Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.836
Filter
1.
Expert Rev Vaccines ; 23(1): 606-618, 2024.
Article in English | MEDLINE | ID: mdl-38813689

ABSTRACT

INTRODUCTION: Rotavirus is a leading cause of severe diarrheal disease and death in children under five years of age worldwide. Vaccination is one of the most important public health interventions to reduce this significant burden. AREAS COVERED: This literature review examined vaccination coverage, hospitalization rate, mortality, genotypic distribution, immunogenicity, cost-effectiveness, and risk versus benefit of rotavirus vaccination in children in South America. Nine out of twelve countries in South America currently include a rotavirus vaccine in their national immunization program with coverage rates in 2022 above 90%. EXPERT OPINION: Introduction of the rotavirus vaccination has led to a marked reduction in hospitalizations and deaths from diarrheal diseases in children under 5 years, particularly infants under 1 year, in several South American countries. In Brazil, hospitalizations decreased by 59% and deaths by 21% (30-38% in infants). In Peru, hospitalizations in infants fell by 46% and deaths by 37% (56% in infants). Overall, data suggest that rotavirus vaccination has reduced rotavirus deaths by 15-50% in various South American countries. There is some evidence that immunity wanes after the age of 1-year old. Ongoing surveillance of vaccine coverage and changes in morbidity and mortality is important to maximize protection against this disease.


Subject(s)
Diarrhea , Hospitalization , Immunization Programs , Rotavirus Infections , Rotavirus Vaccines , Humans , Rotavirus Vaccines/administration & dosage , Rotavirus Vaccines/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/epidemiology , Diarrhea/prevention & control , Diarrhea/epidemiology , Diarrhea/virology , Infant , Hospitalization/statistics & numerical data , South America/epidemiology , Child, Preschool , Vaccination/statistics & numerical data , Cost-Benefit Analysis , Rotavirus/immunology , Vaccination Coverage/statistics & numerical data , Cost of Illness
2.
J Med Virol ; 96(5): e29650, 2024 May.
Article in English | MEDLINE | ID: mdl-38727133

ABSTRACT

To analyze the epidemiological characteristics of group A rotavirus (RVA) diarrhea in Beijing between 2019 and 2022 and evaluate the effectiveness of the RV5 vaccine. Stool specimens were collected from patients with acute diarrhea, and RVA was detected and genotyped. The whole genome of RVA was sequenced by fragment amplification and Sanger sequencing. Phylogenetic trees were constructed using Bayesian and maximum likelihood methods. Descriptive epidemiological methods were used to analyze the characteristics of RVA diarrhea. Test-negative design was used to evaluate the vaccine effectiveness (VE) of the RV5. Compared with 2011-2018, RVA-positive rates in patients with acute diarrhea under 5 years of age and adults decreased significantly between 2019 and 2022, to 9.45% (249/634) and 3.66% (220/6016), respectively. The predominant genotype of RVA had changed from G9-VIP[8]-III between 2019 and 2021 to G8-VP[8]-III in 2022, and P[8] sequences from G8-VP[8]-III strains formed a new branch called P[8]-IIIb. The complete genotype of G8-VP[8]-III was G8-P[8]-I2-R2-C2-M2-A2-N2-T2-E2-H2. The VE of 3 doses of RV5 was 90.4% (95% CI: 28.8%-98.7%) against RVA diarrhea. The prevalence of RVA decreased in Beijing between 2019 and 2022, and the predominant genotype changed to G8P[8], which may be related to RV5 vaccination. Continuous surveillance is necessary to evaluate vaccine effectiveness and improve vaccine design.


Subject(s)
Diarrhea , Feces , Genotype , Phylogeny , Rotavirus Infections , Rotavirus Vaccines , Rotavirus , Humans , Rotavirus/genetics , Rotavirus/classification , Rotavirus/immunology , Rotavirus/isolation & purification , Rotavirus Infections/epidemiology , Rotavirus Infections/virology , Rotavirus Infections/prevention & control , Diarrhea/virology , Diarrhea/epidemiology , Rotavirus Vaccines/administration & dosage , Rotavirus Vaccines/immunology , Child, Preschool , Prevalence , Beijing/epidemiology , Male , Infant , Female , Adult , Feces/virology , Middle Aged , Child , Young Adult , Adolescent , Vaccine Efficacy , Aged , Genome, Viral , Infant, Newborn
3.
Rev Assoc Med Bras (1992) ; 70(4): e20230972, 2024.
Article in English | MEDLINE | ID: mdl-38716934

