Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Viruses ; 14(11)2022 10 25.
Article in English | MEDLINE | ID: mdl-36366439

ABSTRACT

Since their discovery in the 1950s, rhinoviruses (RVs) have been recognized as a major causative agent of the "common cold" and cold-like illnesses, accounting for more than 50% of upper respiratory tract infections. However, more than that, respiratory viral infections are responsible for approximately 50% of asthma exacerbations in adults and 80% in children. In addition to causing exacerbations of asthma, COPD and other chronic lung diseases, RVs have also been implicated in the pathogenesis of lower respiratory tract infections including bronchiolitis and community acquired pneumonia. Finally, early life respiratory viral infections with RV have been associated with asthma development in children. Due to the vast genetic diversity of RVs (approximately 160 known serotypes), recurrent infection is common. RV infections are generally acquired in the community with transmission occurring via inhalation of aerosols, respiratory droplets or fomites. Following the outbreak of coronavirus disease 2019 (COVID-19), exposure to RV and other respiratory viruses was significantly reduced due to social-distancing, restrictions on social gatherings, and increased hygiene protocols. In the present review, we summarize the impact of COVID-19 preventative measures on the incidence of RV infection and its sequelae.


Subject(s)
Asthma , COVID-19 , Communicable Diseases , Picornaviridae Infections , Respiratory Tract Infections , Child , Adult , Humans , Rhinovirus/genetics , COVID-19/prevention & control , Physical Distancing , Asthma/complications , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/complications , Picornaviridae Infections/epidemiology , Picornaviridae Infections/prevention & control , Picornaviridae Infections/complications
2.
Virol J ; 19(1): 36, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246187

ABSTRACT

BACKGROUND: Despite the clinical burden attributable to rhinovirus (RV) infections, the RV transmission dynamics and the impact of interventions on viral transmission remain elusive. METHODS: A total of 3,935 nasopharyngeal specimens were examined, from which the VP4/VP2 gene was sequenced and genotyped. RV transmission clusters were reconstructed using the genetic threshold of 0.005 substitutions/site, estimated from the global VP4/VP2 sequences. A transmission cluster is characterized by the presence of at least two individuals (represent by nodes), whose viral sequences are genetically linked (represent by undirected edges) at the estimated genetic distance threshold supported by bootstrap value of ≥ 90%. To assess the impact of facemask, pleconaril and social distancing on RV transmission clusters, trials were simulated for interventions with varying efficacy and were evaluated based on the reduction in the number of infected patients (nodes) and the reduction in the number of nodes-connecting edges. The putative impact of intervention strategies on RV transmission clusters was evaluated through 10,000 simulations. RESULTS: A substantial clustering of 168 RV transmission clusters of varying sizes were observed. This suggests that RV disease burden observed in the population was largely due to multiple sub-epidemics, predominantly driven by RV-A, followed by RV-C and -B. No misclassification of RV species and types were observed, suggesting the specificity and sensitivity of the analysis. Through 10,000 simulations, it was shown that social distancing may be effective in decelerating RV transmission, by removing more than 95% of nodes and edges within the RV transmission clusters. However, facemask removed less than 8% and 66% of nodes and edges, respectively, conferring moderate advantage in limiting RV transmission. CONCLUSION: Here, we presented a network-based approach of which the degree of RV spread that fuel disease transmission in the region was mapped for the first time. The utilization of RV transmission clusters in assessing the putative impact of interventions on disease transmission at the population level was demonstrated.


Subject(s)
Enterovirus Infections , Picornaviridae Infections , Respiratory Tract Infections , Genotype , Humans , Nasopharynx , Phylogeny , Picornaviridae Infections/epidemiology , Picornaviridae Infections/prevention & control , Rhinovirus/genetics
3.
BMC Infect Dis ; 22(1): 253, 2022 Mar 14.
Article in English | MEDLINE | ID: mdl-35287614

