to SARS-CoV-2, HCoV-OC43, HCoV-NL63, HCoV-HKU1, and HCoV-229E. Incident infections were compared between four population groups: individuals exposed to children, transit users, immunocompromised, and controls. Associations between antibody prevalence indicative of natural infection and demographic variables were assessed using regression analyses. RESULTS: Transit users and those exposed to children were at no greater risk of infection compared to the control group. Fewer infections were detected in the immunocompromised group (p = .03). SARS-CoV-2 seroprevalence was greater in individuals with low income and within ethnic minorities. CONCLUSIONS: Our findings suggest that nonpharmaceutical interventions intended to reduce SAR-CoV-2 transmission protected populations at high risk of exposure. The re-emergence of sCoVs and other common respiratory viruses alongside SARS-CoV-2 may alter infection patterns and increase the risk in vulnerable populations.


Subject(s)
COVID-19 , SARS-CoV-2 , Seasons , Humans , COVID-19/epidemiology , COVID-19/transmission , COVID-19/immunology , Incidence , Male , Female , SARS-CoV-2/immunology , Child , Adult , Middle Aged , Adolescent , Antibodies, Viral/blood , Child, Preschool , Seroepidemiologic Studies , Young Adult , Aged , Risk Factors , Canada/epidemiology , Infant , Pandemics , Immunoglobulin G/blood , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/immunology , Immunoglobulin M/blood
4.
J Zhejiang Univ Sci B ; 25(7): 628-632, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39011682

ABSTRACT

Airborne transmission is among the most frequent types of nosocomial infection. Recent years have witnessed frequent outbreaks of airborne diseases, such as severe acute respiratory syndrome (SARS) in 2002, Middle East respiratory syndrome (MERS) in 2012, and coronavirus disease 2019 (COVID-19), with the latter being on the rampage since the end of 2019 and bringing the effect of aerosols on health back to the fore (Gralton et al., 2011; Wang et al., 2021). An increasing number of studies have shown that certain highly transmissible pathogens can maintain long-term stability and efficiently spread through aerosols (Leung, 2021; Lv et al., 2021). As reported previously, influenza viruses that can spread efficiently through aerosols remain stable for a longer period compared to those that cannot. The World Health Organization (WHO) has stated that aerosol-generating procedures (AGPs) play an important role in aerosol transmission in hospitals (Calderwood et al., 2021). AGPs, referring to medical procedures that produce aerosols, including dental procedures, endotracheal intubation, sputum aspiration, and laparoscopic surgeries, have been reported to be significantly associated with an increased risk of nosocomial infection among medical personnel (Hamilton, 2021).


Subject(s)
Aerosols , COVID-19 , Cross Infection , Endoscopes , SARS-CoV-2 , Humans , Cross Infection/transmission , Cross Infection/prevention & control , COVID-19/transmission , SARS-CoV-2/isolation & purification , Pandemics , Coronavirus Infections/transmission , Pneumonia, Viral/transmission , Disinfection/methods , Betacoronavirus , Air Microbiology
5.
PLoS Pathog ; 20(7): e1012039, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38950065

ABSTRACT

The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) not only caused the COVID-19 pandemic but also had a major impact on farmed mink production in several European countries. In Denmark, the entire population of farmed mink (over 15 million animals) was culled in late 2020. During the period of June to November 2020, mink on 290 farms (out of about 1100 in the country) were shown to be infected with SARS-CoV-2. Genome sequencing identified changes in the virus within the mink and it is estimated that about 4000 people in Denmark became infected with these mink virus variants. However, the routes of transmission of the virus to, and from, the mink have been unclear. Phylogenetic analysis revealed the generation of multiple clusters of the virus within the mink. Detailed analysis of changes in the virus during replication in mink and, in parallel, in the human population in Denmark, during the same time period, has been performed here. The majority of cases in mink involved variants with the Y453F substitution and the H69/V70 deletion within the Spike (S) protein; these changes emerged early in the outbreak. However, further introductions of the virus, by variants lacking these changes, from the human population into mink also occurred. Based on phylogenetic analysis of viral genome data, we estimate, using a conservative approach, that about 17 separate examples of mink to human transmission occurred in Denmark but up to 59 such events (90% credible interval: (39-77)) were identified using parsimony to count cross-species jumps on transmission trees inferred using Bayesian methods. Using the latter approach, 136 jumps (90% credible interval: (117-164)) from humans to mink were found, which may underlie the farm-to-farm spread. Thus, transmission of SARS-CoV-2 from humans to mink, mink to mink, from mink to humans and between humans were all observed.


