Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 600
1.
BMC Infect Dis ; 24(1): 547, 2024 May 31.
Article En | MEDLINE | ID: mdl-38822241

Noroviruses are the second leading cause of death in children under the age of 5 years old. They are responsible for 200 million cases of diarrhoea and 50,000 deaths in children through the word, mainly in low-income countries. The objective of this review was to assess how the prevalence and genetic diversity of noroviruses have been affected by the introduction of rotavirus vaccines in Africa. PubMed, Web of Science and Science Direct databases were searched for articles. All included studies were conducted in Africa in children aged 0 to 5 years old with gastroenteritis. STATA version 16.0 software was used to perform the meta-analysis. The method of Dersimonian and Laird, based on the random effects model, was used for the statistical analyses in order to estimate the pooled prevalence's at a 95% confidence interval (CI). Heterogeneity was assessed by Cochran's Q test using the I2 index. The funnel plot was used to assess study publication bias. A total of 521 studies were retrieved from the databases, and 19 were included in the meta-analysis. The pooled norovirus prevalence's for pre- and post-vaccination rotavirus studies were 15% (95 CI, 15-18) and 13% (95 CI, 09-17) respectively. GII was the predominant genogroup, with prevalence of 87.64% and 91.20% respectively for the pre- and post-vaccination studies. GII.4 was the most frequently detected genotype, with rates of 66.84% and 51.24% respectively for the pre- and post-vaccination studies. This meta-analysis indicates that rotavirus vaccination has not resulted in a decrease in norovirus infections in Africa.


Caliciviridae Infections , Gastroenteritis , Genetic Variation , Norovirus , Rotavirus Infections , Rotavirus Vaccines , Humans , Rotavirus Vaccines/immunology , Rotavirus Vaccines/administration & dosage , Infant , Africa/epidemiology , Child, Preschool , Caliciviridae Infections/epidemiology , Caliciviridae Infections/prevention & control , Caliciviridae Infections/virology , Norovirus/genetics , Norovirus/classification , Norovirus/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/epidemiology , Rotavirus Infections/virology , Gastroenteritis/virology , Gastroenteritis/epidemiology , Gastroenteritis/prevention & control , Infant, Newborn , Prevalence , Rotavirus/genetics , Rotavirus/immunology , Rotavirus/classification , Vaccination/statistics & numerical data
2.
Viruses ; 16(4)2024 03 30.
Article En | MEDLINE | ID: mdl-38675881

Rabbit hemorrhagic disease virus 2 (RHDV2) emerged in the United States in 2018 and has spread in both domestic and wild rabbits nationwide. The virus has a high mortality rate and can spread rapidly once introduced in a rabbit population. Vaccination against RHDV2 provides the best protection against disease and should be considered by all rabbit owners. Here, we investigate the duration of immunity provided by vaccination with the Medgene Platform conditionally licensed commercial vaccine 6 months following the initial series. Rabbits received either the vaccination or a placebo and were challenged with RHDV2 6 months later. All vaccinated rabbits survived challenge whereas 18/19 non-vaccinated controls succumbed to infection within 10 or fewer days post-challenge. These results demonstrate lasting immunity following vaccination with the Medgene RHDV2 vaccine.


Baculoviridae , Caliciviridae Infections , Hemorrhagic Disease Virus, Rabbit , Vaccination , Vaccines, Synthetic , Viral Vaccines , Animals , Hemorrhagic Disease Virus, Rabbit/immunology , Hemorrhagic Disease Virus, Rabbit/genetics , Rabbits , Caliciviridae Infections/prevention & control , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Caliciviridae Infections/veterinary , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Baculoviridae/genetics , Baculoviridae/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology
3.
J Virol ; 97(10): e0093823, 2023 10 31.
Article En | MEDLINE | ID: mdl-37792003

IMPORTANCE: Human norovirus (HuNoV) is highly infectious and can result in severe illnesses in the elderly and children. So far, there is no effective antiviral drug to treat HuNoV infection, and thus, the development of HuNoV vaccines is urgent. However, NoV evolves rapidly, and currently, at least 10 genogroups with numerous genotypes have been found. The genetic diversity of NoV and the lack of cross-protection between different genotypes pose challenges to the development of broadly protective vaccines. In this study, guided by structural alignment between GI.1 and GII.4 HuNoV VP1 proteins, several chimeric-type virus-like particles (VLPs) were designed through surface-exposed loop grafting. Mouse immunization studies show that two of the designed chimeric VLPs induced cross-immunity against both GI.1 and GII.4 HuNoVs. To our knowledge, this is the first designed chimeric VLPs that can induce cross-immune activities across different genogroups of HuNoV, which provides valuable strategies for the development of cross-reactive HuNoV vaccines.