ABSTRACT

BACKGROUND: Human rhinovirus (HRV) is the predominant etiological agent of the common cold in children and adults. A recent study showed that the inhibitory effect of face masks on viral shedding of HRV was less prominent than that on other respiratory viruses. Considering that most Chinese people have worn face masks in public area since the outbreak of coronavirus disease 2019, we aimed to find out whether HRV prevailed among children in 2020 and demonstrate the details of the epidemiological features of HRV under such a special circumstance. METHODS: We summarized the incidences of various respiratory virus infections in patients who visited the Children's Hospital of Fudan University during 2018-2020, and genotyped HRV positive nasopharyngeal specimens collected from 316 inpatients and 72 outpatients that visited the hospital in 2020. RESULTS: There was a major prevalence of HRV among children in the latter half of 2020, with a clear seasonality that HRV-As prevailed in summer while HRV-Cs in autumn. HRV-As were more prone to cause severe lower respiratory tract infections (LRTI), while HRV-Cs were closely associated with childhood wheezing. The predominant genotypes were A11, A28, A47, A82, A101, C40 and C43. Notably, A21, A82 and A101 took up larger proportions in severe cases than in non-severe cases. CONCLUSIONS: Our findings described a major prevalence of HRVs among children in 2020, which highlight the unique transmitting pattern of HRV and help to narrow the targets for antiviral strategies.


Subject(s)
COVID-19 , Picornaviridae Infections , Adult , Child , China/epidemiology , Humans , Masks , Picornaviridae Infections/epidemiology , Picornaviridae Infections/prevention & control , Rhinovirus/genetics
4.
Viruses ; 14(3)2022 03 18.
Article in English | MEDLINE | ID: mdl-35337040

ABSTRACT

Duck viral hepatitis type I (DVH I) is a lethal disease in ducklings caused by duck hepatitis A virus (DHAV). Although the commercial vaccine is available for vaccination of one-day-old ducklings or breeder ducks, the disease is still prevalent due to the delayed immune response in ducklings and variable maternal antibody levels in breeder duck flocks. To explore the feasibility of duck interferon-α (DuIFN-α) for control of DVH I, DuIFN-α was expressed as an elastin-like polypeptide (ELP) fusion protein (ELP-DuIFN-α) in E. coli and purified by inverse phase transition cycling (ITC). After detection of its cytotoxicity, bioactivity, plasma stability and serum half-life, the protective efficacy of ELP-DuIFN-α against DHAV-1 infection of embryos or ducklings was evaluated using different treatment routes at different infection times. The results show that ELP-DuIFN-α was correctly expressed and purified to more than 90% purity after two cycles of ITC. The purified fusion protein had a specific anti-DHAV-1 activity of 6.0 × 104 IU/mg protein, significantly extended plasma stability and serum half-life without overt cytotoxicity. After allantoic injection with ELP-DuIFN-α pre-infection, co-infection or post-infection with DHAV-1, 5/5, 5/5 or 4/5 embryos survived from the virus challenge. After intramuscular injection or oral administration with ELP-DuIFN-α, 3/5 or 4/5 ducklings survived from co-infection with DHAV-1. After oral administration with ELP-DuIFN-α pre-infection, co-infection or post-infection with DHAV-1, 3/5, 4/5 or 4/5 ducklings survived from the virus challenge, and the relative transcription levels of interferon-stimulated genes were significantly higher than the normal control group and virus challenge control group (p < 0.01). These experimental data suggest that ELP-DuIFN-α can be used as a long-lasting anti-DHAV-1 reagent.


Subject(s)
Coinfection , Hepatitis A virus , Hepatitis A , Hepatitis Virus, Duck , Hepatitis, Viral, Animal , Picornaviridae Infections , Poultry Diseases , Animals , Ducks , Escherichia coli , Hepatitis Virus, Duck/genetics , Hepatitis, Viral, Animal/prevention & control , Interferon-alpha , Picornaviridae Infections/prevention & control , Picornaviridae Infections/veterinary
5.
Nutrients ; 13(12)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34959949

ABSTRACT

Acute respiratory infections are an important health concern. Traditionally, polysaccharide-enriched extracts from plants, containing immunomodulatory rhamnogalacturonan-I (RG-1), were used prophylactically. We established the effects of dietary supplementation with carrot-derived RG-I (cRG-I, 0-0.3-1.5 g/day) in 177 healthy individuals (18-65 years) on symptoms following infection with rhinovirus strain 16 (RV16). Primary outcomes were changes in severity and duration of symptoms, and viral load in nasal lavage. Secondary outcomes were changes in innate immune and anti-viral responses, reflected by CXCL10 and CXCL8 levels and cell differentials in nasal lavage. In a nested cohort, exploratory transcriptome analysis was conducted on nasal epithelium. Intake of cRG-I was safe, well-tolerated and accelerated local cellular and humoral innate immune responses induced by RV16 infection, with the strongest effects at 1.5 g/d. At 0.3 g/d, a faster interferon-induced response, induction of the key anti-viral gene EIF2AK2, faster viral clearance, and reduced symptom severity (-20%) and duration (-25%) were observed. Anti-viral responses, viral clearance and symptom scores at 1.5 g/d were in between those of 0 and 0.3 g/d, suggesting a negative feedback loop preventing excessive interferon responses. Dietary intake of cRG-I accelerated innate immune and antiviral responses, and reduced symptoms of an acute respiratory viral infection.