Subject(s)
COVID-19 , Mink , Phylogeny , SARS-CoV-2 , Mink/virology , COVID-19/transmission , COVID-19/virology , COVID-19/epidemiology , COVID-19/veterinary , SARS-CoV-2/genetics , Animals , Denmark/epidemiology , Humans , Pandemics , Farms , Betacoronavirus/genetics , Betacoronavirus/classification , Genome, Viral , Coronavirus Infections/veterinary , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Coronavirus Infections/transmission , Spike Glycoprotein, Coronavirus/genetics
6.
BMJ ; 386: e078918, 2024 07 24.
Article in English | MEDLINE | ID: mdl-39048132

ABSTRACT

OBJECTIVE: To evaluate the personal protective effects of wearing versus not wearing surgical face masks in public spaces on self-reported respiratory symptoms over a 14 day period. DESIGN: Pragmatic randomised superiority trial. SETTING: Norway. PARTICIPANTS: 4647 adults aged ≥18 years: 2371 were assigned to the intervention arm and 2276 to the control arm. INTERVENTIONS: Participants in the intervention arm were assigned to wear a surgical face mask in public spaces (eg, shopping centres, streets, public transport) over a 14 day period (mask wearing at home or work was not mentioned). Participants in the control arm were assigned to not wear a surgical face mask in public places. MAIN OUTCOME MEASURES: The primary outcome was self-reported respiratory symptoms consistent with a respiratory infection. Secondary outcomes included self-reported and registered covid-19 infection. RESULTS: Between 10 February 2023 and 27 April 2023, 4647 participants were randomised of whom 4575 (2788 women (60.9%); mean age 51.0 (standard deviation 15.0) years) were included in the intention-to-treat analysis: 2313 (50.6%) in the intervention arm and 2262 (49.4%) in the control arm. 163 events (8.9%) of self-reported symptoms consistent with respiratory infection were reported in the intervention arm and 239 (12.2%) in the control arm. The marginal odds ratio was 0.71 (95% confidence interval (CI) 0.58 to 0.87; P=0.001) favouring the face mask intervention. The absolute risk difference was -3.2% (95% CI -5.2% to -1.3%; P<0.001). No statistically significant effect was found on self- reported (marginal odds ratio 1.07, 95% CI 0.58 to 1.98; P=0.82) or registered covid-19 infection (effect estimate and 95% CI not estimable owing to lack of events in the intervention arm). CONCLUSION: Wearing a surgical face mask in public spaces over 14 days reduces the risk of self-reported symptoms consistent with a respiratory infection, compared with not wearing a surgical face mask. TRIAL REGISTRATION: ClinicalTrials.gov NCT05690516.


Subject(s)
COVID-19 , Masks , SARS-CoV-2 , Self Report , Humans , Female , Male , COVID-19/prevention & control , COVID-19/epidemiology , Middle Aged , Norway/epidemiology , Adult , Respiratory Tract Infections/prevention & control , Aged , Pandemics/prevention & control , Coronavirus Infections/prevention & control , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Pneumonia, Viral/prevention & control , Pneumonia, Viral/epidemiology , Pneumonia, Viral/transmission , Betacoronavirus
7.
Nat Microbiol ; 9(8): 2038-2050, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39075235

ABSTRACT

Circulating bat coronaviruses represent a pandemic threat. However, our understanding of bat coronavirus pathogenesis and transmission potential is limited by the lack of phenotypically characterized strains. We created molecular clones for the two closest known relatives of SARS-CoV-2, BANAL-52 and BANAL-236. We demonstrated that BANAL-CoVs and SARS-CoV-2 have similar replication kinetics in human bronchial epithelial cells. However, BANAL-CoVs have impaired replication in human nasal epithelial cells and in the upper airway of mice. We also observed reduced pathogenesis in mice and diminished transmission in hamsters. Further, we observed that diverse bat coronaviruses evade interferon and downregulate major histocompatibility complex class I. Collectively, our study demonstrates that despite high genetic similarity across bat coronaviruses, prediction of pandemic potential of a virus necessitates functional characterization. Finally, the restriction of bat coronavirus replication in the upper airway highlights that transmission potential and innate immune restriction can be uncoupled in this high-risk family of emerging viruses.


