1
|
Soni S, Kennedy MA, Wang D, Li F. The role and implication of rotavirus VP8∗ in viral infection and vaccine development. Virology 2025; 609:110563. [PMID: 40378555 DOI: 10.1016/j.virol.2025.110563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/08/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
Rotaviruses (RVs) are major causative agents of diarrhea in both humans and animals worldwide. Despite the successful development of live attenuated vaccines, the efficacy of these vaccines remains low in developing countries and RV infections still result in more than 200,000 deaths in children under 5 years old globally each year. These viruses are also an enteric pathogen for agricultural animals and have caused substantial economic losses annually to the animal livestock industry. Frequent reassortment and the emergence of new RV strains continue to pose a significant challenge to human and agricultural animal health. Attachment to susceptible cells by recognizing cell surface glycans is the first step of the RV lifecycle, which is directed by the RV spike protein VP8∗. VP8∗-host glycan receptor interactions are thought to be strain-specific and play an important role in RV replication fitness, tropism, and cross-species transmission. This review will summarize the current understanding of the roles of VP8∗ in engagement of glycan receptors and its functional consequences in impacting RV replication fitness and host ranges. The current progress towards developing a VP8∗-based RV vaccine is also discussed in the review.
Collapse
Affiliation(s)
- Shalini Soni
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA
| | - Michael A Kennedy
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Dan Wang
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA
| | - Feng Li
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA.
| |
Collapse
|
2
|
Chen Y, Luo G, Song F, Wang X, Zhang S, Ge S, Li T, Zhang J, Xia N. Truncated rotavirus VP4 proteins induce stronger protective immunity compared to P2 - VP8 in animal models. Antiviral Res 2025; 238:106156. [PMID: 40194664 DOI: 10.1016/j.antiviral.2025.106156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/17/2025] [Accepted: 04/05/2025] [Indexed: 04/09/2025]
Abstract
Group A rotavirus (RVA) is the primary causative agent of acute gastroenteritis (AGE) in children under five years of age, resulting in over 120,000 deaths annually. In previous studies, we identified truncated VP4∗ as a potentially more promising vaccine candidate compared to VP8∗ and VP5∗. This study aimed to compare the immunogenicity and protective efficacy of VP4∗ and P2-VP8, the most advanced recombinant rotavirus vaccine undergoing phase 3 clinical trial in various animal models, including mice, guinea pigs, rabbits, and piglets. The results indicated that the binding antibodies and neutralizing antibodies induced by VP4∗ were significantly higher levels compared to P2-VP8. Immunization with VP4∗ provided 100 % protection for mice against challenges with EDIM and LLR strains. Additionally, we were intrigued to discover that the VP4∗ antibody not only inhibited virus adsorption but also prevented the virus from entering cells following pre-adsorption. In summary, VP4∗ demonstrates greater immunogenicity and protective efficacy compared to P2-VP8, making it a more promising candidate antigen for recombinant rotavirus vaccines.
Collapse
MESH Headings
- Animals
- Rotavirus Infections/prevention & control
- Rotavirus Infections/immunology
- Rotavirus Infections/virology
- Capsid Proteins/immunology
- Capsid Proteins/genetics
- Rotavirus Vaccines/immunology
- Rotavirus Vaccines/administration & dosage
- Rotavirus Vaccines/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Rotavirus/immunology
- Rotavirus/genetics
- Mice
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Disease Models, Animal
- Rabbits
- Guinea Pigs
- Swine
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Immunogenicity, Vaccine
- Mice, Inbred BALB C
- Antigens, Viral/immunology
- Antigens, Viral/genetics
- Female
Collapse
Affiliation(s)
- Yaling Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Guoxing Luo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China; Novel Product R&D Department, Xiamen Innovax Biotech Co., Ltd., Xiamen, 361022, Fujian, China
| | - Feibo Song
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xuechun Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Shiyin Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Shengxiang Ge
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China.
| | - Tingdong Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China.
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
3
|
Geng X, Cao X, Jiang J, Li L. Relationship between fecal viral load and peripheral blood Th1/Th2, related cytokines in children with rotavirus infection enteritis. Microb Pathog 2025; 206:107745. [PMID: 40425054 DOI: 10.1016/j.micpath.2025.107745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 04/06/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
This study aimed to examine the association between fecal viral load and peripheral blood Th1/Th2 cytokines in children with rotavirus (RV) infection enteritis. A total of 102 children with RV enteritis and 102 healthy infants were enrolled between April 2022 and April 2024. Fecal viral load was assessed using real-time polymerase chain reaction (RT-PCR), while T lymphocyte subsets and Th1/Th2 cytokines were measured using flow cytometry and ELISA, respectively. Pearson correlation analysis was employed to explore the relationship between RV load and Th1/Th2 cytokines. The RV enteritis group exhibited significantly higher fecal viral load than the control group (P<0.05). Moreover, levels of peripheral blood CD3+, CD4+, and CD4+/CD8+ were significantly lower in the RV enteritis group compared to the control group (P<0.05). Additionally, the levels of IL-6, IL-10, TNF-α, and IFN-γ were significantly elevated in the RV enteritis group (P<0.05). Correlation analysis revealed a negative association between viral load and CD3+, CD4+, and CD4+/CD8+ levels, and a positive association with IL-6, IL-10, TNF-α, and IFN-γ levels (P<0.05). Notably, no significant correlation was found between viral load and CD8+ levels (P>0.05). These findings suggest that viral load correlates with Th1/Th2 cytokines in children with RV infection enteritis, providing potential targets for early diagnosis and treatment.
Collapse
Affiliation(s)
- Xuejing Geng
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xiansheng Cao
- Gastrointestinal Surgery, Hernia and Abdominal Wall Surgery Ward 1, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Jixiang Jiang
- Department of Interventional Vascular, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Lei Li
- Gastrointestinal Surgery, Hernia and Abdominal Wall Surgery Ward 1, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China.
| |
Collapse
|
4
|
Mustafa K, Faryal R, Alam MM, Rana S, Umair M, Shah TA. Epidemiology and clinical features of Rotavirus infection among children in Rawalpindi, Pakistan. PLoS One 2025; 20:e0324037. [PMID: 40392897 PMCID: PMC12091768 DOI: 10.1371/journal.pone.0324037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 04/19/2025] [Indexed: 05/22/2025] Open
Abstract
Group A rotavirus (RVA) associated gastroenteritis is a major cause of infantile morbidity and mortality, globally. Pakistan had the highest rates of gastroenteritis among kids, every year. Our study aimed to assess the RVA disease burden and circulating genotypes in Rawalpindi, before the vaccine's introduction in Pakistan. Stool samples were collected from children < 5 years of age admitted at Benazir Bhutto Hospital, Rawalpindi, from November 2014 to May 2015. Of the 300 stool samples, 47% of children were found positive for RVA antigen on ELISA, with the highest prevalence (52%) in infants less than 7 months of age. Rotavirus positive cases through real-time PCR were 65.5%. Fever and diarrhea were significantly related to RVA infection when compared to RVA-negative cases (P = 0.02). It is the first report on an upsurge of G12P[6] (17.24%) along with the rise of previously declining G3 in the current epidemiological area. The other prevalent types were G1P[8] (12.07%), G1P[4] (6.90%), G1P[6] (5.17%), G3P[6] (5.17%), followed by G2P[6], G3P[4], G9P[4], and G12P[8] each found with a prevalence of (3.45%). This study reports G3P[4], G3P[6] and G12P[4] for the first time in Pakistan. Mixed genotype infections were found in 21% of cases. G12P[6], which was the predominant genotype in this study, was found to be significantly associated with fever (P = 0.03). This study provides valuable data on the significantly highest prevalence of RVA-associated gastroenteritis in kids of Rawalpindi, Pakistan, and elucidates the vast diversity of circulating RVA genotypes. The reported disease burden, genotypes, and clinical symptoms would enable public health dealers to cope with the severity of the disease. It also provides an evolutionary trend of changing genotypes in the country.
Collapse
Affiliation(s)
- Kiren Mustafa
- Department of Virology, National Institute of Health, Chak Shahzad, Islamabad, Pakistan
- Department of Microbiology, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Rani Faryal
- Department of Microbiology, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Muhammad Masroor Alam
- Department of Virology, National Institute of Health, Chak Shahzad, Islamabad, Pakistan
| | - Suleman Rana
- Department of Virology, National Institute of Health, Chak Shahzad, Islamabad, Pakistan
| | - Massab Umair
- Department of Virology, National Institute of Health, Chak Shahzad, Islamabad, Pakistan
| | - Tawaf Ali Shah
- Department of Microbiology, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| |
Collapse
|
5
|
Xu G, Zhou J, Liu K, Wang Y, Tsikari T, Qin F, van den Hil F, Boor PPC, Ayada I, de Vries AC, Li J, Jiang S, Offermans DM, Kainov DE, Janssen HLA, Peppelenbosch MP, Bijvelds MJC, Wang W, Orlova VV, Pan Q, Li P. Macrophage-augmented intestinal organoids model virus-host interactions in enteric viral diseases and facilitate therapeutic development. Nat Commun 2025; 16:4475. [PMID: 40368896 PMCID: PMC12078800 DOI: 10.1038/s41467-025-59639-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
The pathogenesis of enteric viral infections is attributed to both viral replication and the resultant immune-inflammatory response. To recapitulate this complex pathophysiology, we engineer macrophage-augmented organoids (MaugOs) by integrating human macrophages into primary intestinal organoids. Echovirus 1, echovirus 6, rotavirus, seasonal coronavirus OC43 and SARS-CoV-2- known to directly invade the intestine- are used as disease modalities. We demonstrate that these viruses efficiently propagate in MaugOs and stimulate the host antiviral response. However, rotavirus, coronavirus OC43 and SARS-CoV-2, but not the two echoviruses, trigger inflammatory responses. Acetate, a microbial metabolite abundantly present in the intestine, potently inhibits virus-induced inflammatory responses in MaugOs, while differentially affecting viral replication in macrophages and organoids. Furthermore, we provide a proof-of-concept of combining antiviral agent with either anti-inflammatory regimen or acetate to simultaneously inhibit viral infection and inflammatory response in MaugOs. Collectively, these findings demonstrate that MaugOs are innovative tools for studying the complex virus-host interactions and advancing therapeutic development.
Collapse
Affiliation(s)
- Guige Xu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, 271018, China
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiangrong Zhou
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Kuan Liu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yining Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Theano Tsikari
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Fang Qin
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, China
| | - Francijna van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Ibrahim Ayada
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Annemarie C de Vries
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Jiajing Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Shijin Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Dewy M Offermans
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Denis E Kainov
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7028, Trondheim, Norway
| | - Harry L A Janssen
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Wenshi Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, China
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Pengfei Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands.
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Pawłuszkiewicz K, Ryglowski PJ, Idzik N, Błaszczyszyn K, Kucharczyk E, Gaweł-Dąbrowska D, Siczek M, Widelski J, Paluch E. Rotavirus Infections: Pathophysiology, Symptoms, and Vaccination. Pathogens 2025; 14:480. [PMID: 40430800 PMCID: PMC12114175 DOI: 10.3390/pathogens14050480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 05/08/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Rotavirus (RV) is the most common cause of severe acute gastroenteritis (AGE) in children under five years of age. This review summarizes current knowledge on RV infections, with a particular focus on viral structure, pathophysiological mechanisms, and age-dependent clinical presentation. Special attention is given to systemic manifestations, including central nervous system involvement, autoimmune responses such as type 1 diabetes and celiac disease, and rare associations with biliary atresia. The mechanisms of RV-induced diarrhea and vomiting are discussed in detail. Clinical severity scoring systems-such as the Vesikari and Clark scales-and dehydration assessment tools-the Clinical Dehydration Scale (CDS) and the Dehydration: Assessing Kids Accurately (DHAKA) score-are compared. The review highlights differences in disease course between children under and over five years, emphasizing that RV is not limited to early childhood. A major section addresses the global effectiveness of vaccination programs, their role in reducing disease burden, coverage challenges, and decreased efficacy in low-income countries. Particular focus is placed on high-risk groups, including preterm and immunocompromised infants.
Collapse
Affiliation(s)
- Karolina Pawłuszkiewicz
- Faculty of Medicine, Wroclaw Medical University, Wybrzeże L. Pasteura 1, 50-367 Wrocław, Poland; (K.P.); (P.J.R.); (N.I.); (E.K.)
| | - Piotr Józef Ryglowski
- Faculty of Medicine, Wroclaw Medical University, Wybrzeże L. Pasteura 1, 50-367 Wrocław, Poland; (K.P.); (P.J.R.); (N.I.); (E.K.)
| | - Natalia Idzik
- Faculty of Medicine, Wroclaw Medical University, Wybrzeże L. Pasteura 1, 50-367 Wrocław, Poland; (K.P.); (P.J.R.); (N.I.); (E.K.)
| | - Katarzyna Błaszczyszyn
- Jan Mikulicz-Radecki University Hospital in Wroclaw, Borowska 213, 50-556 Wrocław, Poland;
| | - Emilia Kucharczyk
- Faculty of Medicine, Wroclaw Medical University, Wybrzeże L. Pasteura 1, 50-367 Wrocław, Poland; (K.P.); (P.J.R.); (N.I.); (E.K.)
| | - Dagmara Gaweł-Dąbrowska
- Division of Public Health, Faculty of Health Sciences, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland;
| | - Marta Siczek
- Department of Forensic Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 4, 50-345 Wroclaw, Poland;
| | - Jarosław Widelski
- Department of Pharmacognosy with Medicinal Plant Unit, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland
| | - Emil Paluch
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, St. T. Chałubińskiego 4, 50-376 Wrocław, Poland
| |
Collapse
|
7
|
Mametja PM, Motshudi MC, Naidoo CM, Rakau K, Seheri LM, Mkolo NM. Tapping into Metabolomics for Understanding Host and Rotavirus Group A Interactome. Life (Basel) 2025; 15:765. [PMID: 40430193 PMCID: PMC12113392 DOI: 10.3390/life15050765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/27/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Group A rotavirus continues to be a leading global etiological agent of severe gastroenteritis in young children under 5 years of age. The replication of this virus in the host is associated with the occurrence of Lewis antigens and the secretor condition. Moreover, histo-blood group antigens (HBGAs) act as attachment factors to the outer viral protein of VP4 for rotavirus. Therefore, in this study, we employed a metabolomic approach to reveal potential signature metabolic molecules and metabolic pathways specific to rotavirus P[8] strain infection (VP4 genotype), which is associated with the expression of HBGA combined secretor and Lewis (Le) phenotypes, specifically secretor/Le(a+b+). Further integration of the achieved metabolomics results with lipidomic and proteomics metadata analyses was performed. Saliva samples were collected from children diagnosed as negative or positive for rotavirus P[8] strain infection (VP4 genotype), which is associated with the HBGA combined secretor/Le(a+b+). A total of 22 signature metabolic molecules that were downregulated include butyrate, putrescine, lactic acid, and 7 analytes. The upregulated metabolic molecule was 2,3-Butanediol. Significant pathway alterations were also specifically observed in various metabolism processes, including galactose and butanoate metabolisms. Butyrate played a significant role in viral infection and was revealed to exhibit different reactions with glycerolipids, glycerophospholipids, sphingolipids, sterol lipids, and fatty acyls. Moreover, butyrate might interact with protein receptors of free fatty acid receptor 2 (FFAR2) and free fatty acid receptor 3 (FFAR3). The revealed metabolic pathways and molecule might provide fundamental insight into the status of rotavirus P[8] strain infection for monitoring its effects on humans.