ABSTRACT

OBJECTIVE: Our objective was to determine the frequency of rotavirus, adenovirus, and rota-adenovirus co-infections and investigate the fecal leukocyte rate associated with these infections in patients with gastroenteritis. METHODS: This is a retrospective study. We identified patients who were admitted to the pediatric emergency department with acute gastroenteritis and had their stool samples tested for rotavirus and/or adenovirus antigens. Among them, we determined the individuals who underwent stool microscopy tests on the same day and recorded their results. RESULTS: A total of 1,577 patients who underwent testing for rotavirus and/or adenovirus antigens in their stool samples were identified. Among these patients, 583 individuals had concurrent fecal microscopy results. The prevalence of solely rotavirus antigen positivity was 16.4%, solely adenovirus antigen positivity was 2.9%, and rota-adenovirus co-infections were detected in 1.8% of the children. The fecal leukocyte rates in children infected with rotavirus, adenovirus, and rota-adenovirus co-infections were 4.8, 13.3, and 88.9%, respectively. CONCLUSION: The presence of fecal leukocytes was detected at a high rate in cases of viral gastroenteritis, especially in rota-adenovirus co-infections. Therefore, clinicians should not consider only bacterial pathogens in the presence of fecal leukocytes.


Subject(s)
Coinfection , Feces , Gastroenteritis , Rotavirus Infections , Humans , Gastroenteritis/virology , Gastroenteritis/epidemiology , Retrospective Studies , Feces/virology , Female , Male , Child, Preschool , Infant , Rotavirus Infections/epidemiology , Acute Disease , Coinfection/epidemiology , Child , Leukocyte Count , Adenovirus Infections, Human/epidemiology , Adenoviridae Infections/epidemiology , Leukocytes , Rotavirus/isolation & purification , Rotavirus/immunology , Adenoviridae/isolation & purification
4.
Viruses ; 16(5)2024 04 24.
Article in English | MEDLINE | ID: mdl-38793542

ABSTRACT

The suboptimal performance of rotavirus (RV) vaccines in developing countries and in animals necessitates further research on the development of novel therapeutics and control strategies. To initiate infection, RV interacts with cell-surface O-glycans, including histo-blood group antigens (HBGAs). We have previously demonstrated that certain non-pathogenic bacteria express HBGA- like substances (HBGA+) capable of binding RV particles in vitro. We hypothesized that HBGA+ bacteria can bind RV particles in the gut lumen protecting against RV species A (RVA), B (RVB), and C (RVC) infection in vivo. In this study, germ-free piglets were colonized with HBGA+ or HBGA- bacterial cocktail and infected with RVA/RVB/RVC of different genotypes. Diarrhea severity, virus shedding, immunoglobulin A (IgA) Ab titers, and cytokine levels were evaluated. Overall, colonization with HBGA+ bacteria resulted in reduced diarrhea severity and virus shedding compared to the HBGA- bacteria. Consistent with our hypothesis, the reduced severity of RV disease and infection was not associated with significant alterations in immune responses. Additionally, colonization with HBGA+ bacteria conferred beneficial effects irrespective of the piglet HBGA phenotype. These findings are the first experimental evidence that probiotic performance in vivo can be improved by including HBGA+ bacteria, providing decoy epitopes for broader/more consistent protection against diverse RVs.


Subject(s)
Blood Group Antigens , Germ-Free Life , Rotavirus Infections , Rotavirus , Animals , Rotavirus Infections/immunology , Rotavirus Infections/virology , Swine , Rotavirus/immunology , Blood Group Antigens/metabolism , Blood Group Antigens/immunology , Diarrhea/virology , Diarrhea/microbiology , Diarrhea/prevention & control , Swine Diseases/virology , Swine Diseases/microbiology , Swine Diseases/immunology , Virus Shedding , Bacteria/classification , Immunoglobulin A/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Cytokines/metabolism
5.
Int Immunopharmacol ; 133: 112079, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38615376

ABSTRACT

Porcine rotavirus (PoRV) poses a threat to the development of animal husbandry and human health, leading to substantial economic losses. VP6 protein is the most abundant component in virus particles and also the core structural protein of the virus. Firstly, this study developed an antibiotic-resistance-free, environmentally friendly expression vector, named asd-araC-PBAD-alr (AAPA). Then Recombinant Lactiplantibacillus plantarum (L. plantarum) strains induced by arabinose to express VP6 and VP6-pFc fusion proteins was constructed. Subsequently, This paper discovered that NC8/Δalr-pCXa-VP6-S and NC8/Δalr-pCXa-VP6-pFc-S could enhance host immunity and prevent rotavirus infection in neonatal mice and piglets. The novel recombinant L. plantarum strains constructed in this study can serve as oral vaccines to boost host immunity, offering a new strategy to prevent PoRV infection.