Subject(s)
COVID-19 , Chiroptera , Immunity, Innate , SARS-CoV-2 , Virus Replication , Animals , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Chiroptera/virology , Chiroptera/immunology , COVID-19/transmission , COVID-19/virology , COVID-19/immunology , Mice , Cricetinae , Immune Evasion , Epithelial Cells/virology , Epithelial Cells/immunology , Coronavirus Infections/transmission , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus/immunology , Coronavirus/genetics , Coronavirus/classification , Coronavirus/physiology , Coronavirus/pathogenicity , Cell Line , Female
8.
J Virol ; 98(7): e0075324, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38829136

ABSTRACT

Porcine hemagglutinating encephalomyelitis virus (PHEV), a neurotropic betacoronavirus, is prevalent in natural reservoir pigs and infects mice. This raises concerns about host jumping or spillover, but little is known about the cause of occurrence. Here, we revealed that dipeptidyl peptidase 4 (DPP4) is a candidate binding target of PHEV spikes and works as a broad barrier to overcome. Investigations of the host breadth of PHEV confirmed that cells derived from pigs and mice are permissive to virus propagation. Both porcine DPP4 and murine DPP4 have high affinity for the viral spike receptor-binding domain (RBD), independent of their catalytic activity. Loss of DPP4 expression results in limited PHEV infection. Structurally, PHEV spike protein binds to the outer surface of blades IV and V of the DPP4 ß-propeller domain, and the DPP4 residues N229 and N321 (relative to human DPP4 numbering) participate in RBD binding via its linked carbohydrate entities. Removal of these N-glycosylations profoundly enhanced the RBD-DPP4 interaction and viral invasion, suggesting they act as shielding in PHEV infection. Furthermore, we found that glycosylation, rather than structural differences or surface charges, is more responsible for DPP4 recognition and species barrier formation. Overall, our findings shed light on virus-receptor interactions and highlight that PHEV tolerance to DPP4 orthologs is a putative determinant of its cross-species transmission or host range expansion.IMPORTANCEPHEV is a neurotropic betacoronavirus that is circulating worldwide and has raised veterinary and economic concerns. In addition to being a reservoir species of pigs, PHEV can also infect wild-type mice, suggesting a "host jump" event. Understanding cross-species transmission is crucial for disease prevention and control but remains to be addressed. Herein, we show that the multifunctional receptor DPP4 plays a pivotal role in the host tropism of PHEV and identifies the conserved glycosylation sites in DPP4 responsible for this restriction. These findings highlight that the ability of PHEV to utilize DPP4 orthologs potentially affects its natural host expansion.


Subject(s)
Dipeptidyl Peptidase 4 , Host Specificity , Spike Glycoprotein, Coronavirus , Animals , Humans , Mice , Betacoronavirus 1/metabolism , Cell Line , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Coronavirus Infections/metabolism , Coronavirus Infections/transmission , Deltacoronavirus , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl Peptidase 4/genetics , Glycosylation , HEK293 Cells , Protein Binding , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Swine , Swine Diseases/virology , Virus Internalization
9.
Front Public Health ; 12: 1386495, 2024.
Article in English | MEDLINE | ID: mdl-38827618

ABSTRACT

Introduction: Mitigating the spread of infectious diseases is of paramount concern for societal safety, necessitating the development of effective intervention measures. Epidemic simulation is widely used to evaluate the efficacy of such measures, but realistic simulation environments are crucial for meaningful insights. Despite the common use of contact-tracing data to construct realistic networks, they have inherent limitations. This study explores reconstructing simulation networks using link prediction methods as an alternative approach. Methods: The primary objective of this study is to assess the effectiveness of intervention measures on the reconstructed network, focusing on the 2015 MERS-CoV outbreak in South Korea. Contact-tracing data were acquired, and simulation networks were reconstructed using the graph autoencoder (GAE)-based link prediction method. A scale-free (SF) network was employed for comparison purposes. Epidemic simulations were conducted to evaluate three intervention strategies: Mass Quarantine (MQ), Isolation, and Isolation combined with Acquaintance Quarantine (AQ + Isolation). Results: Simulation results showed that AQ + Isolation was the most effective intervention on the GAE network, resulting in consistent epidemic curves due to high clustering coefficients. Conversely, MQ and AQ + Isolation were highly effective on the SF network, attributed to its low clustering coefficient and intervention sensitivity. Isolation alone exhibited reduced effectiveness. These findings emphasize the significant impact of network structure on intervention outcomes and suggest a potential overestimation of effectiveness in SF networks. Additionally, they highlight the complementary use of link prediction methods. Discussion: This innovative methodology provides inspiration for enhancing simulation environments in future endeavors. It also offers valuable insights for informing public health decision-making processes, emphasizing the importance of realistic simulation environments and the potential of link prediction methods.