Collapse
Affiliation(s)
- Phiona Moloi Mametja
- Department of Biology and Environmental Sciences, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (P.M.M.); (M.C.M.); (C.M.N.)
| | - Mmei Cheryl Motshudi
- Department of Biology and Environmental Sciences, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (P.M.M.); (M.C.M.); (C.M.N.)
| | - Clarissa Marcelle Naidoo
- Department of Biology and Environmental Sciences, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (P.M.M.); (M.C.M.); (C.M.N.)
| | - Kebareng Rakau
- Diarrheal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (K.R.); (L.M.S.)
| | - Luyanda Mapaseka Seheri
- Diarrheal Pathogens Research Unit, Department of Virology, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (K.R.); (L.M.S.)
| | - Nqobile Monate Mkolo
- Department of Biology and Environmental Sciences, Sefako Makgatho Health Sciences University, Pretoria 0204, South Africa; (P.M.M.); (M.C.M.); (C.M.N.)
| |
Collapse
|
8
|
Miao Z, Du Y, Dai A, Yang M, Chen C, Yan R, Gao J, Chen Y, Cao K, Jiang D, Zhang X, Wu X, Chen M, You Y, Zhou W, Chen D, Qi J, Zhao S, Lin X, Yang S. Epidemic characteristics and effectiveness of vaccine intervention on rotavirus infection: a real-world observational study in Zhejiang Province, China. Front Public Health 2025; 13:1596899. [PMID: 40416696 PMCID: PMC12098449 DOI: 10.3389/fpubh.2025.1596899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 05/27/2025] Open
Abstract
Background Rotavirus infection, the most common cause of infant infectious diarrhoea and related deaths worldwide, has imposed a high disease burden in China, especially in Zhejiang Province. This study described the overall epidemiological characteristics and trends of reported rotavirus infections in Zhejiang Province from 2005 to 2022 and evaluated the effectiveness of rotavirus vaccines on the incidence of rotavirus infection. Materials and methods Data on reported cases of rotavirus infection from 2005 to 2022 were extracted from the China Disease Prevention and Control Information System. Information on rotavirus vaccination was obtained from the Zhejiang Provincial Viral Diarrhoea Surveillance Site in 2022. Join-point regression, spatial and temporal aggregation analysis, and an age-period-cohort model were used to explore the epidemiological trends of rotavirus infection. Interrupted time series analysis and an overdispersed Poisson model were used to quantify the effectiveness of rotavirus vaccines. Results The average age-standardized reporting incidence rate (ASRIR) of rotavirus infection in Zhejiang Province was 38.58/100,000, particularly in children aged 0-2 years, who had the highest average annual incidence of 951.63/100,000. The annual ASRIR of all ages showed a significant upward trend before 2017 (average percentage change [APC] = 21.64%) and then decreased significantly (APC = -23.02%). However, in children aged 6-19 years, the annual incidence presented a sustained and significant upward trend over time. The rotavirus infection peak showed a seasonal drift in Zhejiang Province, shifting from November before 2014 to January after 2014. Spatiotemporal aggregation revealed two clusters. The spatio-temporal scanning found two spatio-temporal aggregation areas, the first level spatio-temporal aggregation area was distributed in Hangzhou, Jiaxing and Huzhou, and the second level spatio-temporal aggregation area was Lishui. The age-period-cohort model indicated that the risk of rotavirus infection was primarily concentrated in children aged 0-4 years. The vaccine effectiveness (VE) of rotavirus vaccines was 71.62% (95% confidence interval [CI]: 45.21-86.05%) in children aged 2-59 months, in which the VE of the human-bovine reassortant pentavalent vaccine (RV5) was 91.31% (95% CI: 74.39-97.97%). Since the implementation of RV5 vaccination in September 2018, the number of cases of rotavirus infection per month has decreased by 3,061 (65.27%) in Zhejiang Province. Conclusion The disease burden of rotavirus infection in Zhejiang Province was high, especially in children. Rotavirus vaccination have significantly reduced the incidence rate of rotavirus infection. Therefore, the prevention of infectious diarrhoea should be further strengthened, especially through increased coverage with the rotavirus vaccine.
Collapse
Affiliation(s)
- Ziping Miao
- Department of Communicable Diseases Control and Prevention, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yuxia Du
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Anqi Dai
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengya Yang
- Department of the Institution for Drug Clinical Trials, Quzhou People's Hospital, Quzhou, Zhejiang, China
| | - Can Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Rui Yan
- Department of Immunization Program, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jian Gao
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yijuan Chen
- Department of Communicable Diseases Control and Prevention, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Kexin Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Daixi Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaobao Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyue Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengsha Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue You
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenkai Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Dingmo Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaxing Qi
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Shiyong Zhao
- Department of Infectious Diseases, Hangzhou Children’s Hospital, Hangzhou, China
| | - Xianyao Lin
- Department of Infectious Diseases, Hangzhou Children’s Hospital, Hangzhou, China
| | - Shigui Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Department of Emergency Medicine, Second Affiliated Hospital, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
Collaborators
Shigui Yang, Xudong Zhou, Peige Song, Ning Zhang, Hao Lei, Junfang Xu, Jianbing Wang,
Collapse
|
9
|
Ong DS, Harris M, Hart JD, Russell FM. Indirect Effects of Universal Infant Rotavirus Vaccination: A Narrative Systematic Review. Vaccines (Basel) 2025; 13:503. [PMID: 40432114 PMCID: PMC12116122 DOI: 10.3390/vaccines13050503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objective: Rotavirus is a major cause of acute gastroenteritis (AGE) in children <5 years. While rotavirus vaccines are effective in reducing AGE, limited data on their indirect effects exist. The aim of our narrative systematic review was to summarise the indirect effects of rotavirus vaccines on unvaccinated children and adults (PROSPERO: CRD42023418015). Methods: Peer-reviewed articles and conference abstracts were searched through Medline, Embase and PubMed on 8 December 2024. Observational studies of national/regional vaccine introduction were included. We included five outcomes: rotavirus-AGE inpatient admissions, rotavirus-AGE outpatient attendances, all-cause AGE inpatient admissions, all-cause AGE outpatient attendances, and stool rotavirus positivity. Outcome measures reported as percent reduction or individual incidence rates for the pre- and post-introduction periods were transformed to incidence rate ratios (IRRs). Median IRRs and interquartile ranges (IQRs) were calculated for each outcome by age group (<5, 5-19, and >18 years). Results: From an initial 757 articles, 44 studies including 9,327,974 participants were included. In unvaccinated children <5 years, there were reductions in rotavirus-AGE admissions (median IRR: 0.62, IQR: 0.40-0.82), rotavirus-AGE outpatient attendances (0.74, 0.16-0.98), all-cause AGE admissions (0.70, 0.56-0.86), and stool rotavirus positivity (0.42, 0.31-0.57), but not all-cause AGE outpatient attendances (0.92, 0.78-1.17). Few studies reported these outcomes for children and adolescents aged 5-19 years and adults >18 years. Indirect effects appeared to be greater in higher income and lower under-five mortality settings. Conclusions: Understanding these indirect benefits is crucial for evaluating the broader impact and cost-effectiveness of rotavirus immunisation programs.
Collapse
Affiliation(s)
- Darren Suryawijaya Ong
- Asia-Pacific Health, Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (J.D.H.); (F.M.R.)
| | - Matthew Harris
- Asia-Pacific Health, Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (J.D.H.); (F.M.R.)
| | - John D. Hart
- Asia-Pacific Health, Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (J.D.H.); (F.M.R.)
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Fiona M. Russell
- Asia-Pacific Health, Infection, Immunity and Global Health, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (J.D.H.); (F.M.R.)
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
10
|
Mattison CP, Calderwood LE, Cates JE, Donald J, Hall AJ, Schmidt MA, Mirza SA. Seasonality of medically attended norovirus gastroenteritis and its association with climatic factors within an US integrated healthcare system, 2016-2019. PLoS One 2025; 20:e0318077. [PMID: 40343896 PMCID: PMC12063862 DOI: 10.1371/journal.pone.0318077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/09/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND While acute gastroenteritis (AGE) occurs year-round, norovirus has a winter seasonality in the United States. OBJECTIVE We analyzed norovirus seasonality within a US integrated healthcare delivery system from 2016-2019. METHODS Electronic medical records were collected for acute gastroenteritis (AGE) encounters with specific ICD-9/10 codes or clinical stool testing. Norovirus percent positivity was calculated as the 8-week centered rolling average. Temperature and absolute humidity data were measured via weather station. The relationship between these factors and weekly norovirus episodes were modeled via negative binomial models. RESULTS From 2016-2019, there were 198,181 AGE episodes reported; among the 18,998 episodes tested, 892 (5%) were norovirus positive. Norovirus percent positivity peaked in epidemiologic week 7 at 9%. Two negative binomial models showed significant inverse relationships between weekly number of norovirus episodes and both temperature and absolute humidity. CONCLUSION Norovirus AGE exhibited winter seasonality from 2016-2019, associated with lower temperatures and humidity. Understanding this seasonality may help predict peak transmission periods and their impact on healthcare resources.
Collapse
Affiliation(s)
- Claire P. Mattison
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Cherokee Nation Operational Solutions, Tulsa, Oklahoma, United States of America
| | - Laura E. Calderwood
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- Cherokee Nation Operational Solutions, Tulsa, Oklahoma, United States of America
| | - Jordan E. Cates
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Judy Donald
- Kaiser Permanente Northwest, Portland, Oklahoma, United States of America
| | - Aron J. Hall
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Mark A. Schmidt
- Kaiser Permanente Northwest, Portland, Oklahoma, United States of America
| | - Sara A. Mirza
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
11
|
Cai L, Tang B, Kong F, Chang Z, Zhang Y, Zheng Y, Wang L. Disease burden of rotavirus related diarrhea in children under 5 years in China: a meta-analysis. Sci Rep 2025; 15:15973. [PMID: 40341400 PMCID: PMC12062226 DOI: 10.1038/s41598-025-00778-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025] Open
Abstract
Rotavirus (RV) is a leading cause of severe diarrhea among children under five years of age in China. In this meta-analysis, we assessed the disease burden of RV-related diarrhea by analyzing 73 studies retrieved from the PubMed, Web of Science, China National Knowledge Infrastructure, and Wanfang databases (2013-2023). The incidence of RV-related diarrhea ranged from 0.637 /1000 to 31.46/1000 persons. The pooled RV positivity rate for the under-5 age group was 24.7% (95% confidence interval: 22.1-27.4), with higher positivity rates observed among inpatients compared to outpatients (24.1% vs. 22.2%). Notably, the RV positivity rate declined from 27.3 to 21.5% pre- and post- the RotaTeq® licensure and 28.8-22.5% following the COVID-19 pandemic. The G9P[8] genotype was predominant, accounting for 71.7% of the RV cases in the under-5 age group. Given the dynamic nature of the incidence rate of RV-related diarrhea and the prevalence of the G9P[8] genotype, it is imperative to enhance surveillance efforts targeting incidence of RV-related diarrhea and the circulating genotypes of rotavirus.
Collapse
Affiliation(s)
- Li Cai
- Division of Infectious Disease, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Bicheng Tang
- The Sixth People's Hospital of Dongguan, Dongguan, 523129, Guangdong, China
| | - Fanxu Kong
- Division of Infectious Disease, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
- Chinese Field Epidemiology Training Program (CFETP), Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Zhaorui Chang
- Division of Infectious Disease, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yanping Zhang
- Division of Infectious Disease, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yaming Zheng
- Division of Infectious Disease, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
| | - Liping Wang
- Division of Infectious Disease, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
| |
Collapse
|
12
|
Penfold MD, Prabhakar S, Susi A, Rajnik M, Nylund CM, Eberly MD. Pediatric Rotavirus Hospitalization Rates in the Military Health System Before and During the COVID-19 Pandemic. Vaccines (Basel) 2025; 13:492. [PMID: 40432104 PMCID: PMC12115858 DOI: 10.3390/vaccines13050492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Rotavirus gastroenteritis is a vaccine-preventable disease that leads to hospitalization in children less than 5 years of age. Immunizations to prevent rotavirus have greatly altered the epidemiology of significant diarrheal illness. It has been reported that routine immunization rates in children were impacted during the COVID-19 pandemic. Contrary to this fact, rates of many childhood illnesses also decreased. Methods: The Military Health System Data Repository (MDR) contains the health records of all military beneficiaries. We queried the MDR before and during the COVID-19 pandemic to assess for alterations in immunization rates and hospitalization rates and to assess for risk factors for significant (hospitalizations) rotavirus disease. Results: Our study included a cohort of 1.27 million children under the age of 5 years old. There were 186 unique cases of rotavirus-related hospitalizations over the 5-year study period. During COVID-19 Years 1 and 2, there was a decrease in rotavirus-related hospitalizations compared to the pre-pandemic period. During Year 3, there was a return to the pre-pandemic level of rotavirus hospitalization rates. Patients in the northern United States were less likely to be hospitalized from rotavirus when compared to those in the south. The patients at greatest risk were the youngest beneficiaries. Rotavirus vaccination rates declined in this age group during all three years of the pandemic. Conclusions: As the pandemic resulted in less frequent rotavirus immunizations in the Military Health System (MHS), there was not an increase in rotavirus-related hospitalizations above the pre-pandemic baseline.
Collapse
Affiliation(s)
- Matthew D. Penfold
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (S.P.); (A.S.); (M.R.)
| | - Sarah Prabhakar
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (S.P.); (A.S.); (M.R.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Apryl Susi
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (S.P.); (A.S.); (M.R.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Michael Rajnik
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (S.P.); (A.S.); (M.R.)
| | - Cade M. Nylund
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (S.P.); (A.S.); (M.R.)
| | - Matthew D. Eberly
- Pediatric Infectious Diseases, Brooke Army Medical Center, Joint Base San Antonia, Fort Sam Houston, San Antonio, TX 78234, USA
| |
Collapse
|
13
|
Fathi Shaheen MN, Ahmed NI, Elmahdy EM. Epidemiological surveillance of astrovirus, norovirus, rotavirus, and enterovirus in sewage (2022-2023) in Giza, Egypt. JOURNAL OF WATER AND HEALTH 2025; 23:587-601. [PMID: 40448462 DOI: 10.2166/wh.2025.324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 04/07/2025] [Indexed: 06/02/2025]
Abstract
The majority of people with enterically transmitted viruses excrete significant amounts of the virus in their feces for several days or weeks. Therefore, viruses causing diarrhea could be detected in the feces of infected persons and wastewater. In this study, the presence of human astrovirus (AstV), norovirus (NoV), rotavirus (RV), and enterovirus (EntV) was analyzed by real-time RT-PCR in raw sewage (n = 96), treated sewage (n = 96) and diarrheal stool samples (n = 200). Overall, 92.7% (89/96) of raw sewage samples and 48% (46/96) of treated sewage tested positive for at least one virus. The highest detection rates of the four viruses in raw sewage were observed in the winter season. Overall, the mean concentration of the four viruses was 7.3 log10 in raw and 4.8 log10 in treated wastewater, for a total removal of 34% of viral loads. In clinical samples, the most commonly detected virus was EntV followed by RV, NoV, and AstV. The mean concentrations of the four viruses in clinical samples ranged between 2.5 × 101 and 9.86 × 107 GC/g. The results presented here demonstrated that the environmental surveillance of entric viruses in sewage is a useful tool for the study of their transmission dynamics in humans and their molecular epidemiology.
Collapse
Affiliation(s)
- Mohamed Nasr Fathi Shaheen
- Environmental Virology Laboratory, Department of Water Pollution Research, Environment and Climate Change Research Institute, National Research Centre, 12622 Dokki, Cairo, Egypt E-mail:
| | - Nehal Ismail Ahmed
- Environmental Virology Laboratory, Department of Water Pollution Research, Environment and Climate Change Research Institute, National Research Centre, 12622 Dokki, Cairo, Egypt
| | - Elmahdy Mohamed Elmahdy
- Environmental Virology Laboratory, Department of Water Pollution Research, Environment and Climate Change Research Institute, National Research Centre, 12622 Dokki, Cairo, Egypt
| |
Collapse
|
14
|
Malla BA, Dubal ZB, Kumar A, Kumar ORV, Mohmad A, Mani P, Rajak KK, Bhilegaonkar KN. Comparative efficacy of recombinant VP6 protein based in-house Latex Agglutination test with other diagnostic assays for detection of Rotavirus A from calves, piglets and children. Comp Immunol Microbiol Infect Dis 2025; 119:102336. [PMID: 40187271 DOI: 10.1016/j.cimid.2025.102336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
In this study, VP6 protein of rotavirus A (RVA) was expressed in the prokaryotic system for the development of indigenous Latex Agglutination Test for diagnosis of RVA gastroenteritis in animals. Polyclonal anti-rVP6 IgG were raised in rabbits; purified and conjugated to carboxylated beads via covalent coupling for development of in-house LAT. Clinical utility of in-house developed LAT was evaluated on 313 stool samples collected from calves, children and piglets of Bareilly, Uttar Pradesh, India. In-house LAT yielded consistent results at weekly intervals for 2 months. Best visual perception of agglutination was observed when 5 μL beads coupled with 200 μg anti-rVP6 IgG and 10 μL of antigen reacted within reaction time of 2 minutes. Relative sensitivity and specificity of in-house LAT w.r.t RT-PCR was 65.45 % and 95.73 %, respectively; w.r.t commercial LFA was 75.55 % and 95.14 %, respectively and w.r.t RNA-PAGE was 70.27 % and 92.39 %, respectively. The kappa agreement between LAT and RT-PCR was 0.65 (substantial); between LAT and LFA was 0.7 (substantial) and between LAT and RNA-PAGE was 0.56 (moderate). Overall RVA stool positivity from children, calves and piglets with 4 assays was found to be 40 % (40/100), 9 % (9/100) and 16.81 % (19/113), respectively. Higher positivity was recorded in male (45.90 %, 28/61) than in female (30.76 %, 12/39) children. Developed LAT has fulfilled the WHO criteria for point-of-care testing with satisfactory efficacy for detection of RVA. This may serve as a preliminary assay for epidemiological surveillance of RVA antigen in determining rotavirus outbreaks in animal herds under resource-poor settings.