Subject(s)
Antiviral Agents , Chemokine CXCL10/metabolism , Daucus carota/chemistry , Dietary Supplements , Immunity, Innate/drug effects , Interleukin-8/metabolism , Pectins/pharmacology , Pectins/therapeutic use , Phytotherapy , Picornaviridae Infections/drug therapy , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Rhinovirus , Adolescent , Adult , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Male , Middle Aged , Nasal Lavage , Patient Acuity , Pectins/isolation & purification , Picornaviridae Infections/prevention & control , Treatment Outcome , Young Adult
6.
Microbiol Spectr ; 9(2): e0043021, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34494861

ABSTRACT

Measures intended to limit the spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus at the start of the coronavirus disease 2019 (COVID-19) pandemic resulted in a rapid decrease in other respiratory pathogens. Herein, we describe the trends of respiratory pathogens in a major metropolitan health care system central microbiology reference laboratory before and during the COVID-19 pandemic, with attention to when COVID-19 mitigation measures were implemented and relaxed. During the initial lockdown period, COVID-19 was the primary respiratory pathogen detected by multiplex respiratory panels. As COVID-19 containment measures were relaxed, the first non-COVID respiratory viruses to return to prepandemic levels were members of the rhinovirus/enterovirus family. After the complete removal of COVID-19 precautions at the state level, including an end to mask mandates, we observed the robust return of seasonal coronaviruses, parainfluenza virus, and respiratory syncytial virus. Inasmuch as COVID-19 has dominated the landscape of respiratory infections since early 2020, it is important for clinicians to recognize that the return of non-COVID respiratory pathogens may be rapid and significant when COVID-19 containment measures are removed. IMPORTANCE We describe the return of non-COVID respiratory viruses after the removal of COVID-19 mitigation measures. It is important for the public and physicians to recognize that, after months of COVID-19 being the primary driver of respiratory infection, more typical seasonal respiratory illnesses have returned, and this return is out of the normal season for some of these pathogens. Thus, clinicians and the public must now consider both COVID-19 and other respiratory illnesses when a patient presents with symptomatic respiratory illness.


Subject(s)
COVID-19/epidemiology , COVID-19/prevention & control , Communicable Disease Control , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control , Coxsackievirus Infections/epidemiology , Coxsackievirus Infections/prevention & control , Enterovirus/isolation & purification , Humans , Mandatory Programs/statistics & numerical data , Orthomyxoviridae/isolation & purification , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/prevention & control , Picornaviridae Infections/epidemiology , Picornaviridae Infections/prevention & control , Rhinovirus/isolation & purification , SARS-CoV-2/growth & development , Texas/epidemiology
7.
J Med Virol ; 93(10): 6063-6067, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34228369

ABSTRACT

Social restrictions during the coronavirus disease 2019 pandemic strongly affected the epidemiology of influenza and respiratory syncytial virus (RSV). As rhinovirus seemed to spread despite the restrictions, we aimed to analyze rhinovirus epidemiology in children during the pandemic. This register-based study used data from the Finnish Infectious Disease Register. Nationwide rhinovirus findings from July 2015 to March 2021 were included and stratified by age (0-4, 5-9, and 10-14). Cumulative 14-day incidence per 100000 children was calculated. Four thousand five hundred and seventy six positive rhinovirus findings were included, of which 3788 (82.8%) were among children aged 0-4. The highest recorded incidence was 36.2 among children aged 0-4 in October 2017. The highest recorded incidence during the pandemic period was 13.6 in November 2020. The impact of the restrictions was mostly seen among children aged 0-4 years of age in weeks 14-22 in 2020. The incidence has since remained near reference levels in all age groups. Strict restrictions temporarily interrupted the circulation of rhinovirus in spring 2020. Rhinovirus incidence returned to normal levels soon after the harsh restrictions were lifted. These looser social restrictions prevented RSV and influenza seasons but failed to prevent the spread of rhinovirus.