Subject(s)
Capsid Proteins , Lactobacillus plantarum , Swine Diseases , Animals , Swine , Lactobacillus plantarum/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Mice , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/microbiology , Swine Diseases/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/immunology , Rotavirus Infections/virology , Antigens, Viral/immunology , Rotavirus/immunology , Mice, Inbred BALB C , Animals, Newborn
6.
J Virol ; 98(5): e0021224, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38591886

ABSTRACT

Porcine rotaviruses (PoRVs) cause severe economic losses in the swine industry. P[7] and P[23] are the predominant genotypes circulating on farms, but no vaccine is yet available. Here, we developed a bivalent subunit PoRV vaccine using truncated versions (VP4*) of the VP4 proteins from P[7] and P[23]. The vaccination of mice with the bivalent subunit vaccine elicited more robust neutralizing antibodies (NAbs) and cellular immune responses than its components, even at high doses. The bivalent subunit vaccine and inactivated bivalent vaccine prepared from strains PoRVs G9P[7] and G9P[23] were used to examine their protective efficacy in sows and suckling piglets after passive immunization. The immunized sows showed significantly elevated NAbs in the serum and colostrum, and the suckling piglets acquired high levels of sIgA antibodies from the colostrum. Challenging subunit-vaccinated or inactivated-vaccinated piglets with homologous virulent strains did not induce diarrhea, except in one or two piglets, which had mild diarrhea. Immunization with the bivalent subunit vaccine and inactivated vaccine also alleviated the microscopic lesions in the intestinal tissues caused by the challenge with the corresponding homologous virulent strain. However, all the piglets in the challenged group displayed mild to watery diarrhea and high levels of viral shedding, whereas the feces and intestines of the piglets in the bivalent subunit vaccine and inactivated vaccine groups had lower viral loads. In summary, our data show for the first time that a bivalent subunit vaccine combining VP4*P[7] and VP4*P[23] effectively protects piglets against the diarrhea caused by homologous virulent strains.IMPORTANCEPoRVs are the main causes of diarrhea in piglets worldwide. The multisegmented genome of PoRVs allows the reassortment of VP4 and VP7 genes from different RV species and strains. The P[7] and P[23] are the predominant genotypes circulating in pig farms, but no vaccine is available at present in China. Subunit vaccines, as nonreplicating vaccines, are an option to cope with variable genotypes. Here, we have developed a bivalent subunit candidate vaccine based on a truncated VP4 protein, which induced robust humoral and cellular immune responses and protected piglets against challenge with homologous PoRV. It also appears to be safe. These data show that the truncated VP4-protein-based subunit vaccine is a promising candidate for the prevention of PoRV diarrhea.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Capsid Proteins , Rotavirus Infections , Rotavirus Vaccines , Rotavirus , Swine Diseases , Vaccines, Subunit , Animals , Swine , Rotavirus/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Swine Diseases/prevention & control , Swine Diseases/virology , Swine Diseases/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Rotavirus Infections/prevention & control , Rotavirus Infections/veterinary , Rotavirus Infections/immunology , Rotavirus Infections/virology , Rotavirus Vaccines/immunology , Rotavirus Vaccines/administration & dosage , Mice , Female , Mice, Inbred BALB C , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Diarrhea/prevention & control , Diarrhea/virology , Diarrhea/veterinary , Diarrhea/immunology , Genotype , Immunity, Cellular , Vaccination
7.
Vaccine ; 42(15): 3514-3521, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38670845

ABSTRACT

Group A rotavirus (RVA) is the primary etiological agent of acute gastroenteritis (AGE) in children under 5 years of age. Despite the global implementation of vaccines, rotavirus infections continue to cause over 120,000 deaths annually, with a majority occurring in developing nations. Among infants, the P[8] rotavirus strain is the most prevalent and can be categorized into four distinct lineages. In this investigation, we expressed five VP4(aa26-476) proteins from different P[8] lineages of human rotavirus in E. coli and assessed their immunogenicity in rabbits. Among the different P[8] strains, the Wa-VP4 protein, derived from the MT025868.1 strain of the P[8]-1 lineage, exhibited successful purification in a highly homogeneous form and significantly elicited higher levels of neutralizing antibodies (nAbs) against both homologous and heterologous rotaviruses compared to other VP4 proteins derived from different P[8] lineages in rabbits. Furthermore, we assessed the immunogenicity of the Wa-VP4 protein in mice, pigs, and cynomolgus monkeys, observing that it induced robust production of nAbs in all animals. Interestingly, there was no significant difference between in nAb titers against homologous and heterologous rotaviruses in pigs and mankeys. Collectively, these findings suggest that the Wa-VP4* protein may serve as a potential candidate for a rotavirus vaccine.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Capsid Proteins , Macaca fascicularis , Rotavirus Infections , Rotavirus Vaccines , Rotavirus , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Rotavirus Vaccines/immunology , Rotavirus Vaccines/administration & dosage , Antibodies, Viral/immunology , Antibodies, Viral/blood , Swine , Rabbits , Mice , Rotavirus/immunology , Rotavirus/genetics , Capsid Proteins/immunology , Capsid Proteins/genetics , Rotavirus Infections/prevention & control , Rotavirus Infections/immunology , Female , Mice, Inbred BALB C , Humans , Immunogenicity, Vaccine , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/genetics
9.
J Virol ; 96(17): e0070622, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36000839