Caliciviridae Infections , Epitopes , Genotype , Norovirus , Viral Vaccines , Virion , Animals , Humans , Mice , Caliciviridae Infections/immunology , Caliciviridae Infections/prevention & control , Caliciviridae Infections/virology , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Immunization , Norovirus/chemistry , Norovirus/classification , Norovirus/genetics , Norovirus/immunology , Viral Vaccines/chemistry , Viral Vaccines/genetics , Viral Vaccines/immunology , Chimera/genetics , Chimera/immunology , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Virion/chemistry , Virion/genetics , Virion/immunology
4.
J Am Assoc Lab Anim Sci ; 62(4): 327-335, 2023 Jul 05.
Article En | MEDLINE | ID: mdl-37758477

Hypochlorous acid (HOCl), used as a liquid or a fog, has broad antimicrobial and deodorizing effects. Our facility was the first in Taiwan that was built with a system to supply stabilized, biosafe HOCl solution (50 ppm available chlorine concentration, pH 6) into a new animal barrier facility that housed genetically modified mice. The HOCl system creates an extremely clean environment that allows us to raise mice in static, filter-top cages and to handle them on open tables without the need for biologic safety cabinets (BSC). Our animal facility (AF) sometimes receives mice from outside sources that are infected with pathogens, notably murine norovirus (MNV), Helicobacter spp., and trichomonads. We found that our standard operation procedure (SOP) prevented cross-contamination to other mice, including those in adjacent cages. After the removal of infected mice from a room, the remaining mice remained uninfected, without the need for extensive environmental decontamination. Learning this allowed us to use a test-and-removal method to eliminate pathogens. In addition, infected mouse strains that were not commercially available were rederived by using cross-fostering. After finding unexpected infections, we were able to identify all infected mice by widespread screening. We then removed contaminated cages and performed cross-fostering as needed. This approach was able to successfully eliminate murine norovirus, Helicobacter spp., and trichomonads. Over the 12 y in which we managed this AF, we refined our husbandry methods and our approach to the detection and eradication of pathogens by using HOCl fog and solution, the test-and-removal, and cross-fostering.


Caliciviridae Infections , Norovirus , Animals , Mice , Hypochlorous Acid , Caliciviridae Infections/prevention & control , Caliciviridae Infections/veterinary , Housing, Animal , Animal Husbandry/methods
7.
Vaccine ; 41(3): 766-777, 2023 01 16.
Article En | MEDLINE | ID: mdl-36528444

Noroviruses (NoVs) are one of the major causes of acute viral gastroenteritis in humans. Virus-like particles (VLPs) without genomes that mimic the capsid structure of viruses are promising vaccine candidates for the prevention of NoVs infection. To produce large amounts of recombinant protein, including VLPs, the silkworm-expression vector system (silkworm-BEVS) is an efficient and powerful tool. In this study, we constructed a recombinant baculovirus that expresses VP1 protein, the major structural protein of NoV GII.4. Expression analysis showed that the baculovirus-infected silkworm pupae expressed NoV VP1 protein more efficiently than silkworm larval fat bodies. We obtained about 4.9 mg of purified NoV VP1 protein from only five silkworm pupae. The purified VP1 protein was confirmed by dynamic light scattering and electron microscopy to form VLPs of approximately 40 nm in diameter. Antisera from mice immunized with the antigen blocked NoV VLPs binding to histo-blood group antigens of pig gastric mucin and also blocked NoV infection in intestinal epithelial cells derived from human induced pluripotent stem (iPS) cells. Our findings demonstrated that NoV VLP eliciting protective antibodies could be obtained in milligram quantities from a few silkworm pupae using the silkworm-BEVS.