Subject(s)
Picornaviridae Infections/epidemiology , Picornaviridae Infections/prevention & control , Rhinovirus , Adolescent , Age Factors , COVID-19 , Child , Child, Preschool , Finland/epidemiology , Humans , Infant , Infant, Newborn , Pandemics , Physical Distancing , Respiratory Syncytial Virus, Human , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/virology
8.
Vet Microbiol ; 256: 109038, 2021 May.
Article in English | MEDLINE | ID: mdl-33845332

ABSTRACT

Cholesterol 25-hydroxylase (CH25 H), as a host restriction factor, has been reported to take a broad-spectrum antiviral effect. However, the role of CH25H in Senecavirus A (SVA) infection remains unknown. In this study, we first demonstrate that overexpression of CH25H inhibits SVA replication. Consistently, knockdown or knockout of the endogens CH25H promotes SVA infection. Further, the anti-SVA effect of 25-hydroxycholesterol (25HC), which is the product of CH25H, operates via inhibition of viral attachment and replication. On the other hand, the CH25H mutant (CH25H-M) lacking hydroxylase activity still restricts SVA infection, which can selectively interact and degrade SVA 3A protein via the ubiquitin-proteasome manner. Altogether, these results suggest that CH25H has an antiviral function in SVA infection and provides an alternative manner to control SVA.


Subject(s)
Picornaviridae Infections/prevention & control , Picornaviridae/physiology , Steroid Hydroxylases/metabolism , Virus Replication , Animals , Antiviral Agents , Cell Line , Cricetinae , Gene Knockdown Techniques , Gene Knockout Techniques , Mutation , Picornaviridae Infections/virology , Steroid Hydroxylases/genetics
9.
Vet Microbiol ; 256: 109040, 2021 May.
Article in English | MEDLINE | ID: mdl-33812295

ABSTRACT

The objective of this study was to evaluate the efficacy of ultraviolet C light (UVC) for inactivating Senecavirus A (SVA) on three different experimentally contaminated surfaces commonly found in swine farms. An experimental study under controlled conditions assessed the effect of UVC on an SVA isolate on coupons composed of three surface types: cardboard, cloth, and plastic. Each coupon was inoculated with 2 mL of SVA (107.5 TCID50/mL) and 1 mL of PBS or 1 g of feces on the top or bottom surface of the coupon and allowed to dry (90 min at 25℃). Coupons were exposed to UVC in a commercially available pass-through chamber (PTC) for 5 min or in a simulated supply entry room (SER) for 120 min. After exposure, virus isolation was attempted from each coupon and virus titers were determined in cell culture. The efficacy of UVC was determined by the reduction in virus titer for the UVC treated groups compared to their respective non-treated positive controls. UVC was effective at inactivating SVA on plastic surface free of organic material. The plastic coupons inoculated with SVA and PBS had a significantly lower virus titer (>7-log reduction) in both the PTC and SER when compared to their relative positive controls. All other groups in the PTC and SER had a 2-log reduction or less. The reduction in virus titer on the top and bottom inoculated surfaces, following exposure to UVC, were not statistically different. The data from this study provide some guidance when applying UVC for disinfection in the field.


Subject(s)
Disinfection/methods , Picornaviridae Infections/veterinary , Picornaviridae/radiation effects , Swine Diseases/prevention & control , Animals , Clothing , Feces/virology , Paper , Picornaviridae/physiology , Picornaviridae Infections/prevention & control , Picornaviridae Infections/virology , Plastics , Swine , Swine Diseases/virology , Ultraviolet Rays
12.
Euro Surveill ; 25(49)2020 12.
Article in English | MEDLINE | ID: mdl-33303066