ABSTRACT

Rotavirus infects intestinal epithelial cells and is the leading cause of gastroenteritis in infants worldwide. Upon viral infection, intestinal cells produce type I and type III interferons (IFNs) to alert the tissue and promote an antiviral state. These two types of IFN bind to different receptors but induce similar pathways that stimulate the activation of interferon-stimulated genes (ISGs) to combat viral infection. In this work, we studied the spread of a fluorescent wild-type (WT) SA11 rotavirus in human colorectal cancer cells lacking specific interferon receptors and compared it to that of an NSP1 mutant rotavirus that cannot interfere with the host intrinsic innate immune response. We could show that the WT rotavirus efficiently blocks the production of type I IFNs but that type III IFNs are still produced, whereas the NSP1 mutant rotavirus allows the production of both. Interestingly, while both exogenously added type I and type III IFNs could efficiently protect cells against rotavirus infection, endogenous type III IFNs were found to be key to limit infection of human intestinal cells by rotavirus. By using a fluorescent reporter cell line to highlight the cells mounting an antiviral program, we could show that paracrine signaling driven by type III IFNs efficiently controls the spread of both WT and NSP1 mutant rotavirus. Our results strongly suggest that NSP1 efficiently blocks the type I IFN-mediated antiviral response; however, its restriction of the type III IFN-mediated one is not sufficient to prevent type III IFNs from partially inhibiting viral spread in intestinal epithelial cells. Additionally, our findings further highlight the importance of type III IFNs in controlling rotavirus infection, which could be exploited as antiviral therapeutic measures. IMPORTANCE Rotavirus is one of the most common causes of gastroenteritis worldwide. In developing countries, rotavirus infections lead to more than 200,000 deaths in infants and children. The intestinal epithelial cells lining the gastrointestinal tract combat rotavirus infection by two key antiviral compounds known as type I and III interferons. However, rotavirus has developed countermeasures to block the antiviral actions of the interferons. In this work, we evaluated the arms race between rotavirus and type I and III interferons. We determined that although rotavirus could block the induction of type I interferons, it was unable to block type III interferons. The ability of infected cells to produce and release type III interferons leads to the protection of the noninfected neighboring cells and the clearance of rotavirus infection from the epithelium. This suggests that type III interferons are key antiviral agents and could be used to help control rotavirus infections in children.


Subject(s)
Epithelial Cells , Interferons , Intestinal Mucosa , Rotavirus Infections , Rotavirus , Antiviral Agents/immunology , Child , Epithelial Cells/immunology , Epithelial Cells/virology , Gastroenteritis/virology , Humans , Immunity, Innate , Infant , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Interferons/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Mutation , Rotavirus/genetics , Rotavirus/growth & development , Rotavirus/immunology , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Viral Nonstructural Proteins/genetics
10.
J Virol ; 96(16): e0062722, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35924923

ABSTRACT

Rotavirus live-attenuated vaccines, both mono- and pentavalent, generate broadly heterotypic protection. B-cells isolated from adults encode neutralizing antibodies, some with affinity for VP5*, that afford broad protection in mice. We have mapped the epitope of one such antibody by determining the high-resolution cryo-EM structure of its antigen-binding fragment (Fab) bound to the virion of a candidate vaccine strain, CDC-9. The Fab contacts both the distal end of a VP5* ß-barrel domain and the two VP8* lectin-like domains at the tip of a projecting spike. Its interactions with VP8* do not impinge on the likely receptor-binding site, suggesting that the mechanism of neutralization is at a step subsequent to initial attachment. We also examined structures of CDC-9 virions from two different stages of serial passaging. Nearly all the VP4 (cleaved to VP8*/VP5*) spikes on particles from the earlier passage (wild-type isolate) had transitioned from the "upright" conformation present on fully infectious virions to the "reversed" conformation that is probably the end state of membrane insertion, unable to mediate penetration, consistent with the very low in vitro infectivity of the wild-type isolate. About half the VP4 spikes were upright on particles from the later passage, which had recovered substantial in vitro infectivity but had acquired an attenuated phenotype in neonatal rats. A mutation in VP4 that occurred during passaging appears to stabilize the interface at the apex of the spike and could account for the greater stability of the upright spikes on the late-passage, attenuated isolate. IMPORTANCE Rotavirus live-attenuated vaccines generate broadly heterotypic protection, and B-cells isolated from adults encode antibodies that are broadly protective in mice. Determining the structural and mechanistic basis of broad protection can contribute to understanding the current limitations of vaccine efficacy in developing countries. The structure of an attenuated human rotavirus isolate (CDC-9) bound with the Fab fragment of a broadly heterotypic protective antibody shows that protection is probably due to inhibition of the conformational transition in the viral spike protein (VP4) critical for viral penetration, rather than to inhibition of receptor binding. A comparison of structures of CDC-9 virus particles at two stages of serial passaging supports a proposed mechanism for initial steps in rotavirus membrane penetration.