Artificial Virus-Like Particles , Bombyx , Caliciviridae Infections , Gastroenteritis , Norovirus , Animals , Humans , Mice , Antibodies , Antibodies, Viral , Bombyx/chemistry , Bombyx/metabolism , Caliciviridae Infections/prevention & control , Capsid Proteins/genetics , Norovirus/genetics , Norovirus/immunology , Pupa , Swine , Artificial Virus-Like Particles/immunology
8.
Viruses ; 14(12)2022 12 16.
Article En | MEDLINE | ID: mdl-36560815

Norovirus (NoV) is an enteric non-enveloped virus which is the leading cause of gastroenteritis across all age groups. It is responsible for around 200,000 deaths annually and outbreaks are common in small communities such as educational and care facilities. 40% of all NoV outbreaks occur in long-term and acute-care facilities, forming the majority of outbreaks. Nosocomial settings set ideal environments for ease of transmission, especially due to the presence of immunocompromised groups. It is estimated to cost global economies around £48 billion a year, making it a global issue. NoV is transmitted via the faecal-oral route and infection with it results in asymptomatic cases or gastrointestinal disease. It has high mutational rates and this allows for new variants to emerge and be more resistant. The classification system available divides NoV into 10 genogroups and 49 genotypes based on whole amino acid sequencing of VP1 capsid protein and partial sequencing of RdRp, respectively. The most predominant genotypes which cause gastroenteritis in humans include GI.1 and GII.4, where GII.4 is responsible for more extreme clinical implications such as hospitalisation. In addition, GII.4 has been responsible for 6 pandemic strains, the last of which is the GII.4 Sydney (2012) variant. In recent years, the successful cultivation of HuNoV was reported in stem cell-derived human intestinal enteroids (HIEs), which promises to assist in giving a deeper understanding of its underlying mechanisms of infection and the development of more personalized control measures. There are no specific control measures against NoV, therefore common practices are used against it such as hand washing. No vaccine is available, but the HIL-214 candidate passed clinical phase 2b and shows promise.


Caliciviridae Infections , Gastroenteritis , Norovirus , Humans , Caliciviridae Infections/epidemiology , Caliciviridae Infections/prevention & control , Disease Outbreaks , Gastroenteritis/epidemiology , Gastroenteritis/prevention & control , Genotype , Norovirus/genetics , Phylogeny , Clinical Trials, Phase II as Topic
9.
Am J Vet Res ; 83(12)2022 Nov 07.
Article En | MEDLINE | ID: mdl-36327167

OBJECTIVE: To evaluate efficacy of a novel vaccine against rabbit hemorrhagic disease virus 2 (RHDV2) in domestic rabbits. ANIMALS: 40 New Zealand White rabbits obtained from a commercial breeder. PROCEDURES: Rabbits were vaccinated and held at the production facility for the duration of the vaccination phase and transferred to Colorado State University for challenge with RHDV2. Rabbits were challenged with oral suspensions containing infectious virus and monitored for clinical disease for up to 10 days. Rabbits that died or were euthanized following infection were necropsied, and livers were evaluated for viral RNA via RT-PCR. RESULTS: None of the vaccinated animals (0/9) exhibited clinical disease or mortality following infection with RHDV2 while 9/13 (69%) of the control animals succumbed to lethal disease following infection. CLINICAL RELEVANCE: The novel vaccine described herein provided complete protection against lethal infection following RHDV2 challenge. Outside of emergency use, there are currently no licensed vaccines against RHDV2 on the market in the United States; as such, this vaccine candidate would provide an option for control of this disease now that RHDV2 has become established in North America.