ABSTRACT

BackgroundEvidence for face-mask wearing in the community to protect against respiratory disease is unclear.AimTo assess effectiveness of wearing face masks in the community to prevent respiratory disease, and recommend improvements to this evidence base.MethodsWe systematically searched Scopus, Embase and MEDLINE for studies evaluating respiratory disease incidence after face-mask wearing (or not). Narrative synthesis and random-effects meta-analysis of attack rates for primary and secondary prevention were performed, subgrouped by design, setting, face barrier type, and who wore the mask. Preferred outcome was influenza-like illness. Grading of Recommendations, Assessment, Development and Evaluations (GRADE) quality assessment was undertaken and evidence base deficits described.Results33 studies (12 randomised control trials (RCTs)) were included. Mask wearing reduced primary infection by 6% (odds ratio (OR): 0.94; 95% CI: 0.75-1.19 for RCTs) to 61% (OR: 0.85; 95% CI: 0.32-2.27; OR: 0.39; 95% CI: 0.18-0.84 and OR: 0.61; 95% CI: 0.45-0.85 for cohort, case-control and cross-sectional studies respectively). RCTs suggested lowest secondary attack rates when both well and ill household members wore masks (OR: 0.81; 95% CI: 0.48-1.37). While RCTs might underestimate effects due to poor compliance and controls wearing masks, observational studies likely overestimate effects, as mask wearing might be associated with other risk-averse behaviours. GRADE was low or very low quality.ConclusionWearing face masks may reduce primary respiratory infection risk, probably by 6-15%. It is important to balance evidence from RCTs and observational studies when their conclusions widely differ and both are at risk of significant bias. COVID-19-specific studies are required.


Subject(s)
COVID-19/prevention & control , Eye Protective Devices , Influenza, Human/prevention & control , Masks , Picornaviridae Infections/prevention & control , Respiratory Tract Infections/prevention & control , Tuberculosis/prevention & control , COVID-19/transmission , Coronavirus Infections/prevention & control , Coronavirus Infections/transmission , Humans , Influenza, Human/transmission , Picornaviridae Infections/transmission , Respiratory Protective Devices , Respiratory Tract Infections/transmission , SARS-CoV-2 , Tuberculosis/transmission
13.
Expert Rev Vaccines ; 19(7): 599-610, 2020 07.
Article in English | MEDLINE | ID: mdl-32609047

ABSTRACT

INTRODUCTION: Several Picornaviruses are pathogens that generate serious problems for human and animal health worldwide. Vaccination is an attractive approach to fight against picornaviruses. In this regard, the development of low-cost vaccines is a priority to ensure coverage; especially in developing and low-income countries. In this context, plant-made vaccines are a convenient technology since plant cells are low-cost bioreactors capable of producing complex antigens that preserve their antigenic determinants; moreover, they can serve as biocapsules to achieve oral delivery. AREAS COVERED: In the present review the advances in the development of plant-made vaccines against picornaviruses are summarized and placed in perspective. The main diseases that have been targeted using this approach include Poliovirus, Food and mouth disease virus, Hepatitis A virus, and Enterovirus 71. EXPERT OPINION: Several vaccine candidates against picornavirus have been characterized at the preclinical level; with many of them capable of inducing humoral and cellular responses that led to neutralization of pathogens when evaluated in vitro and test animal challenge assays. Plant-made vaccines are a promise to fight picornaviruses; especially in the developing world where limited resources hamper vaccination coverage. A critical analysis of the road ahead for this technology is provided.


Subject(s)
Antigens, Plant/immunology , Picornaviridae Infections/prevention & control , Viral Vaccines/administration & dosage , Animals , Developing Countries , Humans , Picornaviridae Infections/immunology , Plants/immunology , Vaccination , Viral Vaccines/economics , Viral Vaccines/immunology
14.
Vaccine ; 38(30): 4695-4703, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32446833

ABSTRACT

Duck hepatitis A virus type 3 (DHAV-3) is an important pathogen that causes substantial losses in the Chinese duck industry. DHAV-3 is highly fatal to ducklings and there is no licensed vaccine in China available to reduce DHAV-3 infection. Our goal was to develop a live attenuated vaccine candidate against DHAV-3. A field isolated strain, SD, was attenuated by serially passaging in specific-pathogen-free (SPF) chicken embryos, and it lost its pathogenicity after 40 passages. The 70th passaged strain (SD70), which achieved good growth capacity in chicken embryos with a viral titer of 107.5 ELD50/mL, was chosen to be the live attenuated vaccine candidate. The SD70 strain did not cause clinical signs of disease or mortality in 1-day-old ducklings and showed no virulence reversion after seven rounds of in vivo back passages. The minimum effective dose of SD70 was determined to be 102.5 ELD50 via the vaccination route of subcutaneous inoculation. A single dose of the SD70 provided good protection to susceptible ducklings against the lethal DHAV-3 strain. Compared with the genomic sequence of the parent SD strain, the SD70 had 12 amino acid substitutions, some of which may play a role in virulence attenuation. This study demonstrated that the attenuated SD70 strain is a promising vaccine candidate for the prevention of DHAV-3 infection in China. It exhibited safety, good stability and excellent protection.