Subject(s)
Broadly Neutralizing Antibodies , Capsid Proteins , Epitopes, B-Lymphocyte , Rotavirus , Vaccines, Attenuated , Virion , Animals , Broadly Neutralizing Antibodies/immunology , Broadly Neutralizing Antibodies/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/immunology , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/ultrastructure , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/ultrastructure , Mice , Protein Conformation , Rats , Rotavirus/chemistry , Rotavirus/classification , Rotavirus/immunology , Rotavirus/physiology , Serial Passage , Vaccines, Attenuated/chemistry , Vaccines, Attenuated/immunology , Vaccines, Attenuated/metabolism , Virion/immunology , Virion/metabolism , Virion/ultrastructure
11.
Viruses ; 13(12)2021 12 13.
Article in English | MEDLINE | ID: mdl-34960760

ABSTRACT

Rotavirus is the major cause of severe gastroenteritis in children aged <5 years. Introduction of the G1P[8] Rotarix® rotavirus vaccine in Malawi in 2012 has reduced rotavirus-associated hospitalisations and diarrhoeal mortality. However, the impact of rotavirus vaccine on the severity of gastroenteritis presented in children requiring hospitalisation remains unknown. We conducted a hospital-based surveillance study to assess the impact of Rotarix® vaccination on the severity of gastroenteritis presented by Malawian children. Stool samples were collected from children aged <5 years who required hospitalisation with acute gastroenteritis from December 2011 to October 2019. Gastroenteritis severity was determined using Ruuska and Vesikari scores. Rotavirus was detected using enzyme immunoassay. Rotavirus genotypes were determined using nested RT-PCR. Associations between Rotarix® vaccination and gastroenteritis severity were investigated using adjusted linear regression. In total, 3159 children were enrolled. After adjusting for mid-upper arm circumference (MUAC), age, gender and receipt of other vaccines, all-cause gastroenteritis severity scores were 2.21 units lower (p < 0.001) among Rotarix®-vaccinated (n = 2224) compared to Rotarix®-unvaccinated children (n = 935). The reduction in severity score was observed against every rotavirus genotype, although the magnitude was smaller among those infected with G12P[6] compared to the remaining genotypes (p = 0.011). Each one-year increment in age was associated with a decrease of 0.43 severity score (p < 0.001). Our findings provide additional evidence on the impact of Rotarix® in Malawi, lending further support to Malawi's Rotarix® programme.


Subject(s)
Gastroenteritis/prevention & control , Rotavirus Infections/prevention & control , Rotavirus Vaccines/administration & dosage , Rotavirus/immunology , Child, Preschool , Feces/virology , Female , Gastroenteritis/epidemiology , Gastroenteritis/pathology , Gastroenteritis/virology , Genotype , Hospitalization , Humans , Infant , Malawi/epidemiology , Male , Rotavirus/classification , Rotavirus/genetics , Rotavirus/isolation & purification , Rotavirus Infections/epidemiology , Rotavirus Infections/pathology , Rotavirus Infections/virology , Severity of Illness Index , Vaccination , Vaccines, Attenuated/administration & dosage
12.
Int J Mol Sci ; 22(24)2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34948268

ABSTRACT

Rotavirus (RV) and norovirus (NoV) are the leading causes of acute gastroenteritis (AGE) worldwide. Several studies have demonstrated that histo-blood group antigens (HBGAs) have a role in NoV and RV infections since their presence on the gut epithelial surfaces is essential for the susceptibility to many NoV and RV genotypes. Polymorphisms in genes that code for enzymes required for HBGAs synthesis lead to secretor or non-secretor and Lewis positive or Lewis negative individuals. While secretor individuals appear to be more susceptible to RV infections, regarding NoVs infections, there are too many discrepancies that prevent the ability to draw conclusions. A second factor that influences enteric viral infections is the gut microbiota of the host. In vitro and animal studies have determined that the gut microbiota limits, but in some cases enhances enteric viral infection. The ways that microbiota can enhance NoV or RV infection include virion stabilization and promotion of virus attachment to host cells, whereas experiments with microbiota-depleted and germ-free animals point to immunoregulation as the mechanism by which the microbiota restrict infection. Human trials with live, attenuated RV vaccines and analysis of the microbiota in responder and non-responder individuals also allowed the identification of bacterial taxa linked to vaccine efficacy. As more information is gained on the complex relationships that are established between the host (glycobiology and immune system), the gut microbiota and intestinal viruses, new avenues will open for the development of novel anti-NoV and anti-RV therapies.