Caliciviridae Infections , Hemorrhagic Disease Virus, Rabbit , Vaccines , Rabbits , Animals , Hemorrhagic Disease Virus, Rabbit/genetics , Caliciviridae Infections/prevention & control , Caliciviridae Infections/veterinary , Vaccination/veterinary
10.
Viruses ; 14(11)2022 10 27.
Article En | MEDLINE | ID: mdl-36366469

BACKGROUND: Human norovirus (HuNoV) is the leading viral cause of diarrhea, with GII.4 as the predominant genotype of HuNoV outbreaks globally. However, new genogroup variants emerge periodically, complicating the development of anti-HuNoV vaccines; other prophylactic or therapeutic medications specifically for HuNoV disease are lacking. Passive immunization using oral anti-HuNoV antibodies may be a rational alternative. Here, we explore the feasibility of using avian immunoglobulins (IgY) for preventing HuNoV infection in vitro in a human intestinal enteroid (HIE) model. METHODS: Hens were immunized with virus-like particles (VLP) of a GII.4 HuNoV strain (GII.4/CHDC2094/1974/US) by intramuscular injection. The resulting IgY was evaluated for inhibition of binding to histo-blood group antigens (HBGA) and viral neutralization against representative GII.4 and GII.6 clinical isolates, using an HIE model. RESULTS: IgY titers were detected by three weeks following initial immunization, persisting at levels of 1:221 (1:2,097,152) from 9 weeks to 23 weeks. Anti-HuNoV IgY significantly (p < 0.05) blocked VLP adhesion to HBGA up to 1:12,048 dilution (0.005 mg/mL), and significantly (p < 0.05) inhibited replication of HuNoV GII.4[P16] Sydney 2012 in HIEs up to 1:128 dilution (0.08 mg/mL). Neutralization was not detected against genotype GII.6. CONCLUSIONS: We demonstrate the feasibility of IgY for preventing infection of HIE by HuNoV GII.4. Clinical preparations should cover multiple circulating HuNoV genotypes for comprehensive effects. Plans for animal studies are underway.


Blood Group Antigens , Caliciviridae Infections , Norovirus , Humans , Animals , Female , Chickens , Feasibility Studies , Caliciviridae Infections/prevention & control , Caliciviridae Infections/veterinary , Norovirus/genetics , Blood Group Antigens/genetics , Antibodies
11.
J Virol ; 96(22): e0126222, 2022 11 23.
Article En | MEDLINE | ID: mdl-36314817

Rotavirus, a segmented double-stranded RNA virus of the Reoviridae family, is a primary cause of acute gastroenteritis in young children. In countries where rotavirus vaccines are widely used, norovirus (NoV) has emerged as the major cause of acute gastroenteritis. Towards the goal of creating a combined rotavirus-NoV vaccine, we explored the possibility of generating recombinant rotaviruses (rRVs) expressing all or portions of the NoV GII.4 VP1 capsid protein. This was accomplished by replacing the segment 7 NSP3 open reading frame with a cassette encoding, sequentially, NSP3, a 2A stop-restart translation element, and all or portions (P, P2) of NoV VP1. In addition to successfully recovering rRVs with modified SA11 segment 7 RNAs encoding NoV capsid proteins, analogous rRVs were recovered through modification of the segment 7 RNA of the RIX4414 vaccine strain. An immunoblot assay confirmed that rRVs expressed NoV capsid proteins as independent products. Moreover, VP1 expressed by rRVs underwent dimerization and was recognized by conformational-dependent anti-VP1 antibodies. Serially passaged rRVs that expressed the NoV P and P2 were genetically stable, retaining additional sequences of up to 1.1 kbp without change. However, serially passaged rRVs containing the longer 1.6-kb VP1 sequence were less stable and gave rise to virus populations with segment 7 RNAs lacking VP1 coding sequences. Together, these studies suggest that it may be possible to develop combined rotavirus-NoV vaccines using modified segment 7 RNA to express NoV P or P2. In contrast, development of potential rotavirus-NoV vaccines expressing NoV VP1 will need additional efforts to improve genetic stability. IMPORTANCE Rotavirus (RV) and norovirus (NoV) are the two most important causes of acute viral gastroenteritis (AGE) in infants and young children. While the incidence of RV AGE has been brought under control in many countries through the introduction of universal mass vaccination with live attenuated RV vaccines, similar highly effective NoV vaccines are not available. To pursue the development of a combined RV-NoV vaccine, we examined the potential of using RV as an expression vector of all or portions of the NoV capsid protein VP1. Our results showed that by replacing the NSP3 open reading frame in RV genome segment 7 RNA with a coding cassette for NSP3, a 2A stop-restart translation element, and VP1, recombinant RVs can be generated that express NoV capsid proteins. These findings raise the possibility of developing new generations of RV-based combination vaccines that provide protection against a second enteric pathogen, such as NoV.