Subject(s)
Hepatitis Virus, Duck , Hepatitis, Viral, Animal , Picornaviridae Infections , Poultry Diseases , Animals , Chick Embryo , China , Ducks , Hepatitis, Viral, Animal/prevention & control , Picornaviridae Infections/prevention & control , Picornaviridae Infections/veterinary , Poultry Diseases/prevention & control , Vaccines, Attenuated
15.
Pediatr Infect Dis J ; 39(6): 507-511, 2020 06.
Article in English | MEDLINE | ID: mdl-32118855

ABSTRACT

BACKGROUND: Human parechovirus (HPeV) typically infects young children, and although infection is often asymptomatic, some types (eg, HPeV3) are associated with severe clinical manifestations, including central nervous system infection or sepsis-like syndrome, particularly affecting young infants. The third documented national epidemic of HPeV occurred in Australia in 2017-2018. METHODS: Four public laboratories that perform almost all of the HPeV PCR testing in New South Wales provided data regarding HPeV tests performed from July 1, 2017 to June 30, 2018. Limited demographic and clinical data were obtained from electronic medical records for laboratory test-positive cases that presented to each of the 3 pediatric hospitals in New South Wales. RESULTS: Five hundred eighty-one HPeV-positive samples obtained from 395 cases were included in the analysis. The peak of the outbreak occurred in late November 2017 (approximately 35 new cases each week), with the main HPeV epidemic occurring between the spring and summer months of September 2017 to January 2018; although this seasonality was observed primarily in infants less than 12 months of age. Among the 388 pediatric cases, almost half were younger than 2 months (188; 47%) and only 10 were children older than 2 years. The annualized estimated incidence of laboratory confirmed HPeV infection in children was approximately 142.4 cases per 100,000 children younger than 5 years in New South Wales during the epidemic season. CONCLUSIONS: The large burden of HPeV infection and disease identified in young infants in this and previous Australian studies highlight the need for more comprehensive national surveillance of HPeV infections and improved prevention strategies.


Subject(s)
Cost of Illness , Epidemics/statistics & numerical data , Parechovirus/pathogenicity , Picornaviridae Infections/epidemiology , Child, Preschool , Electronic Health Records , Epidemics/prevention & control , Female , Genotype , Hospitalization/statistics & numerical data , Hospitals, Pediatric/statistics & numerical data , Humans , Incidence , Infant , Infant, Newborn , Male , New South Wales/epidemiology , Parechovirus/genetics , Picornaviridae Infections/prevention & control , Seasons , Sequence Analysis, DNA
16.
Am J Epidemiol ; 189(3): 250-260, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31673696

ABSTRACT

The test-negative design is validated in outpatient, but not inpatient, studies of influenza vaccine effectiveness. The prevalence of chronic pulmonary disease among inpatients can lead to nonrepresentative controls. Test-negative design estimates are biased if vaccine administration is associated with incidence of noninfluenza viruses. We evaluated whether control group selection and effects of vaccination on noninfluenza viruses biased vaccine effectiveness in our study. Subjects were enrolled at the University of Michigan and Henry Ford hospitals during the 2014-2015 and 2015-2016 influenza seasons. Patients presenting with acute respiratory infection were enrolled and tested for respiratory viruses. Vaccine effectiveness was estimated using 3 control groups: negative for influenza, positive for other respiratory virus, and pan-negative individuals; it was also estimated for other common respiratory viruses. In 2014-2015, vaccine effectiveness was 41.1% (95% CI: 1.7, 64.7) using influenza-negative controls, 24.5% (95% CI: -42.6, 60.1) using controls positive for other virus, and 45.8% (95% CI: 5.7, 68.9) using pan-negative controls. In 2015-2016, vaccine effectiveness was 68.7% (95% CI: 44.6, 82.5) using influenza-negative controls, 63.1% (95% CI: 25.0, 82.2) using controls positive for other virus, and 71.1% (95% CI: 46.2, 84.8) using pan-negative controls. Vaccination did not alter odds of other respiratory viruses. Results support use of the test-negative design among inpatients.


