1
|
Jeong NY, Cho H, Kim HJ, Choi NK. A broad assessment of rotavirus vaccine safety in infants in Korea: Insights from a data-driven signal detection approach. Hum Vaccin Immunother 2025; 21:2465161. [PMID: 39936376 PMCID: PMC11834447 DOI: 10.1080/21645515.2025.2465161] [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: 12/04/2024] [Revised: 01/15/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025] Open
Abstract
In light of the widespread use of rotavirus vaccines, there is a pressing need to perform thorough, large-scale surveillance to actively monitor safety. This study aimed to identify potential adverse events following rotavirus vaccination in infants. Using a nationwide linked database of the national immunization registry and health insurance claims data, we identified infants vaccinated with the first dose of rotavirus vaccine between January 2016 and October 2022. The self-controlled tree-temporal scan statistics method analyzed all incident diagnoses recorded within 56 days post-vaccination and evaluated all temporal risk windows. Among 1,720,778 rotavirus vaccine recipients 64,752 infants contributed to the analysis, yielding 72,970 incident diagnoses. Of these, 28 clusters were categorized as known adverse drug reactions (ADRs), including infection following immunization (Days 1-2, p<.001), viral infection (Days 1-5, p<.001), urticaria and erythema (Days 3-9, p<.001), acute upper respiratory infections (Days 28-42, p<.001), and pneumonia (Days 9-19 or 28-42, p<.001). Seventeen clusters were classified as ADR-related events, such as the ones clinically related to ADR or lower-level diagnostic codes of ADR. The remaining 26 clusters were classified as signals, including sepsis (Days 1-20, p<.001), meningitis (Days 1-23, p<.001), liver disease (Days 4-11, p<.001), and tubulo-interstitial nephritis (Days 11-38, p<.001). A cluster of intussusceptions was only found in monovalent vaccine-stratified analysis (Days 5-8, p = 0.005). This study confirmed known ADRs following rotavirus vaccination, while identifying potential safety signals requiring further investigation. These findings emphasize the importance of active vaccine surveillance and underscore the need for epidemiological studies with validated outcome definitions to confirm causal relationships between rotavirus vaccination and the detected outcomes.
Collapse
Affiliation(s)
- Na-Young Jeong
- Health Science Convergence Research Institute, Ewha Womans University, Seoul, Republic of Korea
| | - Haerin Cho
- Department of Health Convergence, College of Science and Industry Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Hee-Jin Kim
- Department of Health Convergence, College of Science and Industry Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Nam-Kyong Choi
- Department of Health Convergence, College of Science and Industry Convergence, Ewha Womans University, Seoul, Republic of Korea
- Graduate School of Industrial Pharmaceutical Science, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Yan M, Su A, Meyer D, Sosa GR, Fritsch H, Pitters M, Fischer N, Herrler G, Becher P. Precursor of H-type II histo-blood group antigen and subterminal sialic acids on gangliosides are significantly implicated in cell entry and infection by a porcine P[11] rotavirus. Emerg Microbes Infect 2025; 14:2447608. [PMID: 39726161 PMCID: PMC11727068 DOI: 10.1080/22221751.2024.2447608] [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: 08/01/2024] [Revised: 10/21/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Rotaviruses, non-enveloped viruses with a double-stranded RNA genome, are the leading etiological pathogen of acute gastroenteritis in young children and animals. The P[11] genotype of rotaviruses exhibits a tropism for neonates. In the present study, a binding assay using synthetic oligosaccharides demonstrated that the VP8* protein of P[11] porcine rotavirus (PRV) strain 4555 binds to lacto-N-neotetraose (LNnT) with the sequence Galβ1,4-GlcNAcβ1,3-Galβ1,4-Glc, one of the core parts of histo-blood group antigen (HBGA) and milk glycans. However, infections were significantly inhibited by blocking of endogenous monosialoganglioside (GM) GM1a with cholera toxin B subunit and preincubation of the virus with exogenous GM1a, suggesting that GM1a is involved in the infection of P[11] PRV 4555. In addition to GM1a, preincubation of the virus with exogenous disialogangliosides (GD) GD1a, GD1b, and trisialoganglioside (GT) GT1b also prevented infection. In contrast, exogenous ganglioside GM3 only inhibited infections at an early time point, and exogenous asyalosphingolipids GA1 and LacCer did not show any inhibitory effect on infections. This indicates that P[11] PRV 4555 preferentially utilizes gangliosides containing subterminal sialic acids. Further experiments revealed that P[11] PRV 4555 infections were prevented by preincubation of the virus with Neu5Ac and Neu5Gc. These results confirmed that sialic acids are essential for P[11] PRV 4555 cell entry, despite the classification as NA-resistant strain. Overall, our results proved that P[11] rotavirus not only binds to the Gal-GlcNAc motif but also utilizes gangliosides containing subterminal sialic acids.
Collapse
Affiliation(s)
- Miaomiao Yan
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ang Su
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Denise Meyer
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gleyder Roman Sosa
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Henrik Fritsch
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Malte Pitters
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole Fischer
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Georg Herrler
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Paul Becher
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
3
|
Thakur R, Kaur S. Use of postbiotics and parabiotics from lactobacilli in the treatment of infectious diarrhea. Microb Pathog 2025; 204:107580. [PMID: 40222563 DOI: 10.1016/j.micpath.2025.107580] [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: 07/10/2024] [Revised: 01/30/2025] [Accepted: 04/11/2025] [Indexed: 04/15/2025]
Abstract
Probiotics are effective in the treatment of diarrheal disease which is the second leading cause of death in children below the age of five years via the production of antimicrobial peptides and lactic acid. These live bacteria are known to benefit the host by modulating their gut microbiome and competitively excluding pathogens from the gut. As probiotics are live microbial cells, their safety evaluation is a concern that shifts the focus from the usage of live cells to parabiotics and postbiotics. In recent years attempts have been made to study the efficacy of postbiotics and parabiotics against enteric pathogens. Enteric pathogens are the major cause of diarrhea resulting in watery stools and electrolyte imbalance. Among various gastrointestinal illnesses, 30% are caused by bacteria. These gastrointestinal infections in adults have usually mild to moderate symptoms that disappear spontaneously but, in some cases, they can cause chronic diseases such as typhoid, irritable bowel syndrome, ulcerative colitis and bacteremia. The extensive use of antibiotics for the treatment of bacterial-infection-induced diarrhea has led to the emergence of drug resistance among these enteric pathogens. Drug resistance poses a major threat in the treatment of various other diseases as well. Further, the use of antibiotics is known to disrupt the homeostasis of the gut by killing the normal gut flora thereby worsening the situation. Therefore, the urgent need for new interventions to combat these enteric pathogens along with restoration of gut barrier. Lactobacillus-derived parabiotics and postbiotics have emerged as promising approaches for managing and treating diarrheal diseases. Therefore, our research is focused on studying the efficacy and underlying mechanisms of Lactobacillus spp.-derived postbiotics and parabiotics against enteric pathogens. Understanding these mechanisms helps in combatting diarrhea associated with enteric pathogens and results in reducing the morbidity and mortality rates associated with infectious diarrhea and its complications.
Collapse
Affiliation(s)
- Raman Thakur
- Department of Medical Laboratory Sciences, Lovely Professional University, Punjab, INDIA-144411
| | - Sumanpreet Kaur
- Department of Medical Laboratory Sciences, Lovely Professional University, Punjab, INDIA-144411.
| |
Collapse
|
4
|
Cho M, Been N, Son HS. Analysis of protein determinants of genotype-specific properties of group a rotaviruses using machine learning. Comput Biol Med 2025; 191:110143. [PMID: 40203739 DOI: 10.1016/j.compbiomed.2025.110143] [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: 11/05/2024] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Group A rotaviruses (RVAs) are the leading cause of viral diarrhoea across various host species, including mammals and birds. The VP7 and VP4 proteins of these viruses play critical roles in determining genotype specificity, influencing viral infectivity and host adaptation. This study employed machine-learning techniques to classify RVA genotypes based on the molecular and physicochemical properties of these proteins. A dataset of 94 VP7 and 68 VP4 protein sequences was collected from various host species. Seven machine-learning algorithms-Naïve Bayes (NB), logistic regression (LR), decision tree (DT), random forest (RF), k-nearest neighbour (kNN), support vector machine (SVM), and artificial neural network (ANN)-were used for genotype classification. Feature subsets were configured using ranking-based attribute selection, and classification performance was evaluated using accuracy (ACC), precision, recall, Matthews' correlation coefficient (MCC), and the area under the curve (AUC). kNN demonstrated the highest classification accuracy for both VP7 (ACC = 97.87 %) and VP4 (ACC = 100 %), outperforming NB, LR, DT, RF, SVM, and ANN. For VP7 sequences, key properties influencing genotype classification included hydrophobicity, normalised van der Waals volume, and leucine composition. For VP4, polarity, normalised van der Waals volume, and polarizability were the most significant factors. In summary, the genotype-specific molecular features of VP7 and VP4 proteins served as reliable markers for RVA classification. Our findings highlight the potential of machine-learning approaches to predict RVA genotypes based on the physicochemical properties of amino acids, providing valuable insights into the molecular mechanisms that drive viral evolution, host specificity, and immune evasion.
Collapse
Affiliation(s)
- Myeongji Cho
- Laboratory of Computational Virology & Viroinformatics, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea; Public Health AI Lab, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea; Institute of Health and Environment, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Nara Been
- Laboratory of Computational Virology & Viroinformatics, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea; Public Health AI Lab, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Hyeon S Son
- Laboratory of Computational Virology & Viroinformatics, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea; Public Health AI Lab, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea; Institute of Health and Environment, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea; Interdisciplinary Graduate Program in Bioinformatics, College of Natural Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|
5
|
Shen J, Xiao Y, Bi H, Guo R, Qu M, Ding R, Zhang X, Li H, Han M, Huang S. Rotavirus regulates the gut microhabitat immune response through the IL-22/pSTAT3/RegIIIγ signaling pathway, leading to lactose intolerance. Int Immunopharmacol 2025; 150:114275. [PMID: 39954663 DOI: 10.1016/j.intimp.2025.114275] [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: 01/22/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Lactose intolerance (LI) is a significant contributor to diarrhea in infants and young children. Recent studies suggest a potential link between rotavirus (RV) infection and alterations in gut microbiota, which may play a role in the mechanism of LI. However, the precise underlying mechanism remains unclear and requires urgent clarification. In this study, we combined insights from immunology and gut microbiome research to propose that the activation of the IL-22/pSTAT3/RegIIIγ signaling pathway, triggered by gut microbiota, may be involved. To investigate this, we established a LI animal model using 7-day-old BALB/c mice, which were infected with RV via oral gavage. The syndromes of mice were carefully recorded and compared during the intervention experiment. Notably, we measured the the expression levels of immunocytokines and phosphorylated signaling proteins, including IL-22, phosphorylated STAT3, and RegIIIγ. Additionally, we assessed the relationships between gut microbiota and these key elements. Our results indicated that RV indeed causes LI, as severe diarrhea was observed in the mice during the first three days of RV infection, subsiding after seven days. ELISA results revealed an increase in IL-22 levels, phosphorylated STAT3, and RegIIIγ, suggesting that the mechanism of LI associated with RV is linked to the classical IL-22/pSTAT3/RegIIIγ signaling pathway. Furthermore, our analysis of the connections between gut microbes and this signaling pathway indicated that "Bacteroidetes" and Firmicutes were significantly positively correlated with IL-22 and lactase, respectively. This finding implies that alterations in gut microbiota may serve as a potential switch for the IL-22/pSTAT3/RegIIIγ signaling pathway, leading to increased lactase levels. In summary, our study demonstrated that the development of LI is associated with the activation of the IL-22/pSTAT3/RegIIIγ signaling pathway alongside changes in gut microbes during RV infection. This provides valuable insights into the mechanisms underlying LI and its clinical treatment through moduLation of the gut microbiome.
Collapse
Affiliation(s)
- Jilu Shen
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230000 Anhui Province, China
| | - Yixuan Xiao
- Department of Blood Transfusion, The Children's Hospital, Zhejiang, Zhejiang University School of Medicine, Hangzhou, China
| | - Huijuan Bi
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230000 Anhui Province, China
| | - Rui Guo
- Department of Microbiology, The Institute of Clinical Virology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Mingsheng Qu
- Department of Microbiology, The Institute of Clinical Virology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Rui Ding
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Xiaocheng Zhang
- Department of Microbiology, The Institute of Clinical Virology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Haiwen Li
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Medical University, Hefei City 230601 Anhui, China
| | - Maozhen Han
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China.
| | - Shenghai Huang
- Department of Microbiology, The Institute of Clinical Virology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, China; Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230000 Anhui Province, China; School of Life Sciences, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
6
|
Cullinane A, Nelly M, Dayot L, Lukaseviciute G, Garvey M, Healy J, Gallagher R. Diagnostic Performance of Rapid Antigen Tests to Detect Equine Rotavirus A. Viruses 2025; 17:413. [PMID: 40143340 PMCID: PMC11946396 DOI: 10.3390/v17030413] [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: 01/21/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
This study describes a comparison of the detection of rotavirus in clinical samples from foals using two commercially available rapid antigen detection (RAD) kits, with the detection of rotavirus nucleic acid via a laboratory-based, in-house, real-time reverse transcription polymerase chain reaction (RT-PCR) assay. One hundred and forty freeze-thawed samples (70 that were RT-PCR-positive and 70 that were RT-PCR-negative on original tests) submitted to the diagnostic laboratory over a seven-year period were tested in addition to 123 fresh samples (15 RT-PCR-positive and 108 RT-PCR-negative) submitted over a four- month period in 2024. The analyst performing the RAD tests was blinded to the RT-PCR result as were the two individuals who read the results. Samples with discordant results were re-tested in duplicate using RT-PCR and the two RAD kits. Both kits demonstrated a high level of concordance with the RT-PCR (>95%). However, testing of serial dilutions of RT-PCR positive faeces samples indicated that the RADs failed to detect the virus at the higher dilutions. In conclusion, the RADs evaluated are potentially useful for screening individual foals and for the determination of the urgency of the appropriate treatment and isolation. Negative samples from suspect cases and weak positives should always be submitted to a specialist laboratory for real-time RT-PCR testing.
Collapse
Affiliation(s)
- Ann Cullinane
- Virology Unit, The Irish Equine Centre, Johnstown, Naas, Co., W91 RH93 Kildare, Ireland; (M.N.); (L.D.); (G.L.); (M.G.); (J.H.); (R.G.)
| | | | | | | | | | | | | |
Collapse
|
7
|
Hou G, Son J, Gomez Castro MF, Kawagishi T, Ren X, Roth AN, Antia A, Zeng Q, DeVeaux AL, Feng N, Kohio HP, Baldridge MT, Dermody TS, Zhu S, Ding S. Innate immune sensing of rotavirus by intestinal epithelial cells leads to diarrhea. Cell Host Microbe 2025; 33:408-419.e8. [PMID: 40037352 PMCID: PMC11932023 DOI: 10.1016/j.chom.2025.02.005] [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: 09/03/2024] [Revised: 01/07/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
Diarrhea is the predominant symptom of acute gastroenteritis resulting from enteric infections and a leading cause of death in infants and young children. However, the role of the host response in diarrhea pathogenesis is unclear. Using rotavirus and neonatal mice as a model, we found that oral inoculation of UV-inactivated replication-defective rotavirus consistently induced watery diarrhea by robust activation of cytosolic double-stranded RNA sensing pathways and type III interferon (IFN-λ) secretion. Diarrhea was significantly diminished in mice lacking the IFN-λ receptor. Mechanistically, IFN-λ signaling downregulates the expression of Dra, a chloride and bicarbonate exchanger, which contributes to reduced water absorption. We confirmed these findings in mice inoculated with reovirus, as well as in donor-derived human intestinal organoids and human biopsy samples. Our data highlight a mechanism of rapid diarrhea induction by host innate immune sensing in the gastrointestinal tract and suggest that diarrhea induction is an active host defense strategy to eliminate the pathogen.
Collapse
Affiliation(s)
- Gaopeng Hou
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Juhee Son
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Maria Florencia Gomez Castro
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Takahiro Kawagishi
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Xingxing Ren
- Institute of Immunology and the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Alexa N Roth
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Qiru Zeng
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Anna L DeVeaux
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Ningguo Feng
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hinissan P Kohio
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Megan T Baldridge
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Terence S Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Shu Zhu
- Institute of Immunology and the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
8
|
Bomidi C, Sawyer FM, Shroyer N, Conner M, Estes MK, Blutt SE. Loss of mucin 2 and MHC II molecules causes rare resistance to murine RV infection. J Virol 2025; 99:e0150724. [PMID: 39727412 PMCID: PMC11852729 DOI: 10.1128/jvi.01507-24] [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: 09/03/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Enteric pathogen rotavirus (RV) primarily infects mature enterocytes at the tips of the intestinal villi; however, the role of secretory Paneth and goblet cells in RV pathogenesis remains unappreciated. Atoh1 knockout mice (Atoh1cKO) were used to conditionally delete Paneth, goblet, and enteroendocrine cells in the epithelium to investigate the role of secretory cells in RV infection. Unexpectedly, the number of infected enterocytes and the amount of RV shedding in the stool were greatly decreased following secretory cell deletion. Resistance to RV infection persisted for 7 days after virus inoculation, and Atoh1 knockout mice co-housed with infected wild-type mice were uninfected, based on lack of shedding virus, despite the highly infectious nature of RV. This response was directly proportional to the extent of secretory cell deletion, with infection predominantly occurring in areas containing intact secretory cells. RV infection of Muc2 knockout mice recapitulated the secretory cell deletion phenotype, indicating that goblet cell loss is responsible for attenuated infection. Transcriptome analysis of Atoh1cKO intestine via single-cell RNA sequencing revealed downregulation of MHC II molecules specifically in tip enterocytes, and MHC II-/- mice were likewise resistant to RV infection. These data suggest a previously unknown role for both MUC2 and MHC II expression in susceptibility to RV infection.IMPORTANCERotavirus (RV) is a highly contagious pathogen that primarily infects mature intestinal enterocytes. Murine rotavirus readily infects infant and adult mice, enabling evaluation of RV infection and immunity. We report that mice lacking secretory cells are one of the few genetically modified mouse lines not susceptible to murine rotavirus. Further investigation revealed loss of mucin 2 (MUC2) expression or major histocompatibility complex II (MCH II) expression recapitulated this rare resistance to rotavirus infection, suggesting a previously unrecognized link between secretory cell products and major histocompatibility complex II expression. Furthermore, these mouse models provide a platform to investigate rotavirus pathogenesis.
Collapse
Affiliation(s)
- Carolyn Bomidi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Faith M. Sawyer
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Noah Shroyer
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Margaret Conner
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
9
|
Passarelli GV, Doldan P, Metz-Zumaran C, Keser Y, Boulant S, Stanifer ML. Rotavirus Spreads in a Spatially Controlled Manner. Cells 2025; 14:313. [PMID: 39996783 PMCID: PMC11854656 DOI: 10.3390/cells14040313] [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: 12/30/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Rotavirus is an enteric virus that leads to 200,000 deaths worldwide every year. The live-cell imaging evaluating rotavirus infection of MA104 cells revealed that rotavirus replication and spread occurs in a spatially controlled manner. Specifically, following initial rotavirus infection, the infected cells die, and the second round of infection occurs in the restricted area surrounding the initially infected cell. Interestingly, we found that the time required to establish the secondary infection is shorter compared to the time required for the initial infection. To determine if this increase in the kinetic of secondary infection was due to the early release of viruses or priming of the cells that are infected during the secondary infection, we used a combination of live-cell microscopy, trypsin neutralization assays, and the pharmacological inhibition of calcium signaling. Together, our results show that the second round of infection required rotavirus to be released and accessible to extracellular proteases. In addition, we found that the calcium wave induced upon rotavirus infection was critical for initial infection but did not play a role in the establishment of a secondary infection. Finally, we uncovered that high viral titers released from the initial infection were sufficient to accelerate the rate of the secondary infection.
Collapse
Affiliation(s)
- Gianna V. Passarelli
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32601, USA
| | - Patricio Doldan
- Department of Infectious Disease: Virology, Heidelberg University Hospital, 69205 Heidelberg, Germany
| | - Camila Metz-Zumaran
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32601, USA
- Department of Infectious Disease: Virology, Heidelberg University Hospital, 69205 Heidelberg, Germany
| | - Yagmur Keser
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32601, USA
| | - Steeve Boulant
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32601, USA
- Department of Infectious Disease: Virology, Heidelberg University Hospital, 69205 Heidelberg, Germany
| | - Megan L. Stanifer
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32601, USA
| |
Collapse
|
10
|
Saleem W, Aslam A, Tariq M, Nauwynck H. Intestinal mucus: the unsung hero in the battle against viral gastroenteritis. Gut Pathog 2025; 17:11. [PMID: 39972475 PMCID: PMC11841282 DOI: 10.1186/s13099-025-00684-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
Intestinal mucus plays a crucial role in defending against enteric infections by protecting the vulnerable intestinal epithelial cells both physically and through its various constituents. Despite this, numerous gastroenteritis-causing viruses, such as rotavirus, coronavirus, adenovirus, astrovirus, calicivirus, and enterovirus, continue to pose significant threats to humans and animals. While several studies have examined the interactions between these viruses and intestinal mucus, significant gaps remain in understanding the full protective potential of intestinal mucus against these pathogens. This review aims to elucidate the protective role of intestinal mucus in viral gastroenteritis. It begins with a comprehensive literature overview of (i) intestinal mucus, (ii) enteric viruses of medical and veterinary importance, and (iii) the known interactions between various enteric viruses and intestinal mucus. Following this, a case study is presented to highlight the age-dependent blocking effect of porcine intestinal mucus against transmissible gastroenteritis virus, a porcine coronavirus. Finally, the review discusses future investigation directions to further explore the potential of intestinal mucus as a defense mechanism against viral gastroenteritis to stimulate further research in this dynamic and critical area.
Collapse
Affiliation(s)
- Waqar Saleem
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium.
| | - Ateeqa Aslam
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium
| | - Mehlayl Tariq
- Laboratory of Biomedical Chemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, 53-114, Poland
| | - Hans Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium
| |
Collapse
|
11
|
Gong X, Kuang X, Zheng Y, Lin S, Xiao W, Chen M, Chen J, Teng Z, Pan H. Variations in Prevalence and Characteristics of Rotavirus Diarrhea Among Outpatients - Shanghai Municipality, China, 2017-2023. China CDC Wkly 2025; 7:244-252. [PMID: 39974767 PMCID: PMC11832438 DOI: 10.46234/ccdcw2025.039] [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: 06/27/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Introduction This study investigated temporal changes in rotavirus group A (RVA) prevalence, epidemiological characteristics, and genotype distribution patterns among diarrhea outpatients in Shanghai Municipality, China. Methods We conducted prospective active surveillance of diarrheal disease in pediatric and adult outpatients in Shanghai. Stool specimens were analyzed for five viral and twelve bacterial pathogens. Real-time reverse transcription polymerase chain reaction (rRT-PCR) was employed for RVA detection, followed by genotyping of RVA-positive specimens through partial amplification of VP7 and VP4 genes. Results The study analyzed 2,331 diarrhea cases in children aged 0-14 years and 8,418 cases in individuals aged ≥15 years between January 2017 and December 2023. Overall RVA positivity rates decreased significantly from 7.43% in 2017 to 1.19% in 2023 (P=0.024). The most pronounced decline occurred in children aged 2-5 years, where positivity rates fell from 13.08% to 1.72%. Adults aged ≥30 years also showed a substantial reduction. Among RVA-positive pediatric cases (≤14 years), the proportion of cases aged 6-14 years increased from 2.33% to 18.18%. While G9P[8] remained the predominant strain, its prevalence decreased from 77.78% to 31.25%, concurrent with the emergence of G8P[8] strains. Conclusions RVA prevalence has shown a marked decline since 2018-2019, accompanied by a shift in age distribution toward older children. The diminishing dominance of G9P[8] strains coincided with the emergence of G8P[8] strains. Continued epidemiological and genetic surveillance of rotavirus diarrhea, coupled with real-world effectiveness evaluations of domestic vaccines, remains crucial for optimizing rotavirus immunization strategies.
Collapse
Affiliation(s)
- Xiaohuan Gong
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Xiaozhou Kuang
- Division of Microbiology Detection, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Yaxu Zheng
- Shanghai Academy of Preventive Medicine, Shanghai, China
| | - Sheng Lin
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Wenjia Xiao
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Min Chen
- Division of Microbiology Detection, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Jian Chen
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| | - Zheng Teng
- Division of Microbiology Detection, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Hao Pan
- Division of Infectious Disease Control and Prevention, Shanghai Municipal Center of Disease Control and Prevention, Shanghai, China
| |
Collapse
|
12
|
Yan Q, Chen Y, Gao EB, Lu Y, Wu J, Qiu H. The characteristics of intestinal microflora in infants with rotavirus enteritis, changes in microflora before and after treatment and their clinical values. Sci Rep 2025; 15:4312. [PMID: 39910252 PMCID: PMC11799311 DOI: 10.1038/s41598-025-88312-w] [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: 10/29/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
Rotavirus (RV) is a leading pathogen causing diarrhea in children. In this study, a total of 51 fecal samples from children with RV enteritis, 29 post-treatment fecal samples, and 38 fecal samples from age-matched healthy controls were collected. Microbial DNA was isolated from the samples followed by high throughput Illumina sequencing targeting 16 S rRNA gene. Compared to the healthy group, the RV-infected group exhibited reduced microbial diversity. Both groups shared Firmicutes as the dominant phylum. Additionally, the abundance of Proteobacteria increased significantly in the RV-infected group. At the genus level, among the top 50 most abundant genera, 34 showed significant differences, with these differential genera correlating with certain clinical indicators such as dehydration levels and C-reactive protein (CRP). Notably, there were no significant differences in the microbiota before and after treatment in RV-infected children. Only 8.82% (3/34) of the differential genera in the post-treatment group showed a recovery trend towards the healthy state. This study enhances the understanding of how RV infection alters the gut microbiota structure in children and provides a scientific basis for improving clinical diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Qiyan Yan
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Yuanyuan Chen
- Department of Pediatrics, Jinhua Women's and Children's Hospital, Jinhua, 321000, Zhejiang, China
| | - E-Bin Gao
- School of Life Sciences, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Yanbo Lu
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Junhua Wu
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China.
| | - Haiyan Qiu
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
13
|
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
|
14
|
Nguyen HT, Hoa-Tran TN, Tran HQ, Nguyen TTT. In Vitro Inhibitory Effect of Berberine Against Rotavirus. Chem Biodivers 2025; 22:e202400986. [PMID: 39400499 DOI: 10.1002/cbdv.202400986] [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: 04/18/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Although berberine (BBR) is well known as an active constituent in traditional medicines used in the treatment of gastrointestinal diseases, its potential against viral gastroenteritis has not been specifically reported. This study aims to investigate the antiviral activity of BBR against rotavirus and evaluate its cytotoxicity and pharmacological efficacies, including antioxidant and anti-inflammatory activities in vitro. Using ultraviolet-visible absorption spectroscopy, the saturation concentration of BBR was determined as 2261 μg/mL, indicating that BBR is a poor water-soluble compound. The inhibition rate of nitric oxide (NO) production of BBR solution at a concentration of 283 μg/mL was similar to that of Cardamonin 0.3 μM with a cell viability of 92.46±0.35 %, revealing the anti-inflammatory activity of BBR. The cytotoxicity of the BBR solution depended on its concentration, whereby the 50 % cytotoxicity concentration (CC50) of BBR after 96 h exposure was 664 μg/mL. Investigation of cytopathic effects (CPEs) of MA104 cells treated with BBR and BBR-incubated rotavirus indicates that BBR could effectively inhibit the replication of rotavirus. CPEs were not observed in the cells inoculated with rotavirus (100TCID50) which was pre-incubated with BBR for 96 hours at a BBR concentration of 283 μg/mL. Therefore, the study provides reliable results to demonstrate the ability of BBR to inhibit the replication of rotavirus.
Collapse
Affiliation(s)
- Hue Thi Nguyen
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, 12116, Vietnam
- Faculty of Biomedical Sciences, Phenikaa University, Hanoi, 12116, Vietnam
| | - Thi Nguyen Hoa-Tran
- Department of Virology, National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Huy Quang Tran
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, 12116, Vietnam
- Faculty of Biomedical Sciences, Phenikaa University, Hanoi, 12116, Vietnam
| | - Thuy Thi Thu Nguyen
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, 12116, Vietnam
| |
Collapse
|
15
|
Saha R, Lo M, De P, Deb AK, Indwar P, Miyoshi SI, Kitahara K, Oka T, Dutta S, Chawla-Sarkar M. Epidemiology of viral gastroenteritis in children and genetic diversity of rotavirus strains in Kolkata, West Bengal after introduction of rotavirus vaccine. Vaccine 2025; 45:126637. [PMID: 39731817 DOI: 10.1016/j.vaccine.2024.126637] [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: 09/06/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Despite global rotavirus vaccination efforts, rotavirus remains a leading cause of childhood deaths from acute gastroenteritis. Post-vaccination studies in India, particularly in eastern India, have been limited, despite high prevalence of rotavirus in this region prior to vaccine introduction. This study was conducted to assess the impact of rotavirus vaccine on the epidemiology of rotavirus and other enteric viruses, as well as the changes in the diversity of rotavirus strains among children (≤5 years) with acute gastroenteritis. METHODS A total of 877 stool samples from children hospitalized with acute diarrhea during 2022-2023, were screened for enteric viruses using multiplex PCR. Rotavirus positive samples were genotyped by sequencing and phylogenetic analysis of VP4 and VP7 genes were done. RESULTS AND DISCUSSION Out of 877 diarrheal cases, 47 % tested positive for at least one enteropathogenic virus. Rotavirus was most prevalent (25.9 %), followed by norovirus (11.4 %), adenovirus-F (10.6 %), and astrovirus (5.3 %). Among mixed infections, rotavirus and norovirus co-infections were the most common. Rotavirus infection was highest in children aged 12-24 months, while other enteric viruses were more common in the 6-24 month age group. Clinical severity was higher among rotavirus-infected patients compared to those infected with other enteric viruses. The G3P[8] genotype of rotavirus predominated, with notable increase in G2P[4] and the detection of rare strains like G3P[6] and G11P[25]. G3P[6] was identified for the first time in this region showing Wa-like genome constellation. Unlike pre-vaccine period, G9 genotype was not detected. Mutations in antigenic epitope of circulating strains compared to vaccine strains may affect vaccine efficacy. CONCLUSION The study highlights the persistent burden of childhood diarrhea despite rotavirus vaccination. Subtle alterations in the proportion of other enteric viruses and diversity of circulating rotavirus genotypes in the post-vaccination period were observed. Continuous long-term surveillance is required to evaluate the impact of vaccine in this region.
Collapse
Affiliation(s)
- Ritubrita Saha
- ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Kolkata, India
| | - Mahadeb Lo
- ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Kolkata, India
| | - Papiya De
- ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Kolkata, India
| | - Alok K Deb
- ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Kolkata, India
| | - Pallavi Indwar
- ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Kolkata, India
| | - Shin-Ichi Miyoshi
- Collaborative Research Center of Okayama University for Infectious Diseases in India, ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Kolkata, India; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; Research Center for Intestinal Health Science, Okayama University, Okayama, Japan
| | - Kei Kitahara
- Collaborative Research Center of Okayama University for Infectious Diseases in India, ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Kolkata, India; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tomoichiro Oka
- Department of Virology II, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashi-Murayama, Tokyo, Japan
| | - Shanta Dutta
- Regional Virus Research and Diagnostic Laboratory, ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Beliaghata, Kolkata, West Bengal, India
| | - Mamta Chawla-Sarkar
- ICMR- National Institute for Research in Bacterial Infections (formerly ICMR-NICED), Kolkata, India.
| |
Collapse
|
16
|
Katic L, Mihaljevic B, Pirija M, Goic-Barisic I, Tonkic M, Novak A. Comparison of AccuPower Diarrhea V1&V2 RT-PCR to a Chromatographic Immunoassay for Detecting Viral Pathogens from Human Diarrheal Stool Specimens. Trop Med Infect Dis 2025; 10:33. [PMID: 39998037 PMCID: PMC11860935 DOI: 10.3390/tropicalmed10020033] [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: 12/05/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Viruses are a frequent cause of self-limited diarrhea, with more severe outcomes in immunocompromised patients. This study aimed to compare the performance of Real-Time RT-PCR to chromatographic immunoassays (CIAs) for detecting the major gastrointestinal viruses in human stool. This study was conducted at the University Hospital of Split, Croatia, from October 2023 to May 2024. Stool samples were simultaneously analyzed with CIA (Acro Biotech Rotavirus and Adenovirus Combo Rapid Test Cassette, USA and JusChek Norovirus Rapid Test Cassette, China) and Real-Time RT-PCR (AccuPower Diarrhea V1&V2 Real-Time RT-PCR, Bioneer, Republic of Korea), according to the manufacturers' instructions. Positive percent agreement (PPA), negative percent agreement (NPA), and overall percent agreement (OPA) were calculated. For norovirus, CIA had a low PPA (25%), indicating that it missed 75% of norovirus-positive cases identified by RT-PCR. Adenovirus detection by CIA showed poor agreement with RT-PCR (PPA 0%; NPA 100%). Rotavirus detection presented a relatively better performance with CIA (PPA 90.9% and OPA 84.13%). However, the presence of false positives (15.8%) highlights the need for confirmatory RT-PCR testing. One specimen was sapovirus-RT-PCR-positive, marking the first documented case from human specimens in Croatia. Although CIA provided rapid results, limitations regarding reliability highlight the value of RT-PCR, particularly in the case of ambiguous clinical cases with negative antigenic test results and newly emerged viruses. A two-step diagnostic approach, with initial CIA screening followed by confirmatory RT-PCR, could balance cost-effectiveness with diagnostic accuracy.
Collapse
Affiliation(s)
- Luka Katic
- Department of Medicine, Icahn School of Medicine at Mount Sinai Morningside/West, 1000 Tenth Avenue, New York, NY 10019, USA;
- ESCMID Food- and Water-borne Infections Study Group (EFWISG), 4051 Basel, Switzerland
| | - Boris Mihaljevic
- Department of Clinical Microbiology, University Hospital of Split, 21000 Split, Croatia
| | - Marijo Pirija
- Department of Clinical Microbiology, University Hospital of Split, 21000 Split, Croatia
| | - Ivana Goic-Barisic
- Department of Clinical Microbiology, University Hospital of Split, 21000 Split, Croatia
- School of Medicine Split, University of Split, 21000 Split, Croatia
| | - Marija Tonkic
- Department of Clinical Microbiology, University Hospital of Split, 21000 Split, Croatia
- School of Medicine Split, University of Split, 21000 Split, Croatia
| | - Anita Novak
- ESCMID Food- and Water-borne Infections Study Group (EFWISG), 4051 Basel, Switzerland
- Department of Clinical Microbiology, University Hospital of Split, 21000 Split, Croatia
- School of Medicine Split, University of Split, 21000 Split, Croatia
| |
Collapse
|
17
|
Burke RM, Ramani S, Lynch J, Cooper LV, Cho H, Bandyopadhyay AS, Kirkwood CD, Steele AD, Kang G. Geographic disparities impacting oral vaccine performance: Observations and future directions. Clin Exp Immunol 2025; 219:uxae124. [PMID: 39774633 PMCID: PMC11773816 DOI: 10.1093/cei/uxae124] [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: 08/30/2024] [Revised: 11/01/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Oral vaccines have several advantages compared with parenteral administration: they can be relatively cheap to produce in high quantities, easier to administer, and induce intestinal mucosal immunity that can protect against infection. These characteristics have led to successful use of oral vaccines against rotavirus, polio, and cholera. Unfortunately, oral vaccines for all three diseases have demonstrated lower performance in the highest-burden settings where they are most needed. Rotavirus vaccines are estimated to have >85% effectiveness against hospitalization in children <12 months in countries with low child mortality, but only ~65% effectiveness in countries with high child mortality. Similarly, oral polio vaccines have lower immunogenicity in developing country settings compared with high-resource settings. Data are more limited for oral cholera vaccines, but suggest lower titers among children compared with adults, and, for some vaccines, lower efficacy in endemic settings compared with non-endemic settings. These disparities are likely multifactorial, and available evidence suggests a role for maternal factors (e.g. transplacental antibodies, breastmilk), host factors (e.g. genetic polymorphisms-with the best evidence for rotavirus-or previous infection), and environmental factors (e.g. gut microbiome, co-infections). Overall, these data highlight the rather ambiguous and often contradictory nature of evidence on factors affecting oral vaccine response, cautioning against broad extrapolation of outcomes based on one population or one vaccine type. Meaningful impact on performance of oral vaccines will likely only be possible with a suite of interventions, given the complex and multifactorial nature of the problem, and the degree to which contributing factors are intertwined.
Collapse
Affiliation(s)
- Rachel M Burke
- Global Development Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Julia Lynch
- Office of the Director General, International Vaccine Institute, Seoul, Republic of Korea
| | - Laura V Cooper
- School of Public Health, Imperial College London, London, UK
| | - Haeun Cho
- Department of Data Science and Innovation, International Vaccine Institute, Seoul, Republic of Korea
| | | | - Carl D Kirkwood
- Global Health Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - A Duncan Steele
- Global Health Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Gagandeep Kang
- Global Health Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| |
Collapse
|
18
|
Gebert JT, Scribano FJ, Engevik KA, Huleatt EM, Eledge MR, Dorn LE, Philip AA, Kawagishi T, Greenberg HB, Patton JT, Hyser JM. Viroporin activity is necessary for intercellular calcium signals that contribute to viral pathogenesis. SCIENCE ADVANCES 2025; 11:eadq8115. [PMID: 39823322 PMCID: PMC11740935 DOI: 10.1126/sciadv.adq8115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Viruses engage in a variety of processes to subvert host defenses and create an environment amenable to replication. Here, using rotavirus as a prototype, we show that calcium conductance out of the endoplasmic reticulum by the virus encoded ion channel, NSP4, induces intercellular calcium waves that extend beyond the infected cell and contribute to pathogenesis. Viruses that lack the ability to induce this signaling show diminished viral shedding and attenuated disease in a mouse model of rotavirus diarrhea. This implicates nonstructural protein 4 (NSP4) as a virulence factor and provides mechanistic insight into its mode of action. Critically, this signaling induces a transcriptional signature characteristic of interferon-independent innate immune activation, which is not observed in response to a mutant NSP4 that does not conduct calcium. This implicates calcium dysregulation as a means of pathogen recognition, a theme broadly applicable to calcium-altering pathogens beyond rotavirus.
Collapse
Affiliation(s)
- J. Thomas Gebert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Francesca J. Scribano
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kristen A. Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ethan M. Huleatt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael R. Eledge
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lauren E. Dorn
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Asha A. Philip
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Takahiro Kawagishi
- Departments of Medicine and Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Harry B. Greenberg
- Departments of Medicine and Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John T. Patton
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Joseph M. Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
19
|
Shafqat A, Li M, Zakirullah, Liu F, Tong Y, Fan J, Fan H. A comprehensive review of research advances in the study of lactoferrin to treat viral infections. Life Sci 2025; 361:123340. [PMID: 39730037 DOI: 10.1016/j.lfs.2024.123340] [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: 07/12/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Lactoferrin (Lf) is a naturally occurring glycoprotein known for its antiviral and antibacterial properties and is present in various physiological fluids. Numerous studies have demonstrated its antiviral effectiveness against multiple viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza virus (IFV), herpes simplex virus (HSV), hepatitis B virus (HBV), and human immunodeficiency virus (HIV). Lf, a vital component of the mucosal defense system, plays a crucial role in inhibiting viral infection by binding to both host cells and viral particles, such as the Hepatitis C virus (HCV). This interaction enables Lf to keep viral particles away from their target cells, emphasizing its significance as a fundamental element of mucosal defense against viral infections. Additionally, Lf has the ability to modulate cytokine expression and enhance cellular immune responses. In the innate immune system, Lf serves as a unique iron transporter and helps suppress various pathogens like bacteria, fungi, and viruses. This article summarises the potential antiviral properties of Lf against various viruses, along with its other mentioned functions. The advancement of Lf-based therapies supports the homology of food and medicine, providing a promising avenue to address viral infections and other public health challenges.
Collapse
Affiliation(s)
- Amna Shafqat
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zakirullah
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Feitong Liu
- H&H Group, H&H Research, China Research and Innovation, Guangzhou, China.
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Junfen Fan
- Institute of Cerebrovascular Diseases Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Huahao Fan
- School of Life Sciences, Tianjin University, Tianjin, China.
| |
Collapse
|
20
|
Zhu Y, Ren Q, Liu D, Jiang L, Yang Y, Qiu R, Li Z, Zhang M. Rotavirus-Inspired Nanointerface Engineered Biosensors for All-in-One Cancer Diagnosis. NANO LETTERS 2025; 25:461-469. [PMID: 39680916 DOI: 10.1021/acs.nanolett.4c05210] [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: 12/18/2024]
Abstract
Ultrasensitive and population-scale cancer screening technologies are critical to reducing cancer mortality. However, the current qRT-PCR falls short in high-throughput screening of multiple cancers. Here, a rotavirus-inspired multicancer diagnosis system (RMDS) is developed via nanointerface engineering. RMDS employs Y-shaped DNA (YDNA) probes to encircle the graphene quantum dots (GQDs) for nanointerface modification. The biotransduction mechanisms at the nanointerface are systematically investigated. RMDS greatly enhances the transduction efficiency of biological analytes by optimizing the probe density and configuration. RMDS realizes ultrasensitive detection of the lung cancer KARS G12D mutation with a limit of detection (LoD) of 5.7 aM and the breast cancer-related AKT2 gene (LoD: 3.0 aM). The multiple chambers enable simultaneous diagnosis of multiple cancers and determination of cancer progression. Clinical validation shows RMDS can be a practical solution, which could complement or replace qRT-PCR and become the next-generation all-in-one tool for large-scale population cancer screening.
Collapse
Affiliation(s)
- Yang Zhu
- School of Electronic and Computer Engineering, Peking University, Shenzhen 518055, China
| | - Qinqi Ren
- School of Electronic and Computer Engineering, Peking University, Shenzhen 518055, China
| | - Dexing Liu
- School of Electronic and Computer Engineering, Peking University, Shenzhen 518055, China
| | - Leying Jiang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University, Shenzhen 518055, China
| | - Yongsheng Yang
- School of Electronic and Computer Engineering, Peking University, Shenzhen 518055, China
| | - Rui Qiu
- School of Electronic and Computer Engineering, Peking University, Shenzhen 518055, China
| | - Zigang Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University, Shenzhen 518055, China
| | - Min Zhang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
21
|
Gyem K, Pelden S, Tshering D, Penjor K, Wangchuk R, Dorji S, Tenzin J, Phuyel BL. Aetiological, seasonal and antibiotic susceptibility patterns of diarrhoeal diseases in Bhutan (2016-2022): a retrospective study of surveillance data. BMJ Open 2025; 15:e086332. [PMID: 39762104 PMCID: PMC11749759 DOI: 10.1136/bmjopen-2024-086332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/22/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVES This study aimed to identify the aetiological spectrum, seasonal distribution and antimicrobial resistance patterns of diarrhoeal diseases in Bhutan. STUDY DESIGN AND SETTING The study used a cross-sectional, retrospective analysis of secondary data gathered through a passive, hospital-based sentinel surveillance for diarrhoeal disease across 12 hospitals, representing Bhutan's demographically diverse regions. PARTICIPANTS A total of 3429 participants' data of all age groups who presented with diarrhoea at sentinel hospitals between 1 January 1 2016 and 31 December 2022 were analysed. RESULTS Diarrhoeagenic Escherichia coli (DEC), Shigella, Salmonella and Aeromonas spp. were predominant bacterial pathogens, while Rotavirus, Astrovirus and Norovirus were the leading viral pathogens. Coinfections were observed in 195 cases. Children under nine were significantly affected than the other age groups. Seasonal trends revealed that bacterial pathogen incidence peaked during the summer/monsoon season, viral pathogens were more common in winter and spring, and parasites persisted year-round. Among the antibiotics tested, gentamicin, chloramphenicol, ceftriaxone and tetracycline exhibited high efficacy, with susceptibility rates of 93.4%, 87.2%, 81.5% and 69.5%, respectively. Conversely, high resistance rates were observed for amoxicillin (80.3%), ampicillin (77.4%) and nalidixic acid (69.5%). Multidrug resistance was prevalent, with β-lactamase production contributing to resistance rates of 80.7% to penicillin and 65.4% to fluoroquinolones groups. Cephalosporin resistance was also notable, with rates of 34.4% for cephalexin, 40.0% for cefazolin and 16.9% for ceftriaxone. CONCLUSIONS DEC and Rotavirus were identified as the leading causes of diarrhoea, with significant resistance patterns observed in common bacterial isolates. These findings underscore the need for DEC screening in paediatric cases and emphasise the need for sustained antimicrobial resistance surveillance.
Collapse
Affiliation(s)
- Kinley Gyem
- Enteric Zoonotic and Vector-Borne Disease Laboratory, Royal Centre for Disease Control, Thimphu, Bhutan
| | - Sonam Pelden
- Enteric Zoonotic and Vector-Borne Disease Laboratory, Royal Centre for Disease Control, Thimphu, Bhutan
| | - Dorji Tshering
- Enteric Zoonotic and Vector-Borne Disease Laboratory, Royal Centre for Disease Control, Thimphu, Bhutan
| | - Kinley Penjor
- Enteric Zoonotic and Vector-Borne Disease Laboratory, Royal Centre for Disease Control, Thimphu, Bhutan
| | - Rinzin Wangchuk
- Enteric Zoonotic and Vector-Borne Disease Laboratory, Royal Centre for Disease Control, Thimphu, Bhutan
| | - Sangay Dorji
- Enteric Zoonotic and Vector-Borne Disease Laboratory, Royal Centre for Disease Control, Thimphu, Bhutan
| | - Jigme Tenzin
- Enteric Zoonotic and Vector-Borne Disease Laboratory, Royal Centre for Disease Control, Thimphu, Bhutan
| | - Birdi Lal Phuyel
- Enteric Zoonotic and Vector-Borne Disease Laboratory, Royal Centre for Disease Control, Thimphu, Bhutan
| |
Collapse
|
22
|
Pandey A, Li Z, Gautam M, Ghosh A, Man SM. Molecular mechanisms of emerging inflammasome complexes and their activation and signaling in inflammation and pyroptosis. Immunol Rev 2025; 329:e13406. [PMID: 39351983 PMCID: PMC11742652 DOI: 10.1111/imr.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Inflammasomes are multi-protein complexes that assemble within the cytoplasm of mammalian cells in response to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), driving the secretion of the pro-inflammatory cytokines IL-1β and IL-18, and pyroptosis. The best-characterized inflammasome complexes are the NLRP3, NAIP-NLRC4, NLRP1, AIM2, and Pyrin canonical caspase-1-containing inflammasomes, and the caspase-11 non-canonical inflammasome. Newer inflammasome sensor proteins have been identified, including NLRP6, NLRP7, NLRP9, NLRP10, NLRP11, NLRP12, CARD8, and MxA. These inflammasome sensors can sense PAMPs from bacteria, viruses and protozoa, or DAMPs in the form of mitochondrial damage, ROS, stress and heme. The mechanisms of action, physiological relevance, consequences in human diseases, and avenues for therapeutic intervention for these novel inflammasomes are beginning to be realized. Here, we discuss these emerging inflammasome complexes and their putative activation mechanisms, molecular and signaling pathways, and physiological roles in health and disease.
Collapse
Affiliation(s)
- Abhimanu Pandey
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Zheyi Li
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Manjul Gautam
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Aritra Ghosh
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Si Ming Man
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| |
Collapse
|
23
|
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
|
24
|
Mohy A, Lagoubi Y, Gomez JA, Amadou B, Bouskraoui M. Health economic evaluation of 2-dose and 3-dose rotavirus vaccines in children below 5 years of age in Morocco. Hum Vaccin Immunother 2024; 20:2353480. [PMID: 38757507 PMCID: PMC11110695 DOI: 10.1080/21645515.2024.2353480] [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: 11/27/2023] [Accepted: 05/07/2024] [Indexed: 05/18/2024] Open
Abstract
Following the introduction of rotavirus vaccination into the Moroccan National Immunization Program, the prevalence of the disease has decreased by nearly 50%. However, evidence on the economic value of rotavirus vaccinations in Morocco is limited. This health economic analysis evaluated, from both country payer and societal perspectives, the costs and the cost-effectiveness of three rotavirus vaccines using a static, deterministic, population model in children aged < 5 years in Morocco. Included vaccines were HRV (2-dose schedule), HBRV (3-dose schedule) and BRV-PV 1-dose vial (3-dose schedule). One-way and probabilistic sensitivity analyses were conducted to assess the impact of uncertainty in model inputs. The model predicted that vaccination with HRV was estimated to result in fewer rotavirus gastroenteritis events (-194 homecare events, -57 medical visits, -8 hospitalizations) versus the 3-dose vaccines, translating into 7 discounted quality-adjusted life years gained over the model time horizon. HRV was associated with lower costs versus HBRV from both the country payer (-$1.8 M) and societal (-$4.1 M) perspectives, and versus BRV-PV 1-dose vial from the societal perspective (-$187,000), dominating those options in the cost-effectiveness analysis. However, costs of BRV-PV 1-dose vial were lower than HRV from the payer perspective, resulting in an ICER of approximately $328,376 per QALY, above the assumed cost effectiveness threshold of $3,500. Vaccination with a 2-dose schedule of HRV may be a cost-saving option and could lead to better health outcomes for children in Morocco versus 3-dose schedule rotavirus vaccines.
Collapse
Affiliation(s)
- Ahmed Mohy
- Value Evidence & Outcomes Emerging Markets, GSK, Wavre, Belgium
| | | | - Jorge A. Gomez
- Vaccines, Emerging Markets, GSK, Buenos Aires, Argentina
| | - Barry Amadou
- Vaccines Emerging Markets, GSK, Casablanca, Morocco
| | - Mohammed Bouskraoui
- Faculté de Médecine et de Pharmacie, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Chio CC, Chien JC, Chan HW, Huang HI. Overview of the Trending Enteric Viruses and Their Pathogenesis in Intestinal Epithelial Cell Infection. Biomedicines 2024; 12:2773. [PMID: 39767680 PMCID: PMC11672972 DOI: 10.3390/biomedicines12122773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Enteric virus infection is a major public health issue worldwide. Enteric viruses have become epidemic infectious diseases in several countries. Enteric viruses primarily infect the gastrointestinal tract and complete their life cycle in intestinal epithelial cells. These viruses are transmitted via the fecal-oral route through contaminated food, water, or person to person and cause similar common symptoms, including vomiting, abdominal pain, and diarrhea. Diarrheal disease is the third leading cause of death in children under five years of age, accounting for approximately 1.7 billion cases and 443,832 deaths annually in this age group. Additionally, some enteric viruses can invade other tissues, leading to severe conditions and even death. The pathogenic mechanisms of enteric viruses are also unclear. In this review, we organized the research on trending enteric virus infections, including rotavirus, norovirus, adenovirus, Enterovirus-A71, Coxsackievirus A6, and Echovirus 11. Furthermore, we discuss the gastrointestinal effects and pathogenic mechanisms of SARS-CoV-2 in intestinal epithelial cells, given the gastrointestinal symptoms observed during the COVID-19 pandemic. We conducted a literature review on their pathogenic mechanisms, which serves as a guide for formulating future treatment strategies for enteric virus infections.
Collapse
Affiliation(s)
- Chi-Chong Chio
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (C.-C.C.); (J.-C.C.); (H.-W.C.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Jou-Chun Chien
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (C.-C.C.); (J.-C.C.); (H.-W.C.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Hio-Wai Chan
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (C.-C.C.); (J.-C.C.); (H.-W.C.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Hsing-I Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (C.-C.C.); (J.-C.C.); (H.-W.C.)
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Kwei-Shan, Taoyuan 33305, Taiwan
| |
Collapse
|
28
|
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
|
29
|
Zhang X, Hao P, Mo J, Wang PY, Wang G, Li L, Zheng XJ, Yuan X, Yao W, Jin N, Li C, Ye XS. Local and Noninvasive Glyco-Virus Checkpoint Nanoblockades Restrict Sialylation for Prolonged Broad-Spectrum Epidemic Virus Therapy. ACS NANO 2024; 18:32910-32923. [PMID: 39536146 DOI: 10.1021/acsnano.4c12434] [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: 11/16/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has driven major advances in virus research. The role of glycans in viral infection has been revealed, with research demonstrating that terminal sialic acids are key receptors during viral attachment and infection into host cells. However, there is an urgent demand for universal tools to study the mechanism of sialic acids in viral infections, as well as to develop therapeutic agents against epidemic viruses through the downregulation of terminal sialic acid residues on glycans acting as a glyco-virus checkpoint to accelerate virus clearance. In this study, we developed a robust sialic acids blockade tool termed local and noninvasive glyco-virus checkpoint nanoblockades (LONG NBs), which blocked cell surface sialic acids by endogenously and continuously inhibiting the de novo sialic acids biosynthesis pathway. Furthermore, LONG NBs could accurately characterize the sialic acid-dependent profiles of multiple virus variants and protected the host against partial SARS-CoV-2, rotavirus, and influenza A virus infections after local and noninvasive administration. Our results suggest that LONG NBs represent a promising tool to facilitate in-depth research on the mechanism of viral infection, and serve as a broad-spectrum protectant against existing and emerging viral variants via glyco-virus checkpoint blockade.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Pengfei Hao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun 130000, China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun 130000, China
| | - Juan Mo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Peng-Yu Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Guoqing Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun 130000, China
| | - Letian Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun 130000, China
| | - Xiu-Jing Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Wenlong Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| | - Ningyi Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun 130000, China
| | - Chang Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun 130000, China
| | - Xin-Shan Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, and Chemical Biology Center, Peking University, Beijing 100191, China
| |
Collapse
|
30
|
Standaert B, Raes M, Ethgen O, Benninghoff B, Toumi M. Measuring the Vaccine Success Index: A Framework for Long-Term Economic Evaluation and Monitoring in the Case of Rotavirus Vaccination. Vaccines (Basel) 2024; 12:1265. [PMID: 39591168 PMCID: PMC11598573 DOI: 10.3390/vaccines12111265] [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: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
New vaccination programs