Subject(s)
Contact Tracing , Coronavirus Infections , Disease Outbreaks , Middle East Respiratory Syndrome Coronavirus , Humans , Republic of Korea/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/prevention & control , Coronavirus Infections/epidemiology , Contact Tracing/methods , Disease Outbreaks/prevention & control , Quarantine , Computer Simulation
10.
Vet Microbiol ; 293: 110101, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718529

ABSTRACT

Cross-species transmission of coronaviruses has been continuously posing a major challenge to public health. Pigs, as the major animal reservoirs for many zoonotic viruses, frequently mediate viral transmission to humans. This study comprehensively mapped the relationship between human and porcine coronaviruses through in-depth bioinformatics analysis. We found that human coronavirus OC43 and porcine coronavirus PHEV share a close phylogenetic relationship, evidenced by high genomic homology, similar codon usage patterns and comparable tertiary structure in spike proteins. Inoculation of infectious OC43 viruses in organoids derived from porcine small and large intestine demonstrated that porcine intestinal organoids (pIOs) are highly susceptible to human coronavirus OC43 infection and support infectious virus production. Using transmission electron microscopy, we visualized OC43 viral particles in both intracellular and extracellular compartments, and observed abnormalities of multiple organelles in infected organoid cells. Robust OC43 infections in pIOs result in a significant reduction of organoids viability and widespread cell death. This study bears essential implications for better understanding the evolutionary origin of human coronavirus OC43, and provides a proof-of-concept for using pIOs as a model to investigate cross-species transmission of human coronavirus.


Subject(s)
Computational Biology , Coronavirus Infections , Coronavirus OC43, Human , Intestines , Organoids , Phylogeny , Animals , Organoids/virology , Swine , Humans , Coronavirus Infections/virology , Coronavirus Infections/transmission , Coronavirus Infections/veterinary , Coronavirus OC43, Human/physiology , Coronavirus OC43, Human/genetics , Intestines/virology , Swine Diseases/virology , Swine Diseases/transmission , Genome, Viral
11.
Front Cell Infect Microbiol ; 14: 1378804, 2024.
Article in English | MEDLINE | ID: mdl-38736749

ABSTRACT

Introduction: Seasonal human coronavirus NL63 (HCoV-NL63) is a frequently encountered virus linked to mild upper respiratory infections. However, its potential to cause more severe or widespread disease remains an area of concern. This study aimed to investigate a rare localized epidemic of HCoV-NL63-induced respiratory infections among pediatric patients in Guilin, China, and to understand the viral subtype distribution and genetic characteristics. Methods: In this study, 83 pediatric patients hospitalized with acute respiratory infections and positive for HCoV-NL63 were enrolled. Molecular analysis was conducted to identify the viral subgenotypes and to assess genetic variations in the receptor-binding domain of the spiking protein. Results: Among the 83 HCoV-NL63-positive children, three subgenotypes were identified: C4, C3, and B. Notably, 21 cases exhibited a previously unreported subtype, C4. Analysis of the C4 subtype revealed a unique amino acid mutation (I507L) in the receptor-binding domain of the spiking protein, which was also observed in the previously reported C3 genotype. This mutation may suggest potential increases in viral transmissibility and pathogenicity. Discussion: The findings of this study highlight the rapid mutation dynamics of HCoV-NL63 and its potential for increased virulence and epidemic transmission. The presence of a unique mutation in the C4 subtype, shared with the C3 genotype, raises concerns about the virus's evolving nature and its potential public health implications. This research contributes valuable insights into the understanding of HCoV-NL63's epidemiology and pathogenesis, which is crucial for effective disease prevention and control strategies. Future studies are needed to further investigate the biological significance of the observed mutation and its potential impact on the virus's transmissibility and pathogenicity.


Subject(s)
Coronavirus Infections , Coronavirus NL63, Human , Epidemics , Genotype , Phylogeny , Respiratory Tract Infections , Humans , Coronavirus NL63, Human/genetics , China/epidemiology , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Coronavirus Infections/transmission , Child , Female , Male , Child, Preschool , Respiratory Tract Infections/virology , Respiratory Tract Infections/epidemiology , Infant , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Seasons , Mutation , Adolescent
12.
Int J Infect Dis ; 145: 107097, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38750721

ABSTRACT

BACKGROUND: Removal of zero-COVID restrictions in China led to a surge in COVID-19 cases. In response, countries imposed restrictions on Chinese travelers. However, border policies imposed may not have been informed by accurate data and may not have provided substantial benefits. METHODS: We analyzed quarantines sufficient to prevent additional in-country transmission for February 13-19, 2023 based on World Health Organization (WHO) and self-reported infections to estimate prevalence. RESULTS: We have shown that self-reported prevalence data indicated more stringent border restrictions compared to WHO-published prevalence statistics. No travel restrictions were required for Singapore, South Korea, and Japan so that infections would not be greater than with complete border closure. However, a 1-, 2-, and 3-day quarantine were indicated for England, Germany, and Scotland respectively. A 10-, 13-, and 14-day quarantine were required for Italy, France, and the Philippines, respectively, to prevent an increase in within-country infections due to travel. Vietnam and Thailand required a complete border shutdown. CONCLUSION: Our results demonstrated the necessity for accurate and timely reporting of pandemic statistics to prevent an increase in viral spread. Through the minimum quarantine analysis, countries can use science to determine policy, minimize international friction, and improve the cost-efficiency of interventions.