Capsid Proteins , Gastroenteritis , Norovirus , Rotavirus , Viral Vaccines , Child , Child, Preschool , Humans , Capsid Proteins/genetics , Gastroenteritis/prevention & control , Gastroenteritis/virology , Norovirus/genetics , RNA , Rotavirus/genetics , Vaccines, Combined , Rotavirus Infections/prevention & control , Caliciviridae Infections/prevention & control
12.
J Pediatric Infect Dis Soc ; 11(10): 459-462, 2022 Oct 25.
Article En | MEDLINE | ID: mdl-35849117

A birth cohort design was used to understand whether heterotypic ligand-blocking norovirus antibodies provide cross-protection within the GII genogroup. We found that almost one-half of children who experienced a norovirus GII episode had preexisting antibodies heterotypic to the infecting genotype; therefore, these antibodies did not provide cross-protection.


Caliciviridae Infections , Gastroenteritis , Norovirus , Child , Humans , Infant , Child, Preschool , Norovirus/genetics , Caliciviridae Infections/prevention & control , Gastroenteritis/prevention & control , Ligands , Genotype , Feces
13.
Epidemiology ; 33(5): 650-653, 2022 09 01.
Article En | MEDLINE | ID: mdl-35700200

BACKGROUND: Norovirus and sapovirus cause a large burden of acute gastroenteritis (AGE) in young children. We assessed protection conferred by norovirus and sapovirus AGE episodes against future episodes. METHODS: Between June 2017 and July 2018, we recruited 444 newborns in León, Nicaragua. Weekly household surveys identified AGE episodes over 36 months, and AGE stools were tested by reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) for norovirus genogroup (G)I/GII and sapovirus. We used recurrent-event Cox models and negative control methods to estimate protection conferred by first episodes, controlling for observed and unobserved risk factors, respectively. RESULTS: Sapovirus episodes conferred a 69% reduced hazard of subsequent episodes using the negative control method. Norovirus GI (hazard ratio [HR] = 0.67; 95% confidence interval [CI] = 0.31, 1.3) and GII (HR = 0.20; 95% CI = 0.04, 0.44) episodes also appeared highly protective. Protection against norovirus GII was enhanced following two episodes. CONCLUSIONS: Evidence of natural immunity in early childhood provides optimism for the future success of pediatric norovirus and sapovirus vaccines.


Caliciviridae Infections , Gastroenteritis , Norovirus , Sapovirus , Birth Cohort , Caliciviridae Infections/epidemiology , Caliciviridae Infections/prevention & control , Caliciviridae Infections/virology , Child, Preschool , Gastroenteritis/epidemiology , Gastroenteritis/prevention & control , Gastroenteritis/virology , Genotype , Humans , Infant , Infant, Newborn , Norovirus/genetics , Sapovirus/genetics
14.
Viruses ; 14(5)2022 04 29.
Article En | MEDLINE | ID: mdl-35632680

Feline calicivirus (FCV) is a common pathogen in domestic cats that is highly contagious, resistant to many disinfectants and demonstrates a high genetic variability. FCV infection can lead to serious or even fatal diseases. In this review, the European Advisory Board on Cat Diseases (ABCD), a scientifically independent board of experts in feline medicine from 11 European countries, presents the current knowledge of FCV infection and fills gaps with expert opinions. FCV infections are particularly problematic in multicat environments. FCV-infected cats often show painful erosions in the mouth and mild upper respiratory disease and, particularly in kittens, even fatal pneumonia. However, infection can be associated with chronic gingivostomatitis. Rarely, highly virulent FCV variants can induce severe systemic disease with epizootic spread and high mortality. FCV can best be detected by reverse-transcriptase PCR. However, a negative result does not rule out FCV infection and healthy cats can test positive. All cats should be vaccinated against FCV (core vaccine); however, vaccination protects cats from disease but not from infection. Considering the high variability of FCV, changing to different vaccine strain(s) may be of benefit if disease occurs in fully vaccinated cats. Infection-induced immunity is not life-long and does not protect against all strains; therefore, vaccination of cats that have recovered from caliciviral disease is recommended.