Subject(s)
Caliciviridae Infections/microbiology , Rotavirus Infections/microbiology , Animals , Blood Group Antigens/immunology , Blood Group Antigens/metabolism , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Gastroenteritis/microbiology , Gastrointestinal Microbiome/physiology , Genotype , Glycomics , Humans , Immunity , Norovirus/immunology , Norovirus/pathogenicity , Rotavirus/immunology , Rotavirus/pathogenicity , Rotavirus Infections/immunology , Rotavirus Infections/virology , Vaccine Efficacy , Viral Vaccines
13.
Nat Commun ; 12(1): 7288, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911947

ABSTRACT

Identifying risk factors for impaired oral rotavirus vaccine (ORV) efficacy in low-income countries may lead to improvements in vaccine design and delivery. In this prospective cohort study, we measure maternal rotavirus antibodies, environmental enteric dysfunction (EED), and bacterial gut microbiota development among infants receiving two doses of Rotarix in India (n = 307), Malawi (n = 119), and the UK (n = 60), using standardised methods across cohorts. We observe ORV shedding and seroconversion rates to be significantly lower in Malawi and India than the UK. Maternal rotavirus-specific antibodies in serum and breastmilk are negatively correlated with ORV response in India and Malawi, mediated partly by a reduction in ORV shedding. In the UK, ORV shedding is not inhibited despite comparable maternal antibody levels to the other cohorts. In both India and Malawi, increased microbiota diversity is negatively correlated with ORV immunogenicity, suggesting that high early-life microbial exposure may contribute to impaired vaccine efficacy.


Subject(s)
Gastrointestinal Microbiome , Infant, Newborn, Diseases/prevention & control , Rotavirus Infections/microbiology , Rotavirus Infections/prevention & control , Rotavirus Vaccines/administration & dosage , Rotavirus/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female , Humans , Immunity, Maternally-Acquired , Immunoglobulin A/blood , Immunoglobulin A/immunology , India , Infant , Infant, Newborn , Infant, Newborn, Diseases/blood , Infant, Newborn, Diseases/microbiology , Infant, Newborn, Diseases/virology , Malawi , Male , Milk, Human/chemistry , Milk, Human/immunology , Pregnancy , Prospective Studies , Rotavirus/genetics , Rotavirus/physiology , Rotavirus Infections/blood , Rotavirus Infections/virology , Rotavirus Vaccines/immunology , United Kingdom , Vaccine Efficacy , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Virus Shedding
14.
Pediatr Infect Dis J ; 40(12): 1135-1143, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34870393

ABSTRACT

BACKGROUND: Rotavirus causes 215,000 deaths from severe childhood diarrhea annually. Concerns exist that a monovalent vaccine (RV1) and a pentavalent vaccine (RV5) may be less effective against rotavirus strains not contained in the vaccines. We estimated the vaccine effectiveness (VE) of RV1 and RV5 against severe rotavirus gastroenteritis caused by vaccine (homotypic) and nonvaccine (partially and fully heterotypic) strains. METHODS: After conducting a systematic review, we meta-analyzed 31 case-control studies (N = 27,293) conducted between 2006 and 2020 using a random-effects regression model. RESULTS: In high-income countries, RV1 VE was 10% lower against partially heterotypic (P = 0.04) and fully heterotypic (P = 0.10) compared with homotypic strains (homotypic VE: 90% [95% confidence intervals (CI): 82-94]; partially heterotypic VE: 79% [95% CI: 71-85]; fully heterotypic VE: 80% [95% CI: 65-88]). In middle-income countries, RV1 VE was 14-16% lower against partially heterotypic (P = 0.06) and fully heterotypic (P = 0.04) compared with homotypic strains (homotypic VE: 81% [95% CI: 69-88]; partially heterotypic VE: 67% [95% CI: 54-76]; fully heterotypic VE: 65% [95% CI: 51-75]). Strain-specific RV5 VE differences were less pronounced, and primarily derived from high-income countries. Limited data were available from low-income countries. CONCLUSIONS: Vaccine effectiveness of RV1 and RV5 was somewhat lower against nonvaccine than vaccine strains. Ongoing surveillance is important to continue long-term monitoring for strain replacement, particularly in low-income settings where data are limited.