Subject(s)
COVID-19 , Pandemics , Quarantine , SARS-CoV-2 , Travel , Humans , COVID-19/transmission , COVID-19/prevention & control , COVID-19/epidemiology , China/epidemiology , Pandemics/prevention & control , Coronavirus Infections/transmission , Coronavirus Infections/prevention & control , Coronavirus Infections/epidemiology , Betacoronavirus , Prevalence , Health Policy , Communicable Disease Control/methods , COVID-19 Testing/methods
13.
Placenta ; 152: 39-52, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38788480

ABSTRACT

INTRODUCTION: Several factors influence transmission of 2019-nCoV from mother to fetus during pregnancy, thus the dynamics of vertical transmission is unclear. The role of cellular protective factors, namely a 90 KDa glycoprotein, Early pregnancy-associated protein (Epap-1), expressed by placental endothelial cells in women during early pregnancy would provide an insight into role of placental factors in virus transmission. Since viral spike protein binding to the ACE2 receptors of the host cells promotes virus invasion in placental tissue, an analysis of effects of Epap-1 on the Spike-ACE2 protein binding was studied. METHODS: Epap-1 was isolated from MTP placental tissue. Molecular interaction of Epap-1 and variants of the spike was analyzed in silco. The interaction of Epap-1 with Spike and RBD were analyzed using ELISA and immunofluorescence studies. RESULTS: The results in silico showed an interaction of Epap-1 with S-protein at RBD region involving K417, Y449, Y453, Y456, Y473, Q474, F486, Q498, N501 residues of spike with Y61, F287, I302, N303, N305, S334, N465, G467, N468 residues of Epap-1 leading to interference of S-protein and ACE2 interaction [1]. Further, the interaction is conserved among the variants. The studies in vitro confirm that Epap-1 affects S protein-ACE2 and RBD- ACE2 binding, thus suggesting that during early pregnancy, SARS CoV-2 infection may be protected by Epap-1 protein present in placental tissue. The results were further confirmed by pseudovirus expressing Spike and RBD in an infection assay. DISCUSSION: Epap-1 interferes with Spike and RBD interaction with ACE2, suggesting a possible mechanism of the antiviral environment during pregnancy.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Infectious Disease Transmission, Vertical , Placenta , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Female , Humans , Pregnancy , Angiotensin-Converting Enzyme 2/metabolism , Betacoronavirus/metabolism , Coronavirus Infections/transmission , Coronavirus Infections/metabolism , Coronavirus Infections/virology , COVID-19/transmission , COVID-19/metabolism , Pandemics , Peptidyl-Dipeptidase A/metabolism , Placenta/metabolism , Placenta/virology , Pneumonia, Viral/metabolism , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/virology , Pregnancy Proteins/metabolism , Protein Binding , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/metabolism
14.
Epidemiol Infect ; 152: e90, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38770587

ABSTRACT

We analyzed data from a community-based acute respiratory illness study involving K-12 students and their families in southcentral Wisconsin and assessed household transmission of two common seasonal respiratory viruses - human metapneumovirus (HMPV) and human coronaviruses OC43 and HKU1 (HCOV). We found secondary infection rates of 12.2% (95% CI: 8.1%-17.4%) and 19.2% (95% CI: 13.8%-25.7%) for HMPV and HCOV, respectively. We performed individual- and family-level regression models and found that HMPV transmission was positively associated age of the index case (individual model: p = .016; family model: p = .004) and HCOV transmission was positively associated with household density (family model: p = .048). We also found that the age of the non-index case was negatively associated with transmission of both HMPV (individual model: p = .049) and HCOV (individual model: p = .041), but we attributed this to selection bias from the original study design. Understanding household transmission of common respiratory viruses like HMPV and HCOV may help to broaden our understanding of the overall disease burden and establish methods to prevent the spread of disease from low- to high-risk populations.