Caliciviridae Infections , Calicivirus, Feline , Animals , Caliciviridae Infections/prevention & control , Caliciviridae Infections/veterinary , Cats , Europe , Female , Vaccination
15.
Vaccine ; 40(26): 3588-3596, 2022 06 09.
Article En | MEDLINE | ID: mdl-35595661

BACKGROUND: Young children can suffer severe consequences of norovirus gastroenteritis. We performed a dose-finding study of a bivalent virus-like particle (VLP) vaccine candidate (TAK-214) in healthy 1-8-year-old children. METHODS: In this phase 2 study two age cohorts (1-3 and 4-8 years of age inclusive, N = 120 per cohort) of children enrolled from Finland, Panama and Colombia were initially randomized 1:1:1:1 to four groups which were further split into two equal subgroups, to receive one or two intramuscular doses of four TAK-214 formulations containing 15/15, 15/50, 50/50 or 50/150 µg of GI.1/GII.4c genotype VLPs and 0.5 mg Al(OH)3 at 28 days interval. ELISA Pan-Ig and histoblood group antigen-blocking (HBGA) antibodies against each VLP were measured on days 1, 29, 57 and 210. Parents/guardians recorded solicited local and systemic adverse events (AE) and any unsolicited or serious AEs (SAE). RESULTS: All formulations were well-tolerated across both age cohorts and dosage groups with no vaccine-related SAEs reported. Solicited AEs were mostly mild-to-moderate, resolved quickly, and did not increase after the second dose. Pan-Ig and HBGA responses induced after one dose were only slightly increased by the second dose. Across dose groups at Day 29 after one dose GI.1 Pan Ig seroresponse rates (SRR) were 82-97% and 81-96% and GII.4c SRR were 79-97% and 80-91% in 1-3 and 4-8 year-olds, respectively. Respective rates were to 92-93% and 73-92% for GI.1, and 77-100% and 62-83% for GII.4c at Day 57 following two doses. HBGA responses had similar profiles. Both Pan Ig and HBGA geometric mean titers persisted above baseline up to Day 210. CONCLUSIONS: All dosages of TAK-214 displayed acceptable reactogenicity in 1-8-year-old children and induced robust, durable immune responses after one dose which are further increased after two doses.


Caliciviridae Infections , Norovirus , Vaccines, Virus-Like Particle , Viral Vaccines , Antibodies, Viral , Caliciviridae Infections/prevention & control , Child , Child, Preschool , Double-Blind Method , Humans , Immunogenicity, Vaccine , Infant
16.
Schweiz Arch Tierheilkd ; 164(3): 225-241, 2022 Mar.
Article En | MEDLINE | ID: mdl-35232714

INTRODUCTION: Feline Calicivirus (FCV) is one of the most common viral pathogens in domestic cats worldwide. The first report of FCV dates back to 1957, when FCV was isolated from the gastrointestinal tract of cats in New Zealand. Subsequent reports recognised FCV as a cause of respiratory disease in cats, and at present, feline practitioners worldwide are daily confronted with cats suffering from suspected FCV. The highly mutagenic nature of FCV and its high genetic plasticity enable the virus to successfully survive in the feline population, and pose a special challenge as regards the diagnosis, treatment, and prevention of FCV-induced disease. Upper respiratory tract disease has been considered a common clinical sign of FCV infection. A study from Switzerland demonstrated that oral ulcerations, salivation and gingivitis-stomatitis were more commonly associated with FCV infection than upper respiratory tract disease, and less than half of the cats suspected to have FCV infection were found to be FCV-positive. Furthermore, a study investigating FCV isolates from Switzerland found some evidence that the genetic background of cats might influence their susceptibility to FCV infection. This review article provides a comprehensive summary of the FCV literature, and integrates the results of recent research on FCV's genetic characteristics, the cellular and humoral immunity evoked by FCV vaccination and infection, the diagnosis of FCV, FCV prevention/vaccination, the risk factors associated with FCV, and the hygienic measures necessary in FCV-contaminated areas. After each section, the key points are summarised, and relevant information is outlined to help feline practitioners in FCV diagnosis, treatment and prevention.