Collapse
Affiliation(s)
- Bilal Ahmad Malla
- Division of Veterinary Public Health, ICAR, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India.
| | - Zunjar Baburao Dubal
- Division of Veterinary Public Health, ICAR, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India.
| | - Ajay Kumar
- Division of Biochemistry, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India
| | | | - Aquil Mohmad
- Division of Veterinary Public Health, ICAR, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India
| | - Pashupathi Mani
- Division of Biochemistry, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India
| | - Kaushal Kishor Rajak
- Division of Biological Products, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India
| | - Kiran Narayan Bhilegaonkar
- Division of Veterinary Public Health, ICAR, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India
| |
Collapse
|
15
|
Li Y, Li K, Li Y, Fan C, Zhang L, Ren M, Cao L, Yu W, Yin Z. Post-Marketing Surveillance of Adverse Events Following Rotavirus Vaccination - China, 2013-2023. China CDC Wkly 2025; 7:580-585. [PMID: 40376258 PMCID: PMC12075480 DOI: 10.46234/ccdcw2025.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/18/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction Two live attenuated rotavirus vaccines (RVs) were licensed in China. Passive surveillance for adverse events following immunization (AEFI) provides valuable evidence for potential safety signal detection of RV in China. Methods We obtained data on RV doses administered and RV AEFI reports from the Chinese National Immunization Information System (CNIIS) during January 2013 to December 2023. We conducted a descriptive analysis of RV AEFI characteristics and estimated incidences of RV AEFI. Results During the study period, 77.36 million doses of RV were administered, and 20,556 RV AEFI reports were made, yielding an overall incidence of 26.57 AEFI per 100,000 doses administered; incidences were 26.42 for RV1 and 26.85 for RV5. Among all RV AEFI, 20,334 (98.92%) were non-serious. Vaccine product-related reactions accounted for 95.68% of AEFI reports, including 18,192 (88.50%) common and 1,476 (7.18%) rare vaccine reactions. Among common vaccine reactions, case reports per 100,000 doses administered were 16.85 (13,031 reports) for fever, 5.84 (4,520 reports) for gastrointestinal disorders, and 1.28 (988 reports) for rash. Among rare vaccine reactions, case reports per 100,000 doses were 1.43 (1,104 reports) for allergic rash, 0.07 (56 reports) for thrombocytopenic purpura, 0.03 (26 reports) for febrile convulsion, and 0.01 (5 reports) for intussusception. Conclusions Most RV AEFIs were mild and non-serious, and the incidence of rare vaccine reactions was very low. RVs have reasonable safety surveillance profiles and AEFI evaluation should be continued.
Collapse
Affiliation(s)
- Yuan Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Keli Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chunxiang Fan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lina Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Minrui Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lei Cao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wenzhou Yu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zundong Yin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
16
|
Dey A, Jackson J, Wang H, Lambert SB, McIntyre P, Macartney K, Beard F. Australia's rotavirus immunisation program: Impact on acute gastroenteritis and intussusception hospitalisations over 13 years. Vaccine 2025; 52:126789. [PMID: 39985966 DOI: 10.1016/j.vaccine.2025.126789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Australia was one of the first countries to include rotavirus vaccines in its National Immunisation Program, in 2007. We compared trends in acute gastroenteritis (AGE) and intussusception-coded hospitalisations over 13-year post-vaccine period against five-year pre-vaccine baseline. METHODS In a descriptive before-after study, incidence of hospitalisations with ICD-code of rotavirus AGE (A08.0), other AGE (K52, A01-A09 excluding A08.0) or intussusception (K56.1) between 2002 and 2020 was calculated using population denominators by age and Indigenous status. We used 2002-2006 as pre-vaccine baseline and calculated Incidence Rate Ratios [IRRs] for 2008-2019 and 2020. FINDINGS In children aged <5 years, mean annual hospitalisation rate/100,000 decreased by 85% for rotavirus-coded AGE, from 248.3 in 2002-2006 to 37.6 (IRR 0.15; 95% CI 0.15-0.16) in 2008-2019 (61.4% for Indigenous children, from 680.2 to 262.2), and 46% for other AGE, from 1274.5 to 689.1 (IRR 0.54; CI 0.54-0.55), decreasing further in 2020 to 6.3 (rotavirus-coded) and 445.0 (other AGE). Rates for rotavirus-coded and other AGE declined in 2008-2019 in those aged 5-<20 years (IRR 0.52; CI 0.49-0.56 and 0.86; CI 0.85-0.87, respectively), but increased in 20-<65 years (IRR 2.38; CI 2.01-2.83 and 1.15; CI 1.15-1.16) and ≥65 years (IRR 2.24; CI 1.91-2.62 and 1.24; CI 1.23-1.25). Average annual hospitalisation rate for intussusception in infants was similar in pre-vaccine and post-vaccine periods (IRR 0.97; CI 0.90-1.04). CONCLUSION Over a 13-year period post-rotavirus vaccine introduction we document major sustained declines in hospitalisations coded as rotavirus and other AGE in age groups <20 years, with no change in intussusception hospitalisation rates in infants. Despite small increases in AGE hospitalisations in adults, likely due to increased PCR testing, our findings are consistent with highly favourable risk benefit ratio at whole-of-population level in Australia.
Collapse
Affiliation(s)
- Aditi Dey
- National Centre for Immunisation Research and Surveillance, The Children's Hospital at Westmead, Sydney, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, Australia.
| | - Joanne Jackson
- National Centre for Immunisation Research and Surveillance, The Children's Hospital at Westmead, Sydney, Australia
| | - Han Wang
- National Centre for Immunisation Research and Surveillance, The Children's Hospital at Westmead, Sydney, Australia
| | - Stephen B Lambert
- National Centre for Immunisation Research and Surveillance, The Children's Hospital at Westmead, Sydney, Australia
| | - Peter McIntyre
- National Centre for Immunisation Research and Surveillance, The Children's Hospital at Westmead, Sydney, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, Australia; University of Otago, New Zealand
| | - Kristine Macartney
- National Centre for Immunisation Research and Surveillance, The Children's Hospital at Westmead, Sydney, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Frank Beard
- National Centre for Immunisation Research and Surveillance, The Children's Hospital at Westmead, Sydney, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| |
Collapse
|
17
|
Wenger C, Asare EO, Kwon J, Li X, Mwinjiwa E, Chinkhumba J, Jere KC, Hungerford D, Cunliffe NA, Paltiel AD, Pitzer VE. Cost-effectiveness analysis of alternative infant and neonatal rotavirus vaccination schedules in Malawi. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004341. [PMID: 40209158 PMCID: PMC11984971 DOI: 10.1371/journal.pgph.0004341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/28/2025] [Indexed: 04/12/2025]
Abstract
Rotavirus is the leading cause of severe diarrhea among children under five worldwide, especially in low- and middle-income countries (LMICs). Although vaccination is the best strategy to prevent rotavirus, obstacles leading to poor vaccine effectiveness undermine its impact in LMICs. This study aimed to identify the optimal rotavirus vaccination strategy for Malawi by modeling vaccine impact and cost-effectiveness, comparing the current two-dose Rotarix vaccine schedule to two alternative vaccine delivery schedules and a next-generation neonatal vaccine (RV3-BB) from 2025-2034. The cost-effectiveness of rotavirus vaccine strategies in Malawi was evaluated from the government and societal perspectives using estimates of moderate-to-severe and non-severe rotavirus cases derived from a transmission dynamic model of rotavirus and published estimates of health-seeking behaviors and costs as inputs. A probabilistic sensitivity analysis was performed to evaluate the robustness of our results to parameter uncertainty. Over a ten-year time horizon, the current two-dose Rotarix strategy is predicted to avert over 1.5 million cases and 90,000 disability-adjusted life-years (DALYs) compared to no vaccination and is cost-effective at willingness-to-pay (WTP) thresholds above $105 per DALY averted from the government perspective. Adding a third dose at 14 weeks could avert an additional 1 million cases and 38,000 DALYs, while switching to the neonatal RV3-BB vaccine could avert 1.1 million cases and 41,000 DALYs compared to the current strategy. Whereas adding a third dose of Rotarix would cost $4.1-4.9 million, switching to the neonatal vaccine is expected to save $3.7 million compared to the current strategy. Considering the neonatal vaccine is not yet available, adding a third dose of Rotarix at 14 weeks of age is cost-effective at WTP thresholds above $138 per DALY averted. The neonatal vaccine offers a more cost-effective alternative to Malawi's current rotavirus vaccine, while adding a third dose to the current strategy also provides substantial benefits.
Collapse
Affiliation(s)
- Catherine Wenger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Ernest O. Asare
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Jiye Kwon
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Xiao Li
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), University of Antwerp, Belgium
| | - Edson Mwinjiwa
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, Merseyside, United Kingdom
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Department of Health Systems and Policy, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Jobiba Chinkhumba
- Department of Health Systems and Policy, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Khuzwayo C. Jere
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, Merseyside, United Kingdom
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Medical Laboratory Sciences, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Daniel Hungerford
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, Merseyside, United Kingdom
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Nigel A. Cunliffe
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, Merseyside, United Kingdom
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - A. David Paltiel
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Department of Health Policy & Management, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Virginia E. Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| |
Collapse
|
18
|
Aman AT, Patriani A, Mawarti Y. Efficacy of rotavirus vaccines in Indonesia: A review of genotype distribution and impact. NARRA J 2025; 5:e1681. [PMID: 40352187 PMCID: PMC12059961 DOI: 10.52225/narra.v5i1.1681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/21/2025] [Indexed: 05/14/2025]
Abstract
Rotavirus remains the leading cause of diarrhea among children under five years of age, with an incidence of 31.1-90.9% in Indonesia. Initially, a rotavirus vaccination program was introduced in several provinces of Indonesia in 2022, which would be conducted nationally. This review provides information on the rotavirus genotype distribution in Indonesia, efficacy and effectiveness data of the rotavirus vaccine, and an update on the status of rotavirus vaccine implementation worldwide. The results show a varied distribution of G and P genotypes from 1978 to 2018, with G1-G3, G9, P[4], P[6], and P[8] as the prevalent genotypes, followed by a small proportion of G4, P[9], P[10], and P[11]. Three rotavirus vaccines, which are prequalified by the World Health Organization (WHO) and available in Indonesia, showed an efficacy of 17.6-76.9% in high-mortality countries. The Indonesian government procured ROTAVAC with a G9P[11] genotype for the national immunization program, which showed 31.3-69.1% protective efficacy against severe gastroenteritis caused by other strains. This review suggested that the decision to choose the rotavirus vaccine for the national program should take into account the country's prevalent circulating genotype and the vaccine's efficacy against severe diarrhea. The use of a pentavalent rotavirus vaccine with high efficacy in high-mortality countries can be regarded as the prime choice for the program. Another alternative is the rotavirus vaccine, which showed efficacy data in multiple high-mortality countries. In addition, regular surveillance of the rotavirus genotypes and the clinical manifestations of diarrhea are necessary to design vaccination strategies in Indonesia.
Collapse
Affiliation(s)
- Abu T. Aman
- Indonesia Research Partnership on Infectious Disease (INA-RESPOND) Site 580 Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
- Department of Microbiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Afifah Patriani
- Indonesia Research Partnership on Infectious Disease (INA-RESPOND) Site 580 Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Yuli Mawarti
- Indonesia Research Partnership on Infectious Disease (INA-RESPOND) Site 580 Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada – Dr. Sardjito Hospital, Yogyakarta, Indonesia
- Department of Clinical Microbiology, Installation of Comprehensive Laboratory, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| |
Collapse
|
19
|
Pan Y, Li Z, Miao Q, Shi H, Guo L, Feng L, Tian J. Phylogenitc analysis and immunogenicity comparison of porcine genotype G9 rotavirus in China from 2020-2023. Virol Sin 2025; 40:176-185. [PMID: 39988297 PMCID: PMC12131029 DOI: 10.1016/j.virs.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
As an emerging genotype, the G9 genotype rotaviruses (RVs) are widespread among humans and pigs, and have been reported in many countries and regions in the recent years. Moreover, porcine G9 strains could cross the interspecies barrier to infect human. To investigate the epidemic trends of porcine G9 strains as well as the cross-immunoreactivity among different isolates, an epidemiological investigation about porcine G9 genotype RVs (PoRVs) was performed during the period 2020-2023 in multiple provinces of China. A total of nine representative strains were identified. The phylogenetic analysis based on viral VP7 gene showed that these strains mainly clustered with lineages III and VI, which revealed the predominant G9 PoRVs in China. Moreover, a new lineage, lineage VII, was identified, and strains of this lineage were found to be circulating in Guangdong and Taiwan. Except lineages I and IV, some isolates from other lineages could co-circulate in pigs and humans. Three G9 strains, namely 923H, 923E, and 923X, which belonged to the largest sub-lineage III, were isolated. Then, the significant cross-reactivity was observed among strains of the same or different lineages. This study is the first to systematically investigate the genetic and immunogenetic characteristics of porcine G9 genotype rotavirus in China, as well as the potential cross-species transmission between pigs and humans, providing a valuable direction for the effective prevention of porcine rotavirus.
Collapse
Affiliation(s)
- Yudi Pan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Zixin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Qian Miao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Hongyan Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Longjun Guo
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| | - Jin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150069, China.
| |
Collapse
|
20
|
Prunas O, Asare EO, Sajewski E, Li Y, Pithawala Z, Weinberger DM, Warren JL, Armah GE, Cunliffe NA, Iturriza-Gómara M, Lopman BA, Pitzer VE. Global estimates of rotavirus vaccine efficacy and effectiveness: a rapid review and meta-regression analysis. EClinicalMedicine 2025; 81:103122. [PMID: 40115174 PMCID: PMC11925534 DOI: 10.1016/j.eclinm.2025.103122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/23/2025] Open
Abstract
Background Rotavirus is the leading cause of diarrhoea worldwide, particularly affecting young children. While national rotavirus immunization programs have reduced rotavirus morbidity and mortality, vaccine performance varies considerably between high-income and low-income settings. Methods We updated a previous systematic review of studies reporting rotavirus vaccine efficacy and vaccine effectiveness against severe rotavirus-associated gastroenteritis (RVGE) by performing a rapid review from July 1, 2020 through October 16, 2024. We included randomized controlled trials reporting vaccine efficacy against severe RVGE and case-control and cohort studies reporting vaccine effectiveness against hospitalization with RVGE in children <5 years old for current internationally licensed vaccines. We developed a meta-regression model for vaccine efficacy and effectiveness using widely available country-specific predictors of rotavirus vaccine performance and simultaneously estimated the relationship between vaccine efficacy and effectiveness. We used the model to predict vaccine efficacy and effectiveness for all countries and assessed its predictive accuracy using a modified leave-one-country-out validation approach. Findings Predicted vaccine efficacy ranged from 69.6% to 94.3% across countries in the Americas, European, and Western Pacific Regions, with a decreased efficacy ranging from 18.6% to 85.3% in the African, South-East Asian, and Eastern Mediterranean regions. Estimates of vaccine effectiveness were generally lower than vaccine efficacy when efficacy was greater than 60%, but effectiveness was predicted to be higher when vaccine efficacy was low. A strong correlation (r = 0.63) was found between the observed and predicted vaccine efficacy and effectiveness, with 98.2% of observed efficacy and effectiveness estimates falling within the 95% prediction intervals. Interpretation Our approach enhances the understanding of global variation in rotavirus vaccine performance and can be used to inform predictions of the potential impact of rotavirus vaccines for countries that have yet to introduce them. Higher-quality data on predictor variables and broader regional representation in vaccine trials are required for more robust vaccine performance estimates. Funding National Institutes of Health/National Institute of Allergy and Infectious Diseases (R01AI112970) and the Bill & Melinda Gates Foundation (INV-17940).