Subject(s)
Influenza Vaccines , Influenza, Human/prevention & control , Picornaviridae Infections/prevention & control , Respiratory Syncytial Virus Infections/prevention & control , Adult , Aged , Bias , Case-Control Studies , Chronic Disease , Female , Hospitalization , Humans , Incidence , Influenza, Human/epidemiology , Inpatients , Male , Michigan/epidemiology , Middle Aged , Picornaviridae Infections/epidemiology , Respiratory Syncytial Virus Infections/epidemiology
18.
Front Immunol ; 10: 2660, 2019.
Article in English | MEDLINE | ID: mdl-31849928

ABSTRACT

Senecavirus A (SVA) is an emerging picornavirus causing vesicular disease (VD) clinically indistinguishable from foot-and-mouth disease (FMD) in pigs. Currently there are no vaccines currently available for SVA. Here we developed a recombinant SVA strain (rSVAm SacII) using reverse genetics and assessed its immunogenicity and protective efficacy in pigs. In vivo characterization of the rSVAm SacII strain demonstrated that the virus is attenuated, as evidenced by absence of lesions, decreased viremia and virus shedding in inoculated animals. Notably, while attenuated, rSVA mSacII virus retained its immunogenicity as high neutralizing antibody (NA) responses were detected in inoculated animals. To assess the immunogenicity and protective efficacy of rSVA mSacII, 4-week-old piglets were sham-immunized or immunized with inactivated or live rSVA mSacII virus-based formulations. A single immunization with live rSVA mSacII virus via the intramuscular (IM) and intranasal (IN) routes resulted in robust NA responses with antibodies being detected between days 3-7 pi. Neutralizing antibody responses in animals immunized with the inactivated virus via the IM route were delayed and only detected after a booster on day 21 pi. Immunization with live virus resulted in recall T cell proliferation (CD4+, CD8+, and CD4+/CD8+ T cells), demonstrating efficient stimulation of cellular immunity. Notably, a single dose of the live attenuated vaccine candidate resulted in protection against heterologous SVA challenge, as demonstrated by absence of overt disease and reduced viremia, virus shedding and viral load in tissues. The live attenuated vaccine candidate developed here represents a promising alternative to prevent and control SVA in swine.


Subject(s)
Picornaviridae Infections/veterinary , Picornaviridae/immunology , Swine Diseases/prevention & control , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Immunization , Picornaviridae Infections/prevention & control , Swine , T-Lymphocytes/immunology , Vaccines, Attenuated/immunology , Vaccines, Synthetic/immunology
19.
Article in English | MEDLINE | ID: mdl-31482072

ABSTRACT

Picornaviruses constitute one of the most relevant viral groups according to their impact on human and animal health. Etiologic agents of a broad spectrum of illnesses with a clinical presentation that ranges from asymptomatic to fatal disease, they have been the cause of uncountable epidemics throughout history. Picornaviruses are small naked RNA-positive single-stranded viruses that include some of the most important pillars in the development of virology, comprising poliovirus, rhinovirus, and hepatitis A virus. Picornavirus infectious particles use the fecal-oral or respiratory routes as primary modes of transmission. In this regard, successful viral spread relies on the capability of viral capsids to (i) shelter the viral genome, (ii) display molecular determinants for cell receptor recognition, (iii) facilitate efficient genome delivery, and (iv) escape from the immune system. Importantly, picornaviruses display a substantial amount of genetic variability driven by both mutation and recombination. Therefore, the outcome of their replication results in the emergence of a genetically diverse cloud of individuals presenting phenotypic variance. The host humoral response against the capsid protein represents the most active immune pressure and primary weapon to control the infection. Since the preservation of the capsid function is deeply rooted in the virus evolutionary dynamics, here we review the current structural evidence focused on capsid antibody evasion mechanisms from that perspective.


Subject(s)
Antibodies, Viral/immunology , Biological Evolution , Capsid Proteins/immunology , Capsid/immunology , Host-Pathogen Interactions/immunology , Picornaviridae/immunology , Animals , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/genetics , Genetic Variation , Genome, Viral , Genomics , Humans , Picornaviridae/genetics , Picornaviridae Infections/immunology , Picornaviridae Infections/prevention & control , Picornaviridae Infections/virology , Receptors, Virus/metabolism , Recombination, Genetic , Structure-Activity Relationship , Viral Tropism , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...