INTRODUCTION: Le calicivirus félin (FCV) est l'un des agents pathogènes viraux les plus courants chez les chats domestiques dans le monde. Le premier signalement de FCV remonte à 1957, lorsque le FCV a été isolé du tractus gastro-intestinal de chats en Nouvelle-Zélande. Des rapports ultérieurs ont reconnu le FCV comme une cause de maladie respiratoire chez les chats et, à l'heure actuelle, les praticiens félins du monde entier sont quotidiennement confrontés à des chats suspectés de FCV. La nature hautement mutagène du FCV et sa haute plasticité génétique permettent au virus de survivre avec succès dans la population féline et posent un défi particulier en ce qui concerne le diagnostic, le traitement et la prévention de la maladie induite par le FCV. La maladie des voies respiratoires supérieures a été considérée comme un signe clinique courant d'infection par le FCV. Une étude réalisée en Suisse a démontré que les ulcérations buccales, la salivation et la gingivite-stomatite étaient plus fréquemment associées à une infection à FCV qu'à une autre maladie des voies respiratoires supérieures et moins de la moitié des chats suspectés d'avoir une infection à FCV se sont avérés positifs pour le FCV. De plus, une étude portant sur des isolats de FCV en Suisse a trouvé des preuves que le profil génétique des chats pourrait influencer leur sensibilité à l'infection par le FCV. Cet article de synthèse fournit un résumé complet de la littérature sur le FCV et intègre les résultats de recherches récentes sur les caractéristiques génétiques du FCV, l'immunité cellulaire et humorale évoquée par la vaccination et l'infection au FCV, le diagnostic du FCV, la prévention/vaccination contre le FCV, les facteurs de risque associés avec le FCV et les mesures d'hygiène nécessaires dans les zones contaminées par le FCV. Après chaque section, les points clés sont résumés et des informations pertinentes sont décrites pour aider les praticiens félins dans le diagnostic, le traitement et la prévention du FCV.


Caliciviridae Infections , Calicivirus, Feline , Cat Diseases , Animals , Caliciviridae Infections/diagnosis , Caliciviridae Infections/prevention & control , Caliciviridae Infections/veterinary , Calicivirus, Feline/genetics , Cat Diseases/diagnosis , Cat Diseases/prevention & control , Cats , Switzerland/epidemiology , Vaccination/veterinary
17.
Article En | MEDLINE | ID: mdl-35162592

Human noroviruses (HuNoVs) are a leading cause of acute gastroenteritis among children in China. However, little is known about parents' knowledge of HuNoV infection and their understanding of how to prevent and control the disease. Therefore, we performed an exploratory survey to assess the level of knowledge of HuNoV infection among kindergarten parents. A cross-sectional survey was conducted by investigating kindergarteners' parents through an online self-administered questionnaire between October 2020 and November 2020 in Chengdu, China. A total of 771 questionnaires were received with valid responses, and 81.97% of respondents had heard about NoV before. Among parents who had heard about HuNoV before, they had a poor awareness of incubation period, duration, and high-incidence seasons of HuNoV infection. The respondents also had a low-level awareness of how to clean the places contaminated by vomitus or stool. The multiple-regression analysis confirmed that factors associated with good knowledge regarding HuNoV infection were level of education, occupation, history of infection, and HuNoV learning experience. The most expected approach to learn about HuNoV among parents was the internet, followed by knowledge training in kindergartens, community information, and television. This is the first study to assess kindergarten parents' knowledge and awareness of HuNoV infection. The survey results provide insights that would help in developing effective strategies and educational materials to prevent and control the disease.