Subject(s)
Coronavirus Infections , Family Characteristics , Metapneumovirus , Paramyxoviridae Infections , Humans , Paramyxoviridae Infections/transmission , Paramyxoviridae Infections/epidemiology , Wisconsin/epidemiology , Female , Young Adult , Male , Coronavirus Infections/transmission , Coronavirus Infections/epidemiology , Adult , Adolescent , Child , Coronavirus , Seasons , Middle Aged , Child, Preschool , Respiratory Tract Infections/transmission , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology
15.
J Travel Med ; 31(4)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38630887

ABSTRACT

BACKGROUND: The international flight network creates multiple routes by which pathogens can quickly spread across the globe. In the early stages of infectious disease outbreaks, analyses using flight passenger data to identify countries at risk of importing the pathogen are common and can help inform disease control efforts. A challenge faced in this modelling is that the latest aviation statistics (referred to as contemporary data) are typically not immediately available. Therefore, flight patterns from a previous year are often used (referred to as historical data). We explored the suitability of historical data for predicting the spatial spread of emerging epidemics. METHODS: We analysed monthly flight passenger data from the International Air Transport Association to assess how baseline air travel patterns were affected by outbreaks of Middle East respiratory syndrome (MERS), Zika and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) over the past decade. We then used a stochastic discrete time susceptible-exposed-infected-recovered (SEIR) metapopulation model to simulate the global spread of different pathogens, comparing how epidemic dynamics differed in simulations based on historical and contemporary data. RESULTS: We observed local, short-term disruptions to air travel from South Korea and Brazil for the MERS and Zika outbreaks we studied, whereas global and longer-term flight disruptions occurred during the SARS-CoV-2 pandemic. For outbreak events that were accompanied by local, small and short-term changes in air travel, epidemic models using historical flight data gave similar projections of the timing and locations of disease spread as when using contemporary flight data. However, historical data were less reliable to model the spread of an atypical outbreak such as SARS-CoV-2, in which there were durable and extensive levels of global travel disruption. CONCLUSION: The use of historical flight data as a proxy in epidemic models is an acceptable practice, except in rare, large epidemics that lead to substantial disruptions to international travel.


Subject(s)
Air Travel , COVID-19 , Disease Outbreaks , SARS-CoV-2 , Zika Virus Infection , Humans , Air Travel/statistics & numerical data , COVID-19/epidemiology , COVID-19/transmission , COVID-19/prevention & control , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/prevention & control , Communicable Diseases/epidemiology , Communicable Diseases/transmission , Travel/statistics & numerical data , Aircraft , Global Health
16.
Can J Dent Hyg ; 58(1): 48-63, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38505316

ABSTRACT

Background: Since the outbreak of COVID-19, how to reduce the risk of spreading viruses and other microorganisms while performing aerosolgenerating procedures (AGPs) has become a challenging question within the dental and dental hygiene communities. The purpose of this position paper is to summarize the evidence of the effectiveness of various mitigation methods used to reduce the risk of infection transmission during AGPs in dentistry. Methods: The authors searched 6 databases-MEDLINE, EMBASE, Scopus, Web of Science, Cochrane Library, and Google Scholar-for relevant scientific evidence published between January 2012 and December 2022 to answer 6 research questions about the risk of transmission, methods, devices, and personal protective equipment (PPE) used to reduce contact with microbial pathogens and limit the spread of aerosols. Results: A total of 78 studies fulfilled the eligibility criteria. The literature on the risk of infection transmission including SARS-CoV-2 between dental hygienists and their patients is limited. Although several mouthrinses are effective in reducing bacterial contaminations in aerosols, their effectiveness against SARS-CoV-2 is also limited. The combined use of eyewear, masks, and face shields is effective in preventing contamination of the facial and nasal region while performing AGPs. High-volume evacuation with or without an intraoral suction, low-volume evacuation, saliva ejector, and rubber dam (when appropriate) have shown effectiveness in reducing aerosol transmission beyond the generation site. Finally, the appropriate combination of ventilation and filtration in dental operatories is effective in limiting the spread of aerosols. Discussion and Conclusion: Aerosols produced during clinical procedures can pose a risk of infection transmission between dental hygienists and their patients. The implementation of practices supported by available evidence will ensure greater patient and provider safety in oral health settings. More studies in oral health clinical environments would shape future practices and protocols, ultimately to ensure the delivery of safe clinical care.