Subject(s)
Rotavirus Infections/prevention & control , Rotavirus Vaccines/immunology , Rotavirus/classification , Rotavirus/immunology , Vaccine Efficacy , Case-Control Studies , Child , Diarrhea/virology , Hospitalization , Humans , Infant , Rotavirus/genetics , Rotavirus Infections/virology , Rotavirus Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
15.
Sci Rep ; 11(1): 22037, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764353

ABSTRACT

Conjugate vaccine platform is a promising strategy to overcome the poor immunogenicity of bacterial polysaccharide antigens in infants and children. A carrier protein in conjugate vaccines works not only as an immune stimulator to polysaccharide, but also as an immunogen; with the latter generally not considered as a measured outcome in real world. Here, we probed the potential of a conjugate vaccine platform to induce enhanced immunogenicity of a truncated rotavirus spike protein ΔVP8*. ΔVP8* was covalently conjugated to Vi capsular polysaccharide (Vi) of Salmonella Typhi to develop a bivalent vaccine, termed Vi-ΔVP8*. Our results demonstrated that the Vi-ΔVP8* vaccine can induce specific immune responses against both antigens in immunized mice. The conjugate vaccine elicits high antibody titers and functional antibodies against S. Typhi and Rotavirus (RV) when compared to immunization with a single antigen. Together, these results indicate that Vi-ΔVP8* is a potent and immunogenic vaccine candidate, thus strengthening the potential of conjugate vaccine platform with enhanced immune responses to carrier protein, including ΔVP8*.


Subject(s)
Rotavirus Infections/prevention & control , Rotavirus/immunology , Salmonella typhi/immunology , Typhoid Fever/prevention & control , Vaccines, Combined/immunology , Vaccines, Conjugate/immunology , Viral Proteins/immunology , Animals , Humans , Immunization , Mice , Polysaccharides, Bacterial/immunology , Polysaccharides, Bacterial/pharmacology , Rotavirus Infections/immunology , Typhoid Fever/immunology , Vaccines, Combined/pharmacology , Vaccines, Conjugate/pharmacology , Viral Proteins/pharmacology
16.
Sci Rep ; 11(1): 21760, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34741103

ABSTRACT

Group A rotaviruses (RVA) remain a leading cause of pediatric diarrhea worldwide, in part due to underperformance of currently approved live-attenuated, oral vaccines in low-and-middle income countries. Improved immune correlates of protection (CoP) for existing oral vaccines and novel strategies to evaluate the performance of next-generation vaccines are needed. Use of oral vaccines as challenge agents in controlled human infection models is a potential approach to CoP discovery that remains underexplored. In a live-attenuated, oral rotavirus vaccine (Rotarix, GlaxoSmithKline) efficacy trial conducted among infants in Dhaka, Bangladesh, we explored the potential for the second dose of the two-dose series to be considered a challenge agent through which RVA immunity could be explored, using fecal virus shedding post-dose 2 as a marker of mucosal immunity. Among 180 vaccinated infants who completed the parent study per protocol, the absence of fecal vaccine shedding following the second dose of Rotarix suggested intestinal mucosal immunity generated by the first dose and a decreased risk of RVA diarrhea through 2 years of life (RR 0.616, 95% CI 0.392-0.968). Further development of controlled human infection models for group A rotaviruses, especially in prospective studies with larger sample sizes, may be a promising tool to assess rotavirus vaccine efficacy and CoPs.


Subject(s)
Immunity, Mucosal , Rotavirus Infections/prevention & control , Rotavirus Vaccines/immunology , Administration, Oral , Cohort Studies , Feces/chemistry , Humans , Infant , Rotavirus/immunology , Rotavirus Vaccines/analysis , Vaccines, Attenuated/analysis , Vaccines, Attenuated/immunology
17.
J Infect Dis ; 224(12 Suppl 2): S801-S804, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34528671

ABSTRACT

Safe and effective rotavirus vaccines (RVs) are needed to reduce the enormous public health burden of rotavirus illness in developing countries. Vaccination is critical for effective control of rotavirus infection since it cannot be prevented with improvements in water and sanitation. The International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b) has completed several groundbreaking RV trials (Phase I-Phase IV). The safety, immunogenicity, efficacy, and effectiveness of different RVs were evaluated among both urban and rural populations. In this study, we present the results, policy implications, and lessons learned for successful implementation of these trials as well as future directions for rotavirus vaccination in Bangladesh.


Subject(s)
Diarrhea/prevention & control , Rotavirus Infections/prevention & control , Rotavirus Vaccines , Rotavirus , Bangladesh , Humans , Infant , Rotavirus/immunology , Vaccine Efficacy
18.
J Infect Dis ; 224(12 Suppl 2): S331-S342, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34590142

ABSTRACT

Worldwide, rotavirus is the leading pathogen causing severe diarrhea in children and a major cause of under 5 years mortality. In 1998, the first rotavirus vaccine, RotaShield, was licensed in the United States but a rare adverse event, intussusception, led to its withdrawal. Seven years passed before the next generation of vaccines became available, Rotarix (GSK) and Rotateq (Merck), and 11 years later, 2 additional vaccines from India, Rotavac (Bharat) and Rotasiil (Serum Institute), were recommended by World Health Organization for all children. Today, these vaccines are used in more than 100 countries and have contributed to marked decreases in hospitalizations and deaths from diarrhea. However, these live oral vaccines are less effective in low-income countries with high under 5 years mortality for reasons that are not understood. Efforts to develop new vaccines that avoid the oral route are in progress and will likely be needed to ultimately control rotavirus disease.


Subject(s)
Diarrhea/prevention & control , Rotavirus Infections/prevention & control , Rotavirus Vaccines/administration & dosage , Rotavirus/immunology , Child , Diarrhea/virology , Humans , Infant , Rotavirus Vaccines/adverse effects , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects
19.
J Infect Dis ; 224(12 Suppl 2): S184-S193, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34469564

ABSTRACT

BACKGROUND: To inform the introduction of pneumococcal conjugate vaccine (PCV) and rotavirus vaccine, the World Health Organization (WHO) established the Global Invasive Bacterial Vaccine-Preventable Disease Surveillance Network (GISN) and the Global Rotavirus Surveillance Network (GRSN) in 2008. We investigated whether participation in these networks or other surveillance was associated with vaccine introduction. METHODS: Between 2006 and 2018, among all WHO member states, we used multivariable models adjusting for economic status to assess (1) the association between surveillance for pneumococcal disease or rotavirus disease, including participation in GISN or GRSN and the introduction of the PCV or the rotavirus vaccine, respectively, and (2) the association between the rotavirus disease burden and the rotavirus vaccine introduction among 56 countries participating in GRSN from 2008 to 2018. RESULTS: Countries that participated in or conducted surveillance for invasive pneumococcal disease or rotavirus disease were 3.5 (95% confidence interval [CI], 1.7-7.1) and 4.2 (95% CI, 2.1-8.6) times more likely to introduce PCV or rotavirus respectively, compared to those without surveillance. Among countries participating in GRSN, there was insufficient evidence to demonstrate an association between countries with higher rotavirus positivity and vaccine introduction. CONCLUSIONS: Surveillance should be incorporated into advocacy strategies to encourage the introduction of vaccines, with countries benefiting from data from, support for, and coordination of international disease surveillance networks.


Subject(s)
Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Population Surveillance , Rotavirus Infections/prevention & control , Rotavirus Vaccines/administration & dosage , Vaccines, Conjugate/immunology , Humans , Pneumococcal Infections/epidemiology , Pneumococcal Vaccines/therapeutic use , Rotavirus/immunology , Rotavirus Infections/epidemiology , Rotavirus Vaccines/therapeutic use , Vaccines, Conjugate/therapeutic use
20.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34475219

ABSTRACT

Group A rotaviruses cause severe gastroenteritis in infants and young children worldwide, with P[II] genogroup rotaviruses (RVs) responsible for >90% of global cases. RVs have diverse host ranges in different human and animal populations determined by host histo-blood group antigen (HBGA) receptor polymorphism, but details governing diversity, host ranges, and species barriers remain elusive. In this study, crystal structures of complexes of the major P[II] genogroup P[4] and P[8] genotype RV VP8* receptor-binding domains together with Lewis epitope-containing LNDFH I glycans in combination with VP8* receptor-glycan ligand affinity measurements based on NMR titration experiments revealed the structural basis for RV genotype-specific switching between ßß and ßα HBGA receptor-binding sites that determine RV host ranges. The data support the hypothesis that P[II] RV evolution progressed from animals to humans under the selection of type 1 HBGAs guided by stepwise host synthesis of type 1 ABH and Lewis HBGAs. The results help explain disease burden, species barriers, epidemiology, and limited efficacy of current RV vaccines in developing countries. The structural data has the potential to impact the design of future vaccine strategies against RV gastroenteritis.


Subject(s)
Blood Group Antigens/immunology , Evolution, Molecular , Rotavirus/genetics , Crystallography, X-Ray , Host Specificity/genetics , Humans , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Conformation , Rotavirus/chemistry , Rotavirus/immunology , Viral Nonstructural Proteins/chemistry , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...