Caliciviridae Infections , Gastroenteritis , Norovirus , Caliciviridae Infections/prevention & control , Child , Cross-Sectional Studies , Educational Status , Humans
18.
Emerg Infect Dis ; 28(3): 713-716, 2022 Mar.
Article En | MEDLINE | ID: mdl-35049493

A rapid decrease in viral gastroenteritis during winter 2019-20 and a return of norovirus and rotavirus activity during winter 2020-21 were observed while multiple nonpharmaceutical interventions for coronavirus disease were in effect in Hong Kong. The initial collateral benefit of coronavirus disease countermeasures that reduced the viral gastroenteritis burden is not sustainable.


COVID-19 , Caliciviridae Infections , Norovirus , Rotavirus Infections , Rotavirus , Caliciviridae Infections/epidemiology , Caliciviridae Infections/prevention & control , China/epidemiology , Feces , Humans , Infant , Norovirus/genetics , Rotavirus/genetics , Rotavirus Infections/epidemiology , Rotavirus Infections/prevention & control , SARS-CoV-2
19.
Expert Rev Anti Infect Ther ; 20(2): 279-290, 2022 Feb.
Article En | MEDLINE | ID: mdl-34225537

BACKGROUND: Although most norovirus outbreaks in high-income countries occur in healthcare facilities, information on associations between control measures and outbreak outcomes in these settings is lacking. METHODS: We conducted a systematic review/meta-analysis to assess associations between norovirus outbreak control measures and outcomes in hospitals and long-term care facilities (LTCFs), globally. Using regression analyses stratified by setting (hospital/LTCF), we compared durations, attack rates, and case counts for outbreaks in which control measures were reportedly implemented to those in which they were not. RESULTS: We identified 102 papers describing 162 norovirus outbreaks. Control measures were reportedly implemented in 118 (73%) outbreaks and were associated with 0.6 (95% CI: 0.3-1.1) times smaller patient case counts and 0.7 (95% CI: 0.4, 1.0) times shorter durations in hospitals but 1.5 (95% CI: 1.1-2.2), 1.5 (95% CI: 1.0-2.1) and 1.6 (95% CI: 1.0-2.6) times larger overall, resident and staff case counts, respectively, and 1.4 (95% CI: 1.0-2.0) times longer durations in LTCFs. CONCLUSIONS: Reported implementation of control measures was associated with smaller/shorter outbreaks in hospitals but larger/longer outbreaks in LTCFs. Control measures were likely implemented in response to larger/longer outbreaks in LTCFs, rather than causing them. Prospective observational or intervention studies are needed to determine effectiveness.


Caliciviridae Infections , Cross Infection , Norovirus , Caliciviridae Infections/epidemiology , Caliciviridae Infections/prevention & control , Cross Infection/epidemiology , Cross Infection/prevention & control , Delivery of Health Care , Disease Outbreaks/prevention & control , Humans , Infection Control , Observational Studies as Topic
20.
J Biomol Struct Dyn ; 40(7): 3098-3109, 2022 04.
Article En | MEDLINE | ID: mdl-33170093

Human Norovirus belongs to a family Calciviridae, and was identified in the outbreak of gastroenteritis in Norwalk, due to its seasonal prevalence known as "winter vomiting disease." Treatment of Norovirus infection is still mysterious because there is no effective antiviral drugs or vaccine developed to protect against the infection, to eradicate the infection an effective vaccine should be developed. In this study, capsid protein (A7YK10), small protein (A7YK11), and polyprotein (A7YK09) were utilized. These proteins were subjected to B and T cell epitopes prediction by using reliable immunoinformatics tools. The antigenic and non-allergenic epitopes were selected for the subunit vaccine, which can activate cellular and humoral immune responses. Linkers joined these epitopes together. The vaccine structure was modelled and validated by using Errat, ProSA, and rampage servers. The modelled vaccine was docked with TLR-7. The stability of the docked complex was evaluated by MD simulation. To apply the concept in a wet lab, the reverse translated vaccine sequence was cloned in pET28a (+). The vaccine developed in this study requires experimental validation to ensure its effectiveness against the disease.Communicated by Ramaswamy H. Sarma.


Caliciviridae Infections , Norovirus , Caliciviridae Infections/prevention & control , Computational Biology , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Humans , Molecular Docking Simulation , Vaccines, Subunit , Vaccinology
...