Collapse
Affiliation(s)
- Ottavia Prunas
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Ernest O Asare
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Elizabeth Sajewski
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Yueqi Li
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Zeaan Pithawala
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Daniel M Weinberger
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Joshua L Warren
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - George E Armah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Nigel A Cunliffe
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Miren Iturriza-Gómara
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Virginia E Pitzer
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
21
|
Oliveira Matos AD, Araujo M, Paulino J, Franco FC, Luchs A, Sales-Campos H, Fiaccadori F, Souza M, Silva-Sales M. Mutations in the main antigenic sites of VP7 and VP8* from G3P[8] rotavirus a strains circulating in Brazil may impact immune evasion to rotavirus vaccination. Braz J Microbiol 2025; 56:319-330. [PMID: 39505807 PMCID: PMC11885731 DOI: 10.1007/s42770-024-01542-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024] Open
Abstract
In the post-rotavirus (RVA) vaccination era, uncommon and zoonotic strains have emerged as causative agents of acute gastroenteritis in humans, including the equine-like G3P[8] strains. First identified in 2013, this strain has quickly spread worldwide, reaching the position of the most prevalent genotype in many countries, including Brazil. Here, we report full genotype characterization and phylogenetic analysis of two equine-like G3P[8] strains detected in Goiás, a state in the Cerrado biome of the Brazilian Midwestern region, during the year of 2019. The strains were detected in different socioeconomic and demographic contexts: GO-MR from an asymptomatic adult living in a rural traditional community and GO-H5 from a symptomatic child from the state capital, with access to safe drinking water and essential sanitation services. These strains also displayed different backbone constellations considering the NSP2 gene segment (G3-P[8]-I2-R2-C2-M2-A2-N2-T2-E2-H2 for GO-MR and G3-P[8]-I2-R2-C2-M2-A2-N1-T2-E2-H2 for GO-H5). Furthermore, significant mutations in the main epitope sites of the VP7 and VP8* proteins of the detected strains, and other Brazilian G3P[8] viruses, were found with the comparison to RV1 and RV5 vaccine proteins, indicating a potential ability of these viruses to evade vaccine protection, which may contribute to their prevalence both nationally and globally. In summary, this study corroborates the genetic diversity of equine-like G3P[8] DS-1-like strains circulating worldwide, highlights the epidemiological importance of adults as reservoirs of RVA and shows the substantial differences between these emerging strains and the currently used anti-RVA vaccines, which may partially explain their predominance due to potential evasion of vaccine-induced protection.
Collapse
Affiliation(s)
- Amanda de Oliveira Matos
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
- Laboratory of Mucosal Immunology and Immunoinformatics (LIMIM), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Maísa Araujo
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Jordana Paulino
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Fernanda Craveiro Franco
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Adriana Luchs
- Enteric Diseases Laboratory, Virology Center, Adolfo Lutz Institute, São Paulo, Brazil
| | - Helioswilton Sales-Campos
- Laboratory of Mucosal Immunology and Immunoinformatics (LIMIM), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Fabiola Fiaccadori
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Menira Souza
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil
| | - Marcelle Silva-Sales
- Laboratory of Virology and Cellular Culture (LABVICC), Institute of Tropical Pathology and Public Health, Federal University of Goiás (UFG), Goiânia, Brazil.
| |
Collapse
|
22
|
Erdene E, Munkhjargal O, Batnasan G, Dorjbal E, Oidov B, Byambaa A. Evaluation of Liposome-Encapsulated Vancomycin Against Methicillin-Resistant Staphylococcus aureus. Biomedicines 2025; 13:378. [PMID: 40002791 PMCID: PMC11853440 DOI: 10.3390/biomedicines13020378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Methicillin-resistant Staphylococcus aureus (MRSA) is a global health concern due to its resistance to conventional antibiotics. This study evaluated the efficacy of liposome-encapsulated vancomycin against MRSA using phospholipids extracted from egg yolk. Liposomes were prepared via the freeze-thaw method, yielding vesicles with an average diameter of 157.01 ± 33.04 nm and a polydispersity index (PDI) of 0.0442, indicating uniformity and stability. Antibacterial activity was assessed using the microdilution method. Liposome-encapsulated vancomycin demonstrated complete bacterial growth inhibition (100%) against MRSA ATCC 2758 at dilutions of 101 and 102, compared to only 50% inhibition by free vancomycin at 101. At higher dilutions (103), liposome-encapsulated vancomycin maintained 70% inhibition, whereas free vancomycin was ineffective. In vivo studies using a murine wound infection model revealed that wounds treated with liposome-encapsulated vancomycin achieved superior healing, with complete tissue regeneration observed by day 14. Histological analysis showed reduced inflammation and enhanced tissue recovery in liposome-encapsulated vancomycin-treated groups, compared to fibrosis and persistent necrosis in free vancomycin-treated groups. By enabling sustained drug release and improved bioavailability, liposomal formulations minimized required dosages and systemic toxicity, reducing the risk of resistance development. This study highlights the clinical potential of liposome-encapsulated vancomycin as a scalable, cost-effective treatment for MRSA, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Enkhtaivan Erdene
- Department of Microbiology, Infection Prevention and Control, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia;
- Department of Biomedicine, Etugen University, Ulaanbaatar 14200, Mongolia
| | - Odonchimeg Munkhjargal
- Mongolian Academy of Science, Institute of Chemistry and Chemical Technology, Ulaanbaatar 14200, Mongolia
| | - Galindev Batnasan
- Experimental Animal Center, Institute of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia
| | - Enkhjargal Dorjbal
- Department of Pharmaceutical Chemistry and Pharmacognosy, School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia
| | - Baatarkhuu Oidov
- Department of Infectious Diseases, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia
| | - Ariunsanaa Byambaa
- Department of Microbiology, Infection Prevention and Control, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia;
| |
Collapse
|
23
|
Pun J, Evans C, Chasekwa B, Church JA, Gough E, Mutasa K, Rukobo S, Govha M, Mushayanembwa P, Majo FD, Tavengwa NV, Humphrey JH, Kirkpatrick BD, Kosek M, Ntozini R, Prendergast AJ. Associations Between Histo-blood Group Antigen Status in Mother-Infant Dyads and Infant Oral Rotavirus Vaccine Immunogenicity in Rural Zimbabwe. J Infect Dis 2025; 231:e225-e233. [PMID: 39352457 PMCID: PMC11793023 DOI: 10.1093/infdis/jiae456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/28/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Histo-blood group antigen (HBGA) phenotypes may contribute to poor oral rotavirus vaccine (RVV) immunogenicity, since rotavirus binds intestinal epithelial HBGA glycans, while maternal HBGA status shapes breastmilk composition, which influences the composition of the infant microbiome. We investigated associations between maternal/infant HBGA phenotypes and RVV immunogenicity in rural Zimbabwe. METHODS We undertook salivary FUT2/FUT3 phenotyping in mother-infant pairs. Serum anti-rotavirus immunoglobulin A was measured by enzyme-linked immunosorbent assay. We explored adjusted associations between FUT2/FUT3 status and RVV seroconversion (primary outcome, n = 322) and seropositivity and geometric mean titer (secondary outcomes, n = 776). RESULTS Infants of FUT2- or FUT3-positive women were less likely to seroconvert post-RVV than infants of FUT2- or FUT3-negative women (FUT2 positive [20.1%] vs FUT2 negative [27.5%]: adjusted relative risk [aRR], 0.47; 95% CI, .26-.82; P = .008; FUT3 positive [18.1%] vs FUT3 negative [30.0%]: aRR, 0.45; 95% CI, .25-.78; P = .005). When compared with FUT2-positive infants with FUT2-positive mothers, FUT2-positive infants with FUT2-negative mothers were twice as likely to seroconvert (36.8% vs 21.9%; aRR, 2.12; 95% CI, 1.23-3.63; P = .006). When compared with FUT3-positive infants with FUT3-positive mothers, FUT3-positive infants with FUT3-negative mothers were 3 times as likely to seroconvert (48.3% vs 18.2%; aRR, 2.99; 95% CI, 1.82-4.90; P < .001). CONCLUSIONS Maternal and infant FUT2 and FUT3 status influences infant RVV immunogenicity.
Collapse
Affiliation(s)
- Joshua Pun
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, United Kingdom
| | - Ceri Evans
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, United Kingdom
| | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - James A Church
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ethan Gough
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Sandra Rukobo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Margaret Govha
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume V Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Jean H Humphrey
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Beth D Kirkpatrick
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, College of Medicine, University of Vermont, Burlington
| | - Margaret Kosek
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Andrew J Prendergast
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, United Kingdom
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
24
|
Liu J, Liu G, Wang C, Hu Z, Dahlke HE, Walter MT, Zhang Y, Guo H, Zhang C, Huo Z. Advantages and disadvantages of current human enteric virus surrogates in soils and aquifers. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 963:178497. [PMID: 39827637 DOI: 10.1016/j.scitotenv.2025.178497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/30/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Groundwater is one of the main sources of drinking water, thus, human enteric viruses in groundwater could pose safety risks. Many enteric viruses enter drinking water sources through irrigation or recharge of contaminated water. It is therefore advised to test the potential transport risk with harmless surrogates before wastewater or recycled water is used for irrigation or groundwater recharge. An ideal virus surrogate should be able to mimic the particle size, the surface properties and the inactivation rate of its target virus and should be easy to detect. Particle size should be the first consideration when selecting a suitable virus surrogate in soil and aquifer. The natural bacteriophages could only mimic the viruses that are inherently similar to themselves, and there is only a limited number of readily available bacteriophages. Therefore, once a certain bacteriophage is chosen for study, its particle size, surface properties and inactivation rate are set and unmodifiable for the experiment. Fluorescent microspheres <200 nm could surrogate target viruses in fast-flow subsurface systems, where inactivation can be neglected. However, the current detection limit of fluorescent nanospheres cannot support the detection of small-sized fluorescent microspheres (∼20 nm) through porous media without macropores. Newly emerging DNA tracers not only allow controlling the size and surface properties, but also offer a low detection limit (ideally 1 copy of DNA). Investigating new types of DNA tracers that could either simulate or mimic the inactivation rate of target viruses could widen the use of virus surrogates to study groundwater contamination and drinking water supply safety.
Collapse
Affiliation(s)
- Jiarong Liu
- College of Water Resources and Civil Engineering, China Agricultural University, Beijing 100083, China
| | - Geng Liu
- College of Water Resources and Civil Engineering, China Agricultural University, Beijing 100083, China
| | - Chaozi Wang
- College of Water Resources and Civil Engineering, China Agricultural University, Beijing 100083, China.
| | - Zengjie Hu
- College of Water Resources and Civil Engineering, China Agricultural University, Beijing 100083, China
| | - Helen E Dahlke
- Department of Land, Air, and Water Resources, University of California, Davis, Davis, CA 95616, USA
| | - M Todd Walter
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yuhan Zhang
- College of Water Resources and Civil Engineering, China Agricultural University, Beijing 100083, China
| | - Haoqi Guo
- College of Water Resources and Civil Engineering, China Agricultural University, Beijing 100083, China
| | - Chenglong Zhang
- College of Water Resources and Civil Engineering, China Agricultural University, Beijing 100083, China
| | - Zailin Huo
- College of Water Resources and Civil Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
25
|
Ofosu-Appiah LH, Negoro M, Amexo JX, Amelor DK, Tonto PB, Laryea DO, Yamasaki K, Asiedu-Bekoe F, Sugata K, Hori H, Suganuma N, Taniguchi K. Clinical Impact and Genetic Analysis of Enteric Viruses Associated With Acute Gastroenteritis in Greater Accra, Ghana: A Comprehensive Study of Five Viruses. J Med Virol 2025; 97:e70216. [PMID: 39935201 DOI: 10.1002/jmv.70216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/28/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Enteric viruses are significantly associated with acute gastroenteritis globally. Despite a decrease in severe rotavirus associated diarrhoea, Ghana still records high diarrhoea burden. Meanwhile aetiological investigations in hospital settings do not routinely include viral testing. Rotavirus vaccination is thought to alter enteric viral populations and impact evolution. To better understand virus-specific effects in acute gastroenteritis in both children and adults, we tested fecal samples from 228 patients at two hospitals in Accra from January to December 2019, using multiplex and singleplex PCR assays. The clinical impact of detected viruses was assessed using a modified Vesikari score system. Partial viral genome sequences were obtained by Sanger Sequencing and their genetic diversity and evolutionary history, traced by phylogenetic analyses. At least one enteric virus was found in 86 (37.7%) patient samples, with 36.9% of the population under five infected. Single infections of rotavirus, norovirus, adenovirus, sapovirus and astrovirus were 33, 14, 8, 6, and 1, respectively, while coinfections were 24. Rotavirus accounted for 33.3% of 24 clinically severe cases (modified Vesikari score > 7). Three out of 10 rotavirus cases with evidence of vaccination experienced severe gastroenteritis. Diverse genotypes, including RVA G2P[4], G1P[8], G12P[8] and G12P[6]; AdV F40 and F41; NoV GII.4 Sydney 2012, GII.6 and GI.3, several of which clustered with contemporary strains from the Americas, Europe and Asia, were detected. This study also provides the first report of SaV GI.1, GI.7 and GII.8 detection in humans in Ghana. RVA G2P[4] and AdV F were associated with higher proportions of hospitalizations. While RVA continues to have a profound clinical impact on gastroenteritis, AdV and SaV produce an equally severe disease. In contrast, NoV and AstV showed a generally mild to moderate impact on clinical disease severity.
Collapse
Affiliation(s)
- Lawrence Henry Ofosu-Appiah
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kochi, Japan
- National Public Health and Reference Laboratory, Public Health Division, Ghana Health Service, Accra, Ghana
- Department of Pediatric Infectious Diseases, Institute for Clinical Research, Mie National Hospital Organization, Tsu, Mie, Japan
| | - Manami Negoro
- Department of Pediatric Infectious Diseases, Institute for Clinical Research, Mie National Hospital Organization, Tsu, Mie, Japan
| | - Jennifer Xolali Amexo
- Department of Pediatric Infectious Diseases, Institute for Clinical Research, Mie National Hospital Organization, Tsu, Mie, Japan
| | - Dodzi Kofi Amelor
- National Public Health and Reference Laboratory, Public Health Division, Ghana Health Service, Accra, Ghana
| | - Prince Baffour Tonto
- Department of Pediatric Infectious Diseases, Institute for Clinical Research, Mie National Hospital Organization, Tsu, Mie, Japan
| | - Dennis Odai Laryea
- Disease Surveillance Department, Public Health Division, Ghana Health Service, Accra, Ghana
| | - Keiko Yamasaki
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kochi, Japan
| | - Franklin Asiedu-Bekoe
- Disease Surveillance Department, Public Health Division, Ghana Health Service, Accra, Ghana
| | - Ken Sugata
- Department of Pediatric Infectious Diseases, Institute for Clinical Research, Mie National Hospital Organization, Tsu, Mie, Japan
| | - Hiroki Hori
- Department of Medical Education, Graduate School of Medicine Mie University, Tsu, Mie, Japan
| | - Narufumi Suganuma
- Department of Environmental Medicine, Kochi Medical School, Kochi University, Kochi, Japan
| | - Kiyosu Taniguchi
- Department of Pediatric Infectious Diseases, Institute for Clinical Research, Mie National Hospital Organization, Tsu, Mie, Japan
| |
Collapse
|
26
|
Hubbard S, Wolf J, Oza HH, Arnold BF, Freeman MC, Levy K. Differential Effectiveness of Water, Sanitation, and Handwashing Interventions to Reduce Child Diarrhea in Dry and Rainy Seasons: A Systematic Review and Meta-Analysis of Intervention Trials. ENVIRONMENTAL HEALTH PERSPECTIVES 2025; 133:26001. [PMID: 39903556 PMCID: PMC11793162 DOI: 10.1289/ehp14502] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 11/22/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Studies evaluating how water, sanitation, and/or handwashing (WASH) interventions in low- and middle-income countries impact diarrheal diseases have shown inconsistent results. The prevalence of enteric pathogen infections and diarrhea are highly seasonal and climate-sensitive, which could explain heterogeneous findings. Understanding how season influences the effectiveness of WASH interventions is critical for informing intervention approaches that will be resistant under the varying weather conditions that climate change will bring. METHODS We conducted a systematic review of the literature and meta-analysis to test whether and to what extent the impact of WASH interventions on diarrhea differs by season. We searched the literature for randomized and nonrandomized controlled WASH intervention trials and identified the season in which data were collected-rainy, dry, or both-for each study using proximate land station weather datasets. We compared the relative risk (RR) estimates for the impact of interventions on diarrhea for each study, stratified by season, and analyzed estimates using meta-analysis and meta-regression. This study is registered with PROSPERO, CRD42021231137. RESULTS A total of 50 studies met the inclusion criteria, resulting in 34 drinking water intervention estimates, 8 sanitation intervention estimates, and 14 handwashing intervention estimates. Of the total studies, 60% (n = 30 ) spanned more than one season, with most single-season studies (75%, n = 15 ) occurring exclusively in the dry season. The effect of WASH interventions was stronger in dry seasons than in rainy seasons, with a 33% [95% confidence interval (CI): 24%, 41%] and 18% reduction (95% CI: 5%, 29%) in diarrhea risk, respectively. When stratified by type of intervention, the stronger effect size in dry seasons was consistent for water and handwashing interventions but not for sanitation interventions. CONCLUSIONS Estimates of the seasonal impact of WASH interventions revealed larger effects in the dry season than in the rainy season overall and for water and handwashing interventions in particular. These patterns likely affected previous estimates of intervention effectiveness, which included more dry season estimates. These findings suggest the need to collect data across seasons and report seasonally stratified results to allow for more accurate estimates of the burden of disease impacted by WASH investments and to improve projections of potential impacts of these interventions under future climate conditions. These findings also underscore the need for robust WASH interventions designed to be resistant to seasonal variations in temperature and rainfall now and under future climate change scenarios. https://doi.org/10.1289/EHP14502.
Collapse
Affiliation(s)
- Sydney Hubbard
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Jennyfer Wolf
- Department of Environment, Climate Change and Health, World Health Organization, Geneva, Switzerland
| | - Hemali H. Oza
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Benjamin F. Arnold
- Francis I. Proctor Foundation, University of California, San Francisco, California, USA
| | - Matthew C. Freeman
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Karen Levy
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
27
|
Anzà D, Esposito M, Bertolazzi G, Fallucca A, Genovese C, Maniscalco G, Praticò AD, Scarpaci T, Vitale E, Restivo V. Determinants of Rotavirus Vaccine Acceptance in an Area of Southern Italy with Low Vaccination Coverage: A Case-Control Study by the Health Belief Model Questionnaire. Vaccines (Basel) 2025; 13:63. [PMID: 39852842 PMCID: PMC11769460 DOI: 10.3390/vaccines13010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Rotavirus (RV) is the primary cause of gastroenteritis in children worldwide, contributing significantly to morbidity and mortality, particularly among children under five years of age. The introduction of Rotavirus vaccines (RVV) has markedly reduced RV-related childhood deaths, especially in Europe, where substantial reductions in hospitalizations and disease prevalence have been observed. Despite these advances, RVV uptake in Italy remains below the desired targets, with notable regional disparities. In Sicily, vaccination rates have fluctuated, with current coverage failing to meet national goals. Safety concerns and insufficient parental awareness are major barriers to RVV acceptance. METHODS This case-control study was conducted in Southern Italy to identify factors influencing parental acceptance of RVV. Data were collected from parents using a structured questionnaire that assessed socio-demographic factors, vaccine knowledge, and attitudes based on the Health Belief Model (HBM). RESULTS Overall, 226 parents were enrolled. Higher perceived benefit of RVV was significantly associated with increased vaccine adherence (Odds Ratio = 13.65; 95% Confidence Interval = 6.88-27.09; p < 0.001). CONCLUSIONS These results highlight the need for targeted interventions to improve vaccine coverage and address regional and socio-economic barriers to RVV acceptance. Furthermore, tailored educational campaigns and univocal information from healthcare providers could play pivotal roles in achieving higher vaccine uptake.
Collapse
Affiliation(s)
- Davide Anzà
- Department of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy; (D.A.); (M.E.); (G.B.); (C.G.); (A.D.P.); (E.V.)
| | - Massimiliano Esposito
- Department of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy; (D.A.); (M.E.); (G.B.); (C.G.); (A.D.P.); (E.V.)
| | - Giorgio Bertolazzi
- Department of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy; (D.A.); (M.E.); (G.B.); (C.G.); (A.D.P.); (E.V.)
| | - Alessandra Fallucca
- Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE) “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (A.F.); (G.M.); (T.S.)
| | - Carlo Genovese
- Department of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy; (D.A.); (M.E.); (G.B.); (C.G.); (A.D.P.); (E.V.)
| | - Gabriele Maniscalco
- Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE) “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (A.F.); (G.M.); (T.S.)
| | - Andrea D. Praticò
- Department of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy; (D.A.); (M.E.); (G.B.); (C.G.); (A.D.P.); (E.V.)
| | - Tiziana Scarpaci
- Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE) “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy; (A.F.); (G.M.); (T.S.)
| | - Ermanno Vitale
- Department of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy; (D.A.); (M.E.); (G.B.); (C.G.); (A.D.P.); (E.V.)
| | - Vincenzo Restivo
- Department of Medicine and Surgery, University of Enna Kore, 94100 Enna, Italy; (D.A.); (M.E.); (G.B.); (C.G.); (A.D.P.); (E.V.)
| |
Collapse
|
28
|
Bhat N, Vodicka E, Clifford A, Ananth KB, Bavdekar A, Roy AD, Parashar U, Tate J, Haldar P, Kang G. The evidence base for rotavirus vaccination in India: Current status, future needs. Vaccine 2025; 44:126551. [PMID: 39615343 PMCID: PMC11672240 DOI: 10.1016/j.vaccine.2024.126551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024]
Abstract
Rotavirus is a leading cause of severe diarrheal disease in infants and young children worldwide. Vaccination offers the best protection against this disease, and two rotavirus vaccines were developed in India and included in its routine immunization program. The Government of India's decision to adopt this intervention was supported by a solid base of evidence from clinical trials, as well as substantial research regarding rotavirus disease burden and the potential health and economic value of immunization. Following program implementation, multiple studies were initiated, including three evaluations of effectiveness and several investigations regarding intussusception. These additional data regarding vaccine impact, safety, and delivery from post-introduction evaluations in conditions of real-world use will further strengthen and sustain the immunization program. This manuscript evaluates the status of existing and forthcoming evidence regarding rotavirus vaccination in India through a literature review and consultation with relevant stakeholders. Studies evaluating vaccine impact, effectiveness, safety, health economics, and acceptability, as well as operational and programmatic research, were included in the review. Overall, we found that the evidence base did not contain any major gaps. Nevertheless, additional smaller-scale research studies would be valuable in providing a more complete picture of rotavirus vaccine performance and benefit. Documentation of India's experience with rotavirus vaccines may provide lessons learned for other countries in the Asia region, where rotavirus disease burden remains high, yet vaccine adoption has been slow, as well as for countries worldwide that may be considering implementation of the Indian-made rotavirus vaccines.
Collapse
Affiliation(s)
- Niranjan Bhat
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA.
| | - Elisabeth Vodicka
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA
| | - Allison Clifford
- Center for Vaccine Innovation and Access, PATH, 455 Massachusetts Ave NW, Washington, DC 20001, USA
| | - Kanduri Balaji Ananth
- Center for Vaccine Innovation and Access, PATH, 15th Floor, Dr. Gopal Das Bhawan, 28 Barakhamba Road, New Delhi 110001, India
| | - Ashish Bavdekar
- KEM Hospital Research Centre, Vadu Rural Health Program, P.O. Vadu Budruk, Taluka Shirur, District Pune 412216, India
| | - Arup Deb Roy
- JSI India, Plot No.5 & 6, LSC Shopping Complex, Nelson Mandela Marg, Vasant Kunj, New Delhi 110070, India
| | - Umesh Parashar
- Centers for Disease Control and Prevention, 1600 Clifton Rd. NE, Atlanta, GA 30333, USA
| | - Jacqueline Tate
- Centers for Disease Control and Prevention, 1600 Clifton Rd. NE, Atlanta, GA 30333, USA
| | - Pradeep Haldar
- Former Advisor (RCH), Ministry of Health and Family Welfare, Government of India, Delhi, India
| | - Gagandeep Kang
- Division of Gastrointestinal Sciences, The Wellcome Trust Research Laboratory, Christian Medical College, Vellore, Tamil Nadu 632004, India
| |
Collapse
|
29
|
Zalot MA, Cortese MM, O'Callaghan KP, Casey-Moore MC, L'Etoile N, Smart SL, Honeywood MJ, Mijatovic-Rustempasic S, Tate JE, Davis A, Wittmeyer N, McGann C, Sadaf S, Wilson K, Bowen MD, Gautam R, Parashar UD, Coffin SE, Gibbs KA. Risk of Transmission of Vaccine-Strain Rotavirus in a Neonatal Intensive Care Unit That Routinely Vaccinates. Pediatrics 2025; 155:e2024067621. [PMID: 39652114 DOI: 10.1542/peds.2024-067621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/01/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Many neonatal intensive care units (NICUs) do not give rotavirus vaccines to inpatients due to a theoretical risk of horizontal transmission of vaccine strains. We aimed to determine incidence and clinical significance of vaccine-strain transmission to unvaccinated infants in a NICU that routinely administers pentavalent rotavirus vaccine (RV5). METHODS This prospective cohort study included all patients admitted to a 100-bed NICU for 1 year. Stool specimens were collected weekly; real-time quantitative reverse-transcription polymerase chain reaction was used to detect any RV5 strain. Incidence of transmission to unvaccinated infants was calculated assuming each unvaccinated patient's stool contributed 1 patient-day at risk for transmission. Investigations and geospatial analyses were conducted for suspected transmission events. RESULTS Of 1238 infants admitted, 560 (45%) were premature and 322 (26%) had gastrointestinal pathology. During observation, 226 RV5 doses were administered. Overall, 3448 stool samples were tested, including 2252 from 686 unvaccinated patients. Most (681, 99.3%) unvaccinated patients never tested positive for RV5 strain. Five (<1%) tested RV5 strain positive. The estimated rate of transmission to unvaccinated infants was 5/2252 stools or 2.2/1000 patient-days at risk (95% CI: 0.7-5.2). No gastroenteritis symptoms were identified in transmission cases within 7 days of collection of RV5-positive stool. Of 126 patients for whom the RV5 series was initiated before the discharge date, 55% would have become age-ineligible to start the series if vaccination was allowed only at discharge. CONCLUSIONS Transmission of RV5 strain was infrequent and without clinical consequences. Benefits of allowing vaccine-induced protection against rotavirus disease in infants through in-NICU RV5 vaccination appear to have outweighed risks from vaccine-strain transmission.
Collapse
Affiliation(s)
- Morgan A Zalot
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| | - Margaret M Cortese
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kevin P O'Callaghan
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
- Current affiliation: Centers for Disease Control and Prevention
| | - Mary C Casey-Moore
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Nathan L'Etoile
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| | - Sarah Leeann Smart
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Michelle J Honeywood
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Slavica Mijatovic-Rustempasic
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jacqueline E Tate
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Anna Davis
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| | - Nicole Wittmeyer
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| | - Carolyn McGann
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| | - Salma Sadaf
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| | - Kadedra Wilson
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| | - Michael D Bowen
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Rashi Gautam
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Umesh D Parashar
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Susan E Coffin
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| | - Kathleen A Gibbs
- Department of Pediatrics, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania
| |
Collapse
|
30
|
Thobari JA, Watts E, Carvalho N, Haposan JH, Clark A, Debellut F, Mulyadi AWE, Sundoro J, Nadjib M, Hadinegoro SR, Bines J, Soenarto Y. Cost effectiveness analysis of rotavirus vaccination in Indonesia. Vaccine 2025; 43:126478. [PMID: 39500219 DOI: 10.1016/j.vaccine.2024.126478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Rotavirus (RV) remains the most common cause of morbidity and mortality due to acute gastroenteritis (AGE) in children under five. In Indonesia, RV is responsible for 60 % of severe AGE and 40 % of non-severe AGE in these children. This study assessed the cost-effectiveness of introduction of rotavirus vaccines (RVV) into the National Immunization Program in Indonesia. METHODS We conducted a cost-effectiveness analysis (CEA) of RVV introduction in Indonesia, assuming a three-dose vaccine schedule based on the planned introduction proposed by the Strategic Advisory Group of Experts on Immunization. The analysis involved an initial introduction of an imported RVV (Rotavac®, Bharat Biotech, India) followed by a staged implementation of the locally produced RVV (Bio Farma, Indonesia) from both health system and societal perspectives. The primary outcome measure was the incremental cost (2019 USD) per disability-adjusted life year (DALY) averted, compared to no vaccination. We took model inputs from an Indonesian cost-of-illness study, national information systems and scientific literature, covering disease incidence, hospitalization, mortality, healthcare costs, and vaccine related factors. Our analyses included univariate and probabilitistic sensitivity analyses to assess various parameters. FINDINGS The cost of a 10-year vaccination program is 82.6 million USD and can potentially prevent 7.3 million cases of rotavirus and 0.42 million DALYs. From a societal perspective, the incremental cost-effectiveness ratio (ICER) for the staged program is 464 USD per DALY averted (12 % of Indonesia's gross domestic product (GDP) per capita). From a healthcare sector perspective, ICER is similar at 479 USD (13 % GDP per capita). INTERPRETATION The introduction of RVV into the National Immunization Program is likely to be highly cost-effective in Indonesia. FUNDING This work was supported by funding agreement with the Murdoch Children's Research Institute (MCRI), PATH, and the Indonesian Technical Advisory Group on Immunization (ITAGI).
Collapse
Affiliation(s)
- Jarir At Thobari
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia; Center for Child Health, Pediatric Research Office (CCH/PRO), Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Emma Watts
- Enteric Diseases Research Group, Infection, Immunity, and Global Health, Murdoch Children's Research Institute, Parkville, Australia
| | - Natalie Carvalho
- School of Population and Global Health, University of Melbourne, Parkville, Australia
| | - Jonathan Hasian Haposan
- Center for Child Health, Pediatric Research Office (CCH/PRO), Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia; Enteric Diseases Research Group, Infection, Immunity, and Global Health, Murdoch Children's Research Institute, Parkville, Australia; Department of Biostatistics, Epidemiology, and Population Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Andrew Clark
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | - Asal Wahyuni Erlin Mulyadi
- Center for Child Health, Pediatric Research Office (CCH/PRO), Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia; Faculty of Social and Political Sciences Universitas Sebelas Maret Surakarta, Central Java, Indonesia
| | - Julitasari Sundoro
- Health Economic Working Group, Indonesia Task Advisory Group for Immunization (ITAGI), Indonesia
| | - Mardiati Nadjib
- Health Economic Working Group, Indonesia Task Advisory Group for Immunization (ITAGI), Indonesia
| | - Sri Redzeki Hadinegoro
- Health Economic Working Group, Indonesia Task Advisory Group for Immunization (ITAGI), Indonesia
| | - Julie Bines
- Enteric Diseases Research Group, Infection, Immunity, and Global Health, Murdoch Children's Research Institute, Parkville, Australia; Department of Pediatrics, University of Melbourne, Parkville, Australia
| | - Yati Soenarto
- Center for Child Health, Pediatric Research Office (CCH/PRO), Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
31
|
Han S, Song MS, Song H, Yu J, Choi C, Park SH, Ha SD. Control of rotavirus by sequential stress of disinfectants and gamma irradiation in leafy vegetable industry. Food Res Int 2025; 200:115456. [PMID: 39779116 DOI: 10.1016/j.foodres.2024.115456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
Rotavirus (RV) causes severe gastroenteritis in infants and young children worldwide. Fresh produce has been reported as a source of RV infection during production and harvesting, leading to foodborne illness. Cases of contamination from contact surfaces have also been reported. Therefore, this study applied chemical methods (chlorine dioxide [ClO2], peracetic acid [PAA]), physical methods (gamma irradiation), and a combination of methods (disinfectants + gamma irradiation) to inactivate RV on food contact surfaces (stainless steel) and food (lettuce). Furthermore, the changes in food quality after the combined treatments were assessed. The results of the chemical treatment showed that RV was reduced below the detection limit after treatment for 1 min with 20 ppm ClO2 or 120 ppm PAA in RV suspension. On stainless steel, treatment with 200 ppm ClO2 or 2,000 ppm PAA reduced contaminated RV by more than 4 log. A 5 min treatment with 50 ppm ClO2 or 80 ppm PAA on lettuce reduced the RV by 1.79 and 0.75 log, respectively. Treatment with 4 kGy of gamma irradiation resulted in more than 5 log reduction in suspensions and 3.27 log reduction on food. The sequential treatments, including 30 ppm ClO2 followed by 1.5 kGy gamma irradiation and 80 ppm PAA followed by 1.5 kGy gamma irradiation, showed additional inactivation effects (p < 0.05) compared to each single treatment. No changes in food quality (color difference and texture) were observed after any treatments, suggesting that the combined treatment of both ClO2 and gamma irradiation and PAA and gamma irradiation can be applied in the fresh food industry to reduce RV contamination.
Collapse
Affiliation(s)
- Sangha Han
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Min Su Song
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Hyewon Song
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Jisu Yu
- Lotte R&D Center, 201 Magokjungang-ro, Sangsoe-gu, Seoul, Republic of Korea
| | - Changsun Choi
- Department of Food and Nutrition, School of Food Science and Technology, Chung-Ang University, Anseong, Gyeonggi 17546, Republic of Korea
| | - Si-Hong Park
- Department of Food Science & Technology, Oregon State University, Corvallis, OR, USA
| | - Sang-Do Ha
- Department of Food Safety and Regulatory Science, Advanced Food Safety Research Group, Chung-Ang University, Anseong-si, Gyeonggi-do 17546, Republic of Korea.
| |
Collapse
|
32
|
Zhang Y, Hossain MI, Yeo D, Niu T, Hwang S, Yoon D, Lim DJ, Wang Z, Jung S, Kwon H, Choi C. Impact of storage temperature and ultraviolet irradiation on rotavirus survival on food matrices. Food Res Int 2025; 200:115454. [PMID: 39779111 DOI: 10.1016/j.foodres.2024.115454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
This study investigated the survival of human rotavirus (HRV) on fresh beef, chicken, and lettuce stored at various temperatures, as well as the effect of UV-C exposure on HRV viability on these food surfaces. At 20 °C, the survival rate of three HRV strains (WA, 89-12C2, and DS-1) on beef, chicken, and lettuce decreased within 3 days, with the most significant reduction observed on beef. When stored at 4 °C, a significant reduction in HRV viability was observed by day 7, with the greatest decrease observed on beef, followed by chicken and lettuce. Conversely, storage at -20 °C for up to 28 days did not significantly reduce HRV viability on any of the food surfaces. Exposure to UV-C irradiation at a dosage of 100 mJ/cm2 reduced the viral titers on beef and chicken surfaces by approximately 1 log10 PFU/mL, while those on the surfaces of lettuce were more than 4 log10 PFU/mL. These findings indicate that HRV strains exhibit strong viability on beef, chicken, and lettuce surfaces, enduring extended periods at low temperatures, but display varying susceptibility to UV-C irradiation. Due to the persistence of HRV on contaminated food, implementing effective measures to prevent food contamination is crucial. The findings of this study contribute to the development of a robust sanitation strategy utilizing UV-C to mitigate foodborne HRV transmission.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Md Iqbal Hossain
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Daseul Yeo
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Teng Niu
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Seongwon Hwang
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Danbi Yoon
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Dong Jae Lim
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Zhaoqi Wang
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Soontag Jung
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Hyojin Kwon
- Department of Food Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea
| | - Changsun Choi
- Department of Food and Nutrition, College of Biotechnology and Natural Resources, Chung-Ang University, 4726, Gyeonggi-do 17546, Republic of Korea.
| |
Collapse
|
33
|
Chen S, Ying Z, Liu Y, Li Y, Yu Y, Huang M, Huang Z, Ou Z, Liao Y, Zhang Y, Liu G, Zhao W, Fu R, Shou Q, Zheng M, Liao X, Tu Y, Stek J, Hartzel J, Li C, Zhang J. A phase 3 randomized, open-label study evaluating the immunogenicity and safety of concomitant and staggered administration of a live, pentavalent rotavirus vaccine and an inactivated poliomyelitis vaccine in healthy infants in China. Hum Vaccin Immunother 2024; 20:2324538. [PMID: 38509699 PMCID: PMC10962606 DOI: 10.1080/21645515.2024.2324538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
This open-label, randomized, phase 3 study in China (V260-074; NCT04481191) evaluated the immunogenicity and safety of concomitant and staggered administration of three doses of an oral, live, pentavalent rotavirus vaccine (RV5) and three doses of an intramuscular, inactivated poliomyelitis vaccine (IPV) in 400 healthy infants. The primary objective was the non-inferiority of neutralizing antibody (nAb) responses in the concomitant- versus the staggered-use groups. Antibody responses were measured at baseline and 1-month post-dose 3 (PD3). Parents/legal guardians recorded adverse events for 30 or 15 d after study vaccinations in the concomitant-use or staggered-use groups, respectively. At PD3, >98% of participants seroconverted to all three poliovirus types, and the primary objective was met as lower bounds of the two-sided 95% CI for between-group difference in nAb seroconversion percentages ranged from - 4.3% to - 1.6%, for all poliovirus types, p < .001. At PD3, geometric mean titers (GMTs) of nAb responses to poliovirus types 1, 2, and 3 in the concomitant-use group and the staggered-use group were comparable; 100% of participants had nAb titers ≥1:8 and ≥1:64 for all poliovirus types. Anti-rotavirus serotype-specific IgA GMTs and participants with ≥3-fold rise in postvaccination titers from baseline were comparable between groups. Administration of RV5 and IPV was well tolerated with comparable safety profiles in both groups. The immunogenicity of IPV in the concomitant-use group was non-inferior to the staggered-use group and RV5 was immunogenic in both groups. No safety concerns were identified. These data support the concomitant use of RV5 and IPV in healthy Chinese infants.
Collapse
Affiliation(s)
- Shaomin Chen
- Biological Products Surveillance and Evaluation, Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, Guangdong, China
| | - Zhifang Ying
- Respiratory Virus Vaccine, National Institutes for Food and Drug Control, Beijing, China
| | - Yan Liu
- Division of Hepatitis Virus and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Yuan Li
- Biological Products Surveillance and Evaluation, Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, Guangdong, China
| | - Yebin Yu
- Vaccine Clinical Research Office, Yangchun Center for Disease Control and Prevention, Yangchun, Guangdong, China
| | - Meilian Huang
- Vaccine Clinical Research Office, Yangchun Center for Disease Control and Prevention, Yangchun, Guangdong, China
| | - Zhuhang Huang
- Biological Products Surveillance and Evaluation, Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, Guangdong, China
| | - Zhiqiang Ou
- Biological Products Surveillance and Evaluation, Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, Guangdong, China
| | - Yuyi Liao
- Biological Products Surveillance and Evaluation, Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, Guangdong, China
| | - Yong Zhang
- Division of Hepatitis Virus and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China
| | - Guixiu Liu
- Clinical Research, MSD Research and Development (China) Co. Ltd., Beijing, China
| | - Weiwei Zhao
- Biostatistics and Research Decision Sciences, MSD Research and Development (China) Co. Ltd., Beijing, China
| | - Rong Fu
- Biostatistics and Research Decision Sciences, MSD Research and Development (China) Co. Ltd., Beijing, China
| | - Qiong Shou
- Biostatistics and Research Decision Sciences, MSD Research and Development (China) Co. Ltd., Beijing, China
| | - Minghuan Zheng
- Clinical Research, MSD Research and Development (China) Co. Ltd., Beijing, China
| | - Xueyan Liao
- Clinical Research, MSD Research and Development (China) Co. Ltd., Beijing, China
| | - Yingmei Tu
- Infectious Diseases/Vaccines Clinical Research, Merck & Co. Inc., Rahway, NJ, USA
| | - Jon Stek
- Infectious Diseases/Vaccines Clinical Research, Merck & Co. Inc., Rahway, NJ, USA
| | - Jonathan Hartzel
- Biostatistics and Research Decision Sciences, Merck & Co. Inc., Rahway, NJ, USA
| | - Changgui Li
- Institute for Control of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jikai Zhang
- Directors Office, Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Oluwaseun S, Yang C, Si Tu SJ, Yin J, Song Y, Sun Q, Kanibir N, Hartwig S, Carias C. Health impact of rotavirus vaccination in China. Hum Vaccin Immunother 2024; 20:2386750. [PMID: 39269780 PMCID: PMC11404606 DOI: 10.1080/21645515.2024.2386750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 09/15/2024] Open
Abstract
Rotavirus (RV) vaccines have demonstrated substantial effectiveness in reducing the healthcare burden caused by gastroenteritis (RVGE) worldwide. This study aims to understand the differential impact of RV vaccination in reducing RVGE burden in children under 7 years old in China. A Markov Model was used to investigate the health impact of introducing two different RV vaccines into the Chinese population. The analysis was conducted for RV5, a live pentavalent human-bovine reassortant vaccine, and Lanzhou Lamb RV (LLR), a live-attenuated monovalent RV vaccine, separately, by comparing the strategy of each vaccine to no vaccination within a Chinese birth cohort, including 100,000 children modeled until 7 years of age. The vaccination scenario assumed a vaccination coverage of 2.5%, 2.5%, 90% and 5% for doses one, two, three and no vaccine, respectively, for both vaccines. Strategies with RV5, LLR, and no vaccination were associated with 9,895, 49,069, and 64,746 symptomatic RV infections, respectively. RV5 and LLR were associated with an 85% and 24% reduction in the total symptomatic RV infections, respectively, suggesting that the health benefits of RV5 are at least three-fold greater than those associated with the LLR. Further, strategies with RV5 and LLR resulted in an estimated 206 and 59-year increase in quality-adjusted life years (QALYs), respectively. Sensitivity and scenario analyses supported the robustness of the base-case findings. Use of RV vaccine is expected to improve RV-associated health outcomes and its adoption will help alleviate the burden of RVGE in China. RV5 use will result in significantly better health outcomes.
Collapse
Affiliation(s)
| | | | | | - Jia Yin
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, China
| | - Yan Song
- HEOR, Epidemiology & Market Access, Analysis Group, Boston, MA, USA
| | - Qiang Sun
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, China
| | - Nabi Kanibir
- Global Medical and Scientific Affairs, MSD International GmBH, Luzern, Switzerland
| | | | | |
Collapse
|
35
|
Workneh BS, Mekonen EG, Zegeye AF, Gonete AT, Alemu TG, Tamir TT, Tekeba B, Wassie M, Kassie AT, Ali MS. Rotavirus vaccine dose-two dropout and its associated factors among children who received rotavirus vaccine dose-one in Sub-Saharan African countries: A multilevel analysis of the recent demographic and health survey. Hum Vaccin Immunother 2024; 20:2335730. [PMID: 38575525 PMCID: PMC10996828 DOI: 10.1080/21645515.2024.2335730] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
Rotavirus is the most common cause of diarrhea in children worldwide. In 2016, rotavirus infection resulted in 258 173 300 episodes of diarrhea and 128 500 child deaths in the globe. The study aimed to assess the magnitude of Rotavirus vaccine dose-two dropout and associated factors among children who received rotavirus vaccine dose-one in sub-Saharan African countries. The appended and most recent demographic and health survey (DHS) dataset of 17 sub-Saharan African countries was used for data analysis. A total of 73,396 weighted samples were used. Factors associated with the outcome variable were considered significant if their p-values were ≤ .05 in the multilevel mixed-effect logistic regression model. The overall Rotavirus vaccine dose-two dropouts was 10.77% (95% CI 10.55%, 11.00%), which ranged from 2.77% in Rwanda to 37.67% in Uganda. Being younger, late birth order, having difficulty accessing health facilities, having no media exposure, having no work, having home delivery, having no antenatal follow-up, and having no postnatal checkup were factors significantly associated with the outcome variable. The overall Rotavirus vaccine dose-two dropout was higher in sub-Saharan African countries which implies that vaccine dropout is still a great issue in the region. Special attention should be given to those mothers who are young, who have no work, who give birth at home, who experienced difficulty in accessing health facilities, and late birth orders. Furthermore, targeted interventions should be considered for improving access and utilization of media, antenatal care, and postnatal care services.
Collapse
Affiliation(s)
- Belayneh Shetie Workneh
- Department of Emergency and Critical Care Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Enyew Getaneh Mekonen
- Department of Surgical Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Alebachew Ferede Zegeye
- Department of Medical Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Almaz Tefera Gonete
- Department of Pediatrics and Child Health Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tewodros Getaneh Alemu
- Department of Pediatrics and Child Health Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tadesse Tarik Tamir
- Department of Pediatrics and Child Health Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Berhan Tekeba
- Department of Pediatrics and Child Health Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mulugeta Wassie
- School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Alemneh Tadesse Kassie
- Department of Clinical Midwifery, School of Midwifery, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mohammed Seid Ali
- Department of Pediatrics and Child Health Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
36
|
Omar M, Kassem E, Anis E, Abu-Jabal R, Mwassi B, Shulman L, Cohen D, Muhsen K. Factors associated with antibiotic use in children hospitalized for acute viral gastroenteritis and the relation to rotavirus vaccination. Hum Vaccin Immunother 2024; 20:2396707. [PMID: 39248509 PMCID: PMC11385160 DOI: 10.1080/21645515.2024.2396707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/29/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Evidence on unnecessary antibiotic use in children with acute viral gastroenteritis (AGE) is scarce. We characterized the extent and correlates of antibiotic use among children hospitalized with viral AGE. A single-center study enrolled children aged 0-59 months hospitalized for AGE between 2008 and 2015 in Israel. Information was collected on laboratory tests, diagnoses, antibiotic treatment, and rotavirus vaccination. Stool samples were tested for rotavirus antigen, GII-norovirus, and stool cultures were performed for bacterial enteropathogens. Data from 2240 children were analyzed. Rotavirus vaccine was given to 79% of eligible children. Rotavirus test was performed on 1419 (63.3%) children. Before the introduction of universal rotavirus vaccination (2008-2010), rotavirus positivity in stool samples was 37.0%, which declined to 17.3% during the universal vaccination years (2011-2015). Overall, 1395 participants had viral AGE. Of those, 253 (18.1% [95% CI 16.1-20.2]) had unnecessary antibiotic treatment, mostly penicillin 46.6%, ceftriaxone 34.0% and azithromycin 21.7%. A multivariable analysis showed an inverse association between rotavirus vaccination and unnecessary antibiotic treatment (odds ratio = 0.53 [95% CI 0.31-0.91]), while positive associations were found with performing chest-X-ray test (3.00 [1.73-5.23]), blood (3.29 [95% CI 1.85-5.86]) and urine cultures (7.12 [3.77-13.43]), levels of C-reactive protein (1.02 [1.01-1.02]) and leukocytes (1.05 [1.01-1.09]). The results were consistent in an analysis of children with laboratory-confirmed rotavirus or norovirus AGE, or after excluding children with CRP > 50 mg/L. In conclusion, antibiotic prescription was common among hospitalized children with viral AGE, which was inversely related to rotavirus vaccination, possibly due to less severe illness in the vaccinated children.
Collapse
Affiliation(s)
- Muna Omar
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Eias Kassem
- Department of Pediatrics, Hillel Yaffe Medical Center, Hadera, Israel
| | - Emilia Anis
- Division of Epidemiology, Ministry of Health, Jerusalem, Israel
| | - Roula Abu-Jabal
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Basher Mwassi
- Department of Pediatrics, Hillel Yaffe Medical Center, Hadera, Israel
| | - Lester Shulman
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Central Virology Laboratory, Ministry of Health, Ramat Gan, Israel
| | - Dani Cohen
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Khitam Muhsen
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
37
|
Light PM, Singh NS, Alhaffar M, Allison LE, Mounier-Jack S, Ratnayake R, Checchi F, Abdelmagid N. Decision-making for childhood vaccination in crisis settings: a survey of practice & barriers. Confl Health 2024; 18:77. [PMID: 39716298 DOI: 10.1186/s13031-024-00638-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Children, particularly those who have received no routine vaccinations (zero-dose children), are at high risk of vaccine-preventable diseases in humanitarian crisis settings. However, the decision-making processes underlying vaccine intervention design and delivery in such settings are poorly understood. The present study investigated the decision-making practices of organisations involved in childhood vaccination in humanitarian crisis settings globally via an online survey. METHODS Individuals involved in the design or delivery of childhood vaccination programmes in humanitarian crisis settings were invited to fill out a self-administered online survey. Respondents were asked about factors influencing intervention design and vaccine delivery; use of technical guidance, specifically the WHO decision-making framework for vaccination in acute humanitarian emergencies (WHO Framework); and practices for reaching zero-dose children. RESULTS Fourteen responses were received. Large international organisations and UN agencies were overrepresented in the sample. Technical guidance was considered of high importance when designing vaccine interventions. However, the WHO Framework is not available in relevant languages and has not been well-distributed to local and national actors. Awareness of initiatives to reach zero-dose children was high within our sample, though this may not accurately reflect global awareness. Security and resource availability were key barriers to vaccine delivery and reaching zero-dose children. Problems with vaccine access in our sample pertained primarily to issues with the procurement system rather than vaccine cost. CONCLUSIONS The WHO Framework should be provided in more languages, and vaccination actors at local and national level should be engaged to improve its practicality and increase awareness of its aims. In order to reach zero-dose children, vaccines must be made available for use in expanded age groups, which is sometimes not currently feasible within the Gavi/UNICEF procurement system. Clarifying this policy would allow relevant organisations to reach more zero-dose children. Additionally, security is a key barrier impeding vaccine delivery, including for zero-dose children. Safe operational space for humanitarian actors in conflict must be maintained and global conflict resolution mechanisms improved.
Collapse
Affiliation(s)
- Page M Light
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, International Health London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Neha S Singh
- Department of Global Health and Development, Faculty of Public Health and Policy, School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Mervat Alhaffar
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, International Health London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
- Syria Research Group (SyRG), Co-Hosted Between London School of Hygiene & Tropical Medicine, National University of Singapore Saw Swee Hock School of Public Health, Singapore, Singapore
| | - Lauren E Allison
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, International Health London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Sandra Mounier-Jack
- Department of Global Health and Development, Faculty of Public Health and Policy, School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Ruwan Ratnayake
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, International Health London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Francesco Checchi
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, International Health London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Nada Abdelmagid
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, International Health London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.
| |
Collapse
|
38
|
Kostanić V, Kunić V, Prišlin Šimac M, Lolić M, Sukalić T, Brnić D. Comparative Insights into Acute Gastroenteritis in Cattle Caused by Bovine Rotavirus A and Bovine Coronavirus. Vet Sci 2024; 11:671. [PMID: 39729011 DOI: 10.3390/vetsci11120671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Acute gastroenteritis (AGE) in cattle significantly impacts the economy due to relatively high morbidity and mortality and decreased production. Its multifactorial nature drives its global persistence, involving enteric viruses, bacteria, protozoa, and environmental factors. Bovine Rotavirus A (BoRVA) and bovine coronavirus (BCoV) are among the most important enteric RNA viruses causing AGE in cattle. These viruses infect intestinal enterocytes, leading to cell damage and consequently to malabsorption and diarrhea. BoRVA primarily affects calves under 14 days old with gastrointestinal clinical signs, while BCoV affects all ages, causing gastrointestinal and respiratory distress. The economic impact of BoRVA and BCoV, along with their interspecies transmission potential, warrants attention. This concise review discusses the molecular structure, epidemiology, pathogenesis, clinical signs, diagnosis, treatment, and preventive measures of BoRVA and BCoV while providing a comparative analysis. By offering practical guidance on managing such viral infections in cattle, these comparative insights may prove valuable for veterinarians in clinical practice.
Collapse
Affiliation(s)
- Vjekoslava Kostanić
- Department of Virology, Croatian Veterinary Institute, 10000 Zagreb, Croatia
| | - Valentina Kunić
- Department of Virology, Croatian Veterinary Institute, 10000 Zagreb, Croatia
| | | | - Marica Lolić
- Laboratory for Diagnostics, Croatian Veterinary Institute, 32100 Vinkovci, Croatia
| | - Tomislav Sukalić
- Laboratory for Diagnostics, Croatian Veterinary Institute, 48260 Križevci, Croatia
| | - Dragan Brnić
- Department of Virology, Croatian Veterinary Institute, 10000 Zagreb, Croatia
| |
Collapse
|
39
|
Vita D, Lemos M, Neto Z, Evans M, Francisco NM, Fortes F, Fernandes E, Cunha C, Istrate C. High Detection Rate of Rotavirus Infection Among Children Admitted with Acute Gastroenteritis to Six Public Hospitals in Luanda Province After the Introduction of Rotarix ® Vaccine: A Cross-Sectional Study. Viruses 2024; 16:1949. [PMID: 39772256 PMCID: PMC11680217 DOI: 10.3390/v16121949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Rotavirus group A (RVA) is a major cause of pediatric acute gastroenteritis (AGE). Vaccination is an effective public health strategy and Angola implemented it in 2014. This hospital-based study aimed to estimate the prevalence of RVA infection and the severity of AGE in children under five years of age treated at six hospitals in Luanda Province. Between April 2021 and May 2022, 1251 fecal samples were screened by an immunochromatographic rapid test (SD Bioline). Data on socio-demographic profile, nutritional status, and clinical assessment were obtained. The association of RVA infection and AGE severity with possible risk factors was evaluated with a binary logistic regression model. Overall, the detection rate was 57.8% and girls tend to be more often infected than boys (55.2%). Infection was more common in the youngest group (1 to 6 months, 60.3%). Important sources of RVA infection were drinking water kept in tanks (57.9%) and private sanitary facilities with piped water (61%). Surprisingly, according to the Vesikari Scale score, the most severe symptoms were observed in children vaccinated with two doses (80.7%). RVA prevalence remains high despite vaccination, and further studies should address the association between infection sources and disease severity, as well as the causes underlying vaccine (un)effectiveness.
Collapse
Affiliation(s)
- Dikudila Vita
- Faculty of Medicine, Agostinho Neto University, Luanda P.O. Box 116, Angola (M.L.); (E.F.)
| | - Manuel Lemos
- Faculty of Medicine, Agostinho Neto University, Luanda P.O. Box 116, Angola (M.L.); (E.F.)
| | - Zoraima Neto
- National Institute for Health Research, Luanda P.O. Box 3635, Angola
| | - Mathebula Evans
- School of Health Systems and Public Health, Faculty of Health Science, University of Pretoria, Pretoria 0084, South Africa;
| | | | - Filomeno Fortes
- Global Health and Tropical Medicine (GHTM), Institute of Hygiene and Tropical Medicine (IHMT), NOVA University (UNL), 1349-008 Lisbon, Portugal; (F.F.); (C.C.)
| | - Ema Fernandes
- Faculty of Medicine, Agostinho Neto University, Luanda P.O. Box 116, Angola (M.L.); (E.F.)
| | - Celso Cunha
- Global Health and Tropical Medicine (GHTM), Institute of Hygiene and Tropical Medicine (IHMT), NOVA University (UNL), 1349-008 Lisbon, Portugal; (F.F.); (C.C.)
| | - Claudia Istrate
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Interdisciplinary Center for Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal
| |
Collapse
|
40
|
Kanai Y, Kotaki T, Sakai S, Ishisaka T, Matsuo K, Yoshida Y, Hirai K, Minami S, Kobayashi T. Rapid production of recombinant rotaviruses by overexpression of NSP2 and NSP5 genes with modified nucleotide sequences. J Virol 2024; 98:e0099624. [PMID: 39494903 PMCID: PMC11650980 DOI: 10.1128/jvi.00996-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024] Open
Abstract
Reverse genetics systems for rotaviruses (RV) facilitate the generation of genetically engineered RVs by transfection of 11 plasmids encoding 11 genomic viral RNA segments. In addition to viral genome expression, overexpression of NSP2 and NSP5 has been used to increase the rescue efficiency of recombinant RVs. Here, we showed that the overexpression of nucleotide sequence-modified NSP2 and NSP5 enabled the rapid and efficient production of recombinant RVs. Using improved reverse genetics, we established a reverse genetics system for human and bovine RV clinical isolates, as well as laboratory strains of bovine RV (NCDV and UK) and porcine RV (Gottfried). In addition, we rescued low-replicating recombinant RVs carrying a mutant NSP4 lacking the double-layered particle-binding domain, which was deficient in the efficient production of mature virions. These advancements in reverse genetics enabled the generation of molecular clones of RV clinical isolates and recombinant RVs harboring critical amino acid mutations, offering a versatile platform for investigating RV biology and pathogenesis.IMPORTANCERecombinant rotavirus (RV) synthesis via reverse genetics relies on both the viral propagation capacity and the efficiency of the experimental system. Since the establishment of our reverse genetics system, several enhancements have been implemented to augment the rescue efficiency. Nevertheless, challenges persist in generating RV clinical strains and recombinant viruses with low replication capacities. Notably, this improved reverse genetics system successfully facilitated the establishment of molecular clones of human and bovine RV clinical isolates. Fecal samples from patients with RV typically harbor quasi-species or, occasionally, multiple genotypes of RV. In the present study, we performed the genetic sequencing of clinical viral strains during the early propagation stages in cultured cells. Subsequently, infectious viruses were synthesized, allowing the characterization of circulating viruses in nature. This approach provides valuable insights into the genetic diversity and dynamics of RV populations and contributes to a more comprehensive understanding of viral pathogenesis and evolution.
Collapse
Affiliation(s)
- Yuta Kanai
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tomohiro Kotaki
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Satoko Sakai
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Toshie Ishisaka
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kayoko Matsuo
- Kumamoto Prefectural Aso Livestock Hygiene Service Center, Aso, Japan
| | - Yukino Yoshida
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Katsuhisa Hirai
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shohei Minami
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| |
Collapse
|
41
|
Peng R, Wang M, Shahar S, Xiong G, Zhang Q, Pang L, Wang H, Kong X, Li D, Duan Z. Epidemiological, molecular, and evolutionary characteristics of G1P[8] rotavirus in China on the eve of RotaTeq application. Front Cell Infect Microbiol 2024; 14:1453862. [PMID: 39717546 PMCID: PMC11666228 DOI: 10.3389/fcimb.2024.1453862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/05/2024] [Indexed: 12/25/2024] Open
Abstract
Introduction This study, conducted in China prior to RotaTeq's launch, examined the epidemiological, molecular, and evolutionary features of the G1P[8] genotype RVA in children admitted with diarrhea, to aid in evaluating its efficacy and impact on G1P[8] RVA in China. Methods Data from the Chinese viral diarrhea surveillance network were collected from January 2016 to December 2018. RVA strains identified as the G1P[8] genotype were subjected to whole-genome sequencing. Neutralizing epitope, amino acid selection pressure, and evolution dynamics analyses on VP7 and VP4 were performed using BioEdit v.7.0.9.0 and PyMOL v.2.5.2, four algorithms (MEME, SLAC, FEL, and FUBAR) in the Datamonkey online software, and the MCMC model in BEAST v. 1.10.4, respectively. Phylogenetic and identity features of 11 genes were assessed by DNAStar and MEGA v.7. Results Results showed that the detection rate of G1P[8] in China from 2016 to 2018 was generally low with significant seasonality. The whole genome of G1P[8] of four 2016 childhood diarrhea specimens was successfully sequenced. Phylogenetic and neutralizing epitope analysis showed that Rotavin-M1 might have better protection on G1P[8] prevalent in China than Rotarix and RotaTeq. Two conserved N-glycosylation sites on VP7 of Chinese G1P[8] might affect the protective effect of the vaccine. Evolution rate and selection pressure analysis identified the possibility of rapidly evolving and adapting to the new environment introduced by vaccines of G1P[8], whereas positive selection specific to VP4 indicated the potential tendency to select for dominant traits. Identity and phylogeny analysis showed that Chinese G1P[8] from before 2018 was generally stable with possible genetic recombination among local strains. Discussion These findings not only are of great significance for predicting the prevalence of G1P [8] in China, but also provide data reference for evaluating rotavirus vaccine efficacy.
Collapse
Affiliation(s)
- Rui Peng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Biosciences, Faculty of Sciences, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Mengxuan Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Saleha Shahar
- Department of Biosciences, Faculty of Sciences, Universiti Teknologi Malaysia, Johor Bahru, Malaysia
| | - Guangping Xiong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qing Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lili Pang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hong Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiangyu Kong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dandi Li
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhaojun Duan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Health Commission Key Laboratory for Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
42
|
Dinana Z, Doan YH, Maharani AT, Fitria AL, Yamani LN, Juniastuti, Wahyuni RM, Soegijanto S, Soetjipto, Utsumi T, Matsui C, Deng L, Takemae N, Kageyama T, Katayama K, Lusida MI, Shoji I. Unusual G9P[4] Rotavirus Emerged After the Dynamic Changes in Rotavirus Genotypes From Equine-Like G3 to Typical Human G1/G3 in Indonesia. J Med Virol 2024; 96:e70106. [PMID: 39670413 DOI: 10.1002/jmv.70106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/31/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024]
Abstract
Inter-genogroup reassortment of Rotavirus A (RVA) strains has highlighted the spread of unusual RVA strains worldwide. We previously reported the equine-like G3 RVA as the predominant strain in Indonesia in 2015-2016. However, since July 2017, typical human genotypes G1 and G3 have replaced these strains completely. To understand how dynamic changes in RVA occur in Indonesia, we performed a detailed epidemiological study. A total of 356 stool specimens were collected from hospitalized children in Sidoarjo, Indonesia between 2018 and 2022. Whole-genome sequencing was performed for all 26 RVA-positive samples using next-generation sequencing. Twenty-four samples were determined to be the unusual RVA G9P[4], while two were G9P[6]. Detailed analysis revealed that seven G9P[4] strains had the typical DS-1-like backbone, while the other strains exhibited a double-reassortant profile (G9-N1) on the DS-1-like backbone. The Bayesian evolutionary analyses suggested that the Indonesian G9P[4] strains share a common ancestor with previously reported G9P[4] strains in the VP7 and VP4 genes. G9P[4] DS-1-like strains were identified as the predominant genotype in Indonesia in 2021 for the first time. These results suggest that the G9P[4] strains were generated from the previous G9P[4] strains that had undergone further intra-reassortments with the other circulating strains.
Collapse
Affiliation(s)
- Zayyin Dinana
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Research Center on Global Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Yen Hai Doan
- Office of Laboratory Emergency Preparedness, Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo, Japan
| | - Aussie Tahta Maharani
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Anisa Lailatul Fitria
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Laura Navika Yamani
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Research Center on Global Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Juniastuti
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Research Center on Global Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Rury Mega Wahyuni
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Soegeng Soegijanto
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Soetjipto
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Research Center on Global Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Takako Utsumi
- Division of Infectious Disease Control, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Chieko Matsui
- Division of Infectious Disease Control, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Lin Deng
- Division of Infectious Disease Control, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Nobuhiro Takemae
- Office of Laboratory Emergency Preparedness, Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tsutomu Kageyama
- Office of Laboratory Emergency Preparedness, Center for Emergency Preparedness and Response, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kazuhiko Katayama
- Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute, Graduate School of Infection Control Sciences, Laboratory of Viral Infection I, Tokyo, Japan
| | - Maria Inge Lusida
- Laboratory of Viral Diarrhea, Indonesia-Japan Collaborative Research Center for Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
- Research Center on Global Emerging and Re-emerging Infectious Diseases, Institute of Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| | - Ikuo Shoji
- Division of Infectious Disease Control, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Hyogo, Japan
| |
Collapse
|
43
|
Zhao Y, Zhu X, Lan Q, Wei Z, Shang P, Song L, Hu S, Chen L, Gan M, Niu L, Wang Y, Shen L, Zhu L. 1α,25-hydroxyvitamin D 3 alleviated rotavirus infection induced ferroptosis in IPEC-J2 cells by regulating the ATF3-SLC7A11-GPX4 axis. Int J Biol Macromol 2024; 283:137484. [PMID: 39528192 DOI: 10.1016/j.ijbiomac.2024.137484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Rotavirus (RV) mainly infects mature intestinal epithelial cells and impairs intestinal absorption function, which leads to the death of infected cells and eventually fatal diarrhea. Ferroptosis is a novel regulatory cell death pattern, which can be caused by virus infection. 1α,25-hydroxyvitamin D3 (1,25D3) has an anti-RV infection effect and can regulate ferroptosis. However, whether RV infection can induce ferroptosis, and whether 1,25D3 can inhibit RV infection by regulating ferroptosis has not yet been studied. Present study shows that RV infection or erastin treatment induces IPEC-J2 cell death, which results in mitochondrial shrinkage, decreased mitochondrial membrane potential (MMP) and glutathione (GSH) content, increased MMP, intracellular Fe2+, reactive oxygen species (ROS), and malondialdehyde (MDA) contents. Meanwhile, ferrostatin-1 (Fer-1), liproxstatin-1 (Lip-1), and deferoxamine (DFO) treatment can effectively reverse the increase of intracellular Fe2+, ROS and MDA levels induced by RV infection. Moreover, RV infection increases activating transcription factor 3 (ATF3) mRNA and protein expressions, and inhibited SLC7A11 and glutathione peroxidase 4 (GPX4) expressions, which was partially alleviated by siATF3. 1,25D3 treatment significantly eliminates RV induced ferroptosis via ATF3-SLC7A11-GPX4 axis. Therefore, these results reveals that RV infection induces ferroptosis in IPEC-J2 cell and 1,25D3 alleviates RV induced ferroptosis by regulating the ATF3-SLC7A11-GPX4 axis.
Collapse
Affiliation(s)
- Ye Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoxiao Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Qingyuan Lan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Ziang Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Pan Shang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Song
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shijie Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mailin Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lili Niu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Linyuan Shen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Li Zhu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
44
|
Mirhoseinian M, Jalilvand S, Yaghooti MM, Kachooei A, Latifi T, Feizi M, Motamedi-Rad M, Azadmanesh K, Marashi SM, Roohvand F, Shoja Z. Full genome sequence analysis of the predominant and uncommon G9P[4] rotavirus strains circulating in Tehran, Iran, 2021-2022: Evidence for inter and intra-genotype recombination. Virology 2024; 600:110250. [PMID: 39321558 DOI: 10.1016/j.virol.2024.110250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Group A rotaviruses (RVAs) are a major cause of acute gastroenteritis in children under 5 years of age worldwide. Herein, the genetic sequences of 11 RNA segments from three uncommon G9P[4] RVA strains found in the stool samples of children under 5 years of age in Iran were analyzed using next-generation sequencing (NGS) technology. The genomic constellations of these three uncommon G9P[4] strains indicated the presence of the double and quadruple reassortants of two G9P[4] strains, containing the VP7/NSP2 and VP7/VP2/NSP2/NSP4 genes on a DS-1-like genetic background, respectively. The genome of one strain indicated a Wa-like genetic backbone in a single-reassortant with the VP4 of the DS1-like human strains. With the exception of VP1, VP2, VP7, NSP2, NSP3, and NSP4 genes, which clustered with RVA of human origins belonging to cognate gene sequences of genogroup 1/2 genotypes/lineages, the remaining five genes (VP8/VP4, VP3, VP6, NSP1, NSP5) displayed direct evidence of recombination. It is presumed that the presence of uncommon G9P[4] strains in Iran is not linked to vaccination pressure, but rather to the high prevalence of RVA co-infection or the direct import of these uncommon RVA reassortants strains from other countries (especially those that have implemented RV vaccination).
Collapse
Affiliation(s)
- Mahtab Mirhoseinian
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Atefeh Kachooei
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Latifi
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Mahsa Feizi
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Sayed Mahdi Marashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
45
|
Lv Y, Tong Z, Liu J, Zhang Z, Wang C, Zeng Y, Liu P, Zong X, Chen G, Chen H, Tan C. Molecular Characterization and Pathogenicity Analysis of Porcine Rotavirus A. Viruses 2024; 16:1842. [PMID: 39772152 PMCID: PMC11680200 DOI: 10.3390/v16121842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Porcine rotavirus A (RVA) is one of the major etiological agents of diarrhea in piglets and constitutes a significant threat to the swine industry. A molecular epidemiological investigation was conducted on 2422 diarrhea samples from Chinese pig farms to enhance our understanding of the molecular epidemiology and evolutionary diversity of RVA. The findings revealed an average RVA positivity rate of 42% (943/2422), and the study included data from 26 provinces, primarily in the eastern, southern and southwestern regions. Genetic evolutionary analysis revealed that G9 was the predominant genotype among the G-type genotypes, accounting for 25.32% of the total. The VP4 genotypes were P[7] (36.49%) and P[23] (36.49%). The predominant genotypic combinations of RVA were G9P[23] and G9P[7]. Eleven RVA strains were obtained via MA104 cell isolation. A rat model was established to assess the pathogenicity of these strains, with three strains exhibiting high pathogenicity in the model. Specifically, the RVA Porcine CHN HUBEI 2022 (Q-1), RVA Porcine CHN SHANXI 2022 (3.14-E), and RVA Porcine CHN HUBEI 2022 (5.11-U) strains were shown to cause diarrhea in the rats and damage the intestinal villi during the proliferation phase of the infection, leading to characteristic lesions in the small intestine. These data indicate that continuous monitoring of RVA can provide essential data for the prevention and control of this virus.
Collapse
Affiliation(s)
- Yaning Lv
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Ze Tong
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Jiaqi Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Zhaoran Zhang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Chenchen Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Yan Zeng
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Pingxuan Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Xin Zong
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Guosheng Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| |
Collapse
|
46
|
Song F, Zeng Y, Sheng R, Lin Y, Wang X, Hong C, Luo G, Wang Y, Fang M, He S, Zhang S, Zheng Q, Li T, Ge S, Zhang J, Xia N. VP8 Mosaic Nanoparticles Elicit Cross-Neutralizing Immune Responses and Provide Protection Against Heterotypic Rotavirus Challenge in Mice. ACS NANO 2024; 18:31809-31822. [PMID: 39497609 DOI: 10.1021/acsnano.4c07061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Group A rotaviruses (RVA) remain one of the dominant pathogens causing diarrhea in children under 5 years of age worldwide, despite a sharp decrease of RVA-associated diarrhea and mortality since the introduction of rotavirus vaccines. The decreased effectiveness of live attenuated rotavirus vaccines, coupled with the emergence of new rotavirus genotypes and the risk of cross-species virus transmission, underscores the necessity to develop more effective and broad-spectrum rotavirus vaccines. In this study, we utilized nanoparticles coupled with the SpyCatcher-SpyTag system to effectively display the truncated VP8-1 protein. The modular display of the monovalent VP8-1 proteins markedly increased the immunogenicity of VP8-1. Furthermore, the bivalent display of VP8-1 proteins from simian rotavirus SA11 and lamb rotavirus LLR on the same particle not only increased immunogenicity against homotypic antigens but also elicited robust heterotypic immune responses and conferred effective protection against a distant heterotypic rotavirus with sequence identities of only 62%-66% in an adult mouse model. Therefore, mosaic VP8 nanoparticles could be considered as a viable strategy for the development of the next-generation broad-spectrum rotavirus vaccine.
Collapse
Affiliation(s)
- Feibo Song
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yuanjun Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Roufang Sheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yunyun Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuechun Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Congming Hong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Guoxing Luo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yingbin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shuizhen He
- Haicang Hospital of Xiamen, Xiamen 361026, China
| | - Shiyin Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qingbing Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Tingdong Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shengxiang Ge
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
47
|
Pitzer VE, Ndeketa L, Asare EO, Hungerford D, Lopman BA, Jere KC, Cunliffe NA. Impact of rotavirus vaccination in Malawi from 2012 to 2022 compared to model predictions. NPJ Vaccines 2024; 9:227. [PMID: 39562592 PMCID: PMC11576906 DOI: 10.1038/s41541-024-01008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
Rotarix® vaccine was introduced into the Malawi national immunization program in October 2012. We analyzed data on children <5 years old hospitalized with acute gastroenteritis from January 2012 to June 2022, and compared to pre-vaccination data from 1997 to 2009. We estimated vaccine coverage before, during, and after the COVID-19 pandemic using data from rotavirus-negative children. We compared the observed weekly number of rotavirus-associated gastroenteritis (RVGE) cases by age to predictions from a previously developed mathematical model to estimate overall vaccine effectiveness. The number of RVGE and rotavirus-negative acute gastroenteritis cases declined substantially following vaccine introduction. Vaccine coverage among rotavirus-negative controls was >90% with two doses by July 2014, and declined to a low of ~80% in October 2020 before returning to pre-pandemic levels by July 2021. Our models captured the post-vaccination trends in RVGE incidence. Comparing observed RVGE cases to the model-predicted incidence without vaccination, overall effectiveness was estimated to be modest at 36.0% (95% prediction interval: 33.6%, 39.9%), peaking in 2014, and was highest in infants (52.5%; 95% prediction interval: 50.1%, 54.9%). Our mathematical models provide a validated platform for assessing strategies to improve rotavirus vaccine impact in low-income settings.
Collapse
Affiliation(s)
- Virginia E Pitzer
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA.
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA.
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, UK.
| | - Latif Ndeketa
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
| | - Ernest O Asare
- Department of Epidemiology of Microbial Disease, Yale School of Public Health, Yale University, New Haven, CT, USA
- Public Health Modeling Unit, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Daniel Hungerford
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Benjamin A Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Khuzwayo C Jere
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Malawi-Liverpool-Wellcome Programme, Blantyre, Malawi
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Department of Medical Laboratory Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Nigel A Cunliffe
- NIHR Global Health Research Group on Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
48
|
Xiao G, Zhu T, Wang Z, Xie X, Shu M, Gao S, Wang L, Zhou W, Deng J, Xie Y, Yu F. Pentavalent Rotavirus Vaccine Coverage and Trends in Rotavirus Detection Before and After This Vaccination in Chengdu, China. Pediatr Infect Dis J 2024; 43:e397-e399. [PMID: 38985999 DOI: 10.1097/inf.0000000000004441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Pentavalent rotavirus vaccine (RV5) coverage and changes in rotavirus detection in the prevaccine and postvaccine era in Chengdu were investigated. The results showed that the coverage of RV5 had been increasing but still relatively low. Nevertheless, the dramatical decline in the rotavirus detection was observed after the introduction of RV5. Efforts to improve the coverage of rotavirus vaccination should continue to maximize the public health benefits.
Collapse
Affiliation(s)
- Guoguang Xiao
- From the Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu, China
- West China Xiamen Hospital of Sichuan University, Xiamen, China
| | - Tingting Zhu
- Department of Clinical Laboratory, West China Second Hospital, Sichuan University, Chengdu, China
| | - Zhiling Wang
- From the Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, Ministry of Education, China
| | - Xiaoping Xie
- Department of Pediatrics, People's Hospital of Dujiangyan, Chengdu, China
| | - Min Shu
- From the Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu, China
- West China Xiamen Hospital of Sichuan University, Xiamen, China
| | - Shan Gao
- From the Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Liyuan Wang
- From the Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu, China
| | - Wei Zhou
- Department of Clinical Laboratory, West China Second Hospital, Sichuan University, Chengdu, China
| | - Jianjun Deng
- From the Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu, China
- West China Second UNIV. Hospital, SCU, Qingbaijiang Women's & Children's Hospital, Chengdu, China
| | - Yongmei Xie
- From the Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, Ministry of Education, China
| | - Fan Yu
- Department of Clinical Laboratory, West China Second Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, Ministry of Education, China
| |
Collapse
|
49
|
Yi Y, Liu J, Zhang Y, Zeng B, Lin L, Li C, Yang F, Zhang H, Xie R, Huang Z, Kang M, Jiang Y. Effectiveness of Lanzhou Lamb Rotavirus Vaccine and RotaTeq Against Hospitalized Rotavirus Infections Among Children During 2020-2023 in Guangdong Province, China: A Test-Negative Case-Control Study. Infect Dis Ther 2024; 13:2301-2317. [PMID: 39283583 PMCID: PMC11499462 DOI: 10.1007/s40121-024-01040-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/29/2024] [Indexed: 10/16/2024] Open
Abstract
INTRODUCTION The evidence regarding the effectiveness of Lanzhou Lamb Rotavirus Vaccine (LLR) and RotaTeq (RV5) against gastroenteritis (RVGE) caused by emerging genotypes in Chinese children remains limited. METHODS We conducted a test-negative case-control study using gastroenteritis surveillance data from four cities (2020-2023) in Guangdong Province, China. Children aged 2 months to 5 years hospitalized with acute gastroenteritis were enrolled. Cases were rotavirus-positive; controls were rotavirus-negative. Vaccine effectiveness (VE) was estimated using multivariable logistic regressions. RESULTS Among 2650 children, 218 (8.2%) were rotavirus-positive, predominantly G8P[8]. Also, 1543 (58.23%) children were unvaccinated, while 632 (23.85%) and 475 (17.92%) received at least one dose of RV5 and LLR, respectively. Adjusted RV5 VE against any RVGE severity was 51.7% [95% confidence interval (CI) - 58.1-85.3%]) for one dose, 37.6% (95% CI - 58.5-75.4%) for two doses, and 64.1% (95% CI 38.0-79.2%) for three doses. For LLR, VE against any RVGE severity was 38.7% (95% CI 5.7-60.2%) for one dose, 74.6% (95% CI 35.3-90.0%) for two doses, and 58.8% (95% CI - 217.6-94.6%) for three doses. Against severe RVGE, RV5 VE was 67.2% (95% CI - 144.7-95.6%) for one dose, 74.0% (95% CI - 92.1-96.5%) for two doses, and 86.6% (95% CI 56.8-95.9%) for three doses. For LLR, VE against severe RVGE was 57.7% (95% CI 20.3-77.6%) for one dose, 73.4% (95% CI 11.9-92.0%) for two doses, and - 27.8% (95% CI - 949.7-84.4%) for three doses. CONCLUSIONS Both RV5 and LLR provided protection against RVGE, including the emerging G8P[8] genotype. Three doses of RV5 offered strong protection, while two doses of LLR also appeared to be an effective strategy against rotavirus infection.
Collapse
Affiliation(s)
- Yao Yi
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China
| | - Jun Liu
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China
| | - Yingtao Zhang
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China
| | - Biao Zeng
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China
| | - Liling Lin
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China
| | - Caixia Li
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China
| | - Fen Yang
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China
| | - Hailong Zhang
- Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China
| | - Ruili Xie
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China
| | - Zhuhang Huang
- Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, Guangdong, China
| | - Min Kang
- Guangdong Center for Disease Control and Prevention, No. 160, Qunxian Road, Panyu District, Guangzhou, Guangdong, China.
| | - Yawen Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, 66 Gongchang Road, Guangming District, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen, Guangdong, China.
| |
Collapse
|
50
|
Wang K, Wang Y, Yang L, Li J, Li P, Yang C, Jia L, Qiu S, Song H, Li P. Genomic analysis of an acute gastroenteritis outbreak caused by rotavirus C in Hebei, China. Virol J 2024; 21:242. [PMID: 39358760 PMCID: PMC11448206 DOI: 10.1186/s12985-024-02486-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Rotavirus group C is an important cause of sporadic cases and outbreaks of gastroenteritis worldwide. Whole-Genome sequences of human rotavirus C (RVC) in public databases are limited. We performed genome sequencing to analyze a RVC outbreak of acute gastroenteritis in China. Samples from 22 patients were screened for pathogens using RT-PCR, and six samples were positive for rotavirus. Whole-Genome sequencing analysis showed that the outbreak strain SJZ217 belongs to the G4-P[2]-I2-R2-C2-M3-A2-N2-T2-E2-H2 genotype and shares almost identical genomic sequences with Chungnam isolated in Korea. Phylogenetic analysis revealed strain SJZ217 also fell into a cluster with rotavirus C strains from Japan and Europe. Reassortment in the VP4 fragment was observed. These results helped to understand the genetic diversity and possible spread of RVC strains.
Collapse
Affiliation(s)
- Kaiying Wang
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China
| | - Yun Wang
- Tianjin Binhai New Area Center for Disease Control and Prevention, Tianjin, 300450, China
| | - Lang Yang
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China
| | - Jinhui Li
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China
| | - Peihan Li
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China
| | - Chaojie Yang
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China
| | - Leili Jia
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China
| | - Shaofu Qiu
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China.
| | - Hongbin Song
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China.
| | - Peng Li
- Chinese PLA Center for Disease Control and Prevention, 20 Dongda Street, Fengtai District, Beijing, 100071, China.
| |
Collapse
|