Contexte: Depuis l'éclosion de la COVID-19, la façon de réduire le risque de propagation de virus et d'autres microorganismes tout en effectuant des interventions générant des aérosols (IGA) est devenue un enjeu complexe au sein des communautés de la médecine dentaire et de l'hygiène dentaire. L'objectif de cet exposé de position est de résumer les données probantes de l'efficacité des diverses méthodes d'atténuation utilisées pour réduire le risque de transmission des infections pendant les IGA en médecine dentaire. Méthodes: Les auteurs ont effectué des recherches dans MEDLINE, EMBASE, Scopus, Web of Science, Cochrane Library et Google Scholar pour trouver des preuves scientifiques pertinentes publiées entre janvier 2012 et décembre 2022 afin de répondre à 6 questions de recherche sur le risque de transmission, les méthodes, les dispositifs et l'équipement de protection individuelle (EPI) utilisés pour réduire le contact avec les agents pathogènes microbiens et limiter la propagation des aérosols. Résultats: Au total, 78 études ont satisfait aux critères d'admissibilité. La documentation est limitée en ce qui concerne le risque de transmission des infections, y compris le SRAS-CoV-2, entre les hygiénistes dentaires et leurs patients. Bien que plusieurs rince-bouches soient efficaces pour réduire la contamination bactérienne dans les aérosols, leur efficacité contre le SRAS-CoV-2 est limitée. L'utilisation combinée de lunettes, de masques et d'écrans faciaux est efficace pour prévenir la contamination de la région faciale et nasale lors de l'exécution d'IGA. L'évacuation à volume élevé avec ou sans aspiration intraorale, l'évacuation à faible volume, l'aspirateur de salive et la digue dentaire en caoutchouc (le cas échéant) ont démontré une efficacité à réduire la transmission des aérosols au-delà du site de production. Enfin, la combinaison appropriée de ventilation et de filtration dans les salles de traitement dentaire permet de limiter efficacement la propagation des aérosols. Discussion et conclusion: Les aérosols produits lors des interventions cliniques peuvent présenter un risque de transmission des infections entre les hygiénistes dentaires et leurs patients. La mise en oeuvre de pratiques appuyées par les données probantes disponibles assurera une plus grande sécurité des patients et des prestataires dans les milieux de santé buccodentaire. Un plus grand nombre d'études dans les environnements cliniques de santé buccodentaire permettrait de façonner les pratiques et les protocoles futurs dans le but d'assurer la prestation sécuritaire des soins cliniques.


Subject(s)
Aerosols , COVID-19 , Dental Hygienists , Personal Protective Equipment , SARS-CoV-2 , Humans , COVID-19/transmission , COVID-19/prevention & control , COVID-19/epidemiology , Canada/epidemiology , Pandemics/prevention & control , United States/epidemiology , Infectious Disease Transmission, Patient-to-Professional/prevention & control , Coronavirus Infections/transmission , Coronavirus Infections/prevention & control , Coronavirus Infections/epidemiology , Betacoronavirus , Pneumonia, Viral/transmission , Pneumonia, Viral/prevention & control , Pneumonia, Viral/epidemiology , Infection Control, Dental/methods
17.
Science ; 383(6679): 141-145, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38207045
18.
J Virol ; 97(12): e0133823, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38009916

ABSTRACT

IMPORTANCE: Betacoronaviruses, including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and mouse hepatitis virus (MHV), exploit the lysosomal exocytosis pathway for egress. However, whether all betacoronaviruses members use the same pathway to exit cells remains unknown. Here, we demonstrated that porcine hemagglutinating encephalomyelitis virus (PHEV) egress occurs by Arl8b-dependent lysosomal exocytosis, a cellular egress mechanism shared by SARS-CoV-2 and MHV. Notably, PHEV acidifies lysosomes and activates lysosomal degradative enzymes, while SARS-CoV-2 and MHV deacidify lysosomes and limit the activation of lysosomal degradative enzymes. In addition, PHEV release depends on V-ATPase-mediated lysosomal pH. Furthermore, this is the first study to evaluate ßCoV using lysosome for spreading through the body, and we have found that lysosome played a critical role in PHEV neural transmission and brain damage caused by virus infection in the central nervous system. Taken together, different betacoronaviruses could disrupt lysosomal function differently to exit cells.


Subject(s)
Betacoronavirus 1 , Coronavirus Infections , Exocytosis , Lysosomes , Neurons , Animals , Mice , Betacoronavirus 1/metabolism , Lysosomes/enzymology , Lysosomes/metabolism , Lysosomes/virology , Murine hepatitis virus/metabolism , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Neurons/virology , SARS-CoV-2/metabolism , Swine/virology , Hydrogen-Ion Concentration , Vacuolar Proton-Translocating ATPases/metabolism , Coronavirus Infections/pathology , Coronavirus Infections/transmission , Coronavirus Infections/virology
19.
J Virol ; 97(11): e0082923, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37882520

ABSTRACT

IMPORTANCE: Several coronaviruses (CoVs) have been detected in domesticated, farmed, and wild meso-carnivores, causing a wide range of diseases and infecting diverse species, highlighting their important but understudied role in the epidemiology of these viruses. Assessing the viral diversity hosted in wildlife species is essential to understand their significance in the cross-species transmission of CoVs. Our focus here was on CoV discovery in meso-carnivores in the Northeast United States as a potential "hotspot" area with high density of humans and urban wildlife. This study identifies novel alphacoronaviruses circulating in multiple free-ranging wild and domestic species in this area and explores their potential epidemiological importance based on regions of the Spike gene, which are relevant for virus-host interactions.


Subject(s)
Alphacoronavirus , Carnivora , Feces , Saliva , Animals , Humans , Alphacoronavirus/classification , Alphacoronavirus/genetics , Alphacoronavirus/isolation & purification , Animals, Domestic/virology , Animals, Wild/virology , Carnivora/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/veterinary , Feces/virology , Host Microbial Interactions , New England/epidemiology , Saliva/virology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Viral Zoonoses/transmission , Viral Zoonoses/virology
20.
Article in Spanish | IBECS | ID: ibc-226406

ABSTRACT

Introducción La infección por SARS-CoV-2 durante la gestación y su repercusión en el recién nacido eran, en los primeros meses de la pandemia, desconocidas. Recientes estudios han aportado información sobre la afectación clínica en el recién nacido y su evolución. En este trabajo se muestra cómo varía la inmunidad pasiva en el recién nacido con relación al momento de infección SARS-CoV-2 materno. Población y método Estudio observacional, prospectivo y longitudinal en un hospital de tercer nivel. Se recogieron datos epidemiológicos y clínicos de las madres y sus recién nacidos desde mayo del 2020 hasta junio del 2021. Resultados Se ha incluido a un total de 109 madres y 109 neonatos. El 28,4% de las infecciones maternas fueron en el primer trimestre, el 24,8% en el segundo y el 58,8% en el tercero. El 56% de las infecciones maternas fueron sintomáticas, solo una gestante con infección respiratoria grave ingresó en Cuidados Intensivos. La edad gestacional media de los recién nacidos fue de 39 semanas, con un peso medio de 3.232g y un perímetro craneal de 35cm. Ocho recién nacidos hijos de madre con SARS-CoV-2 requirieron ingreso en la UCI neonatal: 2 por ictericia, 2 por distrés respiratorio, uno por prematuridad moderada y 3 por otras causas no relacionadas con infección atribuible a SARS-CoV-2. Los anticuerpos tipo IgG fueron positivas en el 56,9% de los recién nacidos. De las madres infectadas durante el primer trimestre, las IgG fueron positivas en el 32,2% de los recién nacidos, en el segundo trimestre resultaron positivos el 81,5% y en el tercero, el 58,8%. Ningún neonato presentó IgM positivas. Conclusiones La infección por SARS-CoV-2 durante la gestación proporciona anticuerpos IgG a la mitad de los recién nacidos. La presencia de anticuerpos en el recién nacido es más probable cuando la infección se ha producido en el segundo trimestre de gestación (AU)


Introduction SARS-CoV-2 infection during pregnancy and its impact on the newborn were, in the first months of the pandemic, unknown. Recent studies have provided information on the clinical involvement in the newborn and its evolution. This work shows how passive immunity varies in the newborn in relation to the moment of maternal SARS-CoV-2 infection during pregnancy. Population and method Observational, prospective and longitudinal study in a third level hospital. Epidemiological and clinical data from mothers and their newborns were collected from May 2020 to June 2021. Results A total of 109 mothers and 109 neonates have been included. 28.4% of maternal infections were in the first trimester, 24.8% during the second and 58.8% in the third. 56% of maternal infections were symptomatic and only one pregnant woman with severe respiratory infection was admitted to intensive care. The mean gestational age of the newborns was 39 weeks, with a mean weight of 3232g and a head circumference of 35cm. Eight newborns born from mothers with SARS-CoV-2 required admission to the neonatal ICU: 2 due to jaundice, 2 due to respiratory distress, 1 due to moderate prematurity, and 3 due to other causes unrelated to infection attributable to SARS-CoV-2. IgG-type antibodies were positive in 56.9% of newborns. Of the mothers infected during the 1st trimester, IgG were positive in 32.2% of the newborns, in the second trimester 81.5% were positive and in the third 58.8%. No neonate had positive IgM. Conclusions SARS-CoV-2 infection during pregnancy provides IgG antibodies to half of newborns. The presence of antibodies in the newborn is more likely when the infection has occurred in the second trimester of pregnancy (AU)


Subject(s)
Humans , Female , Pregnancy , Infant, Newborn , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Immunity, Maternally-Acquired , Maternal-Fetal Exchange , Immunoglobulins/immunology , Prospective Studies , Longitudinal Studies
SELECTION OF CITATIONS
SEARCH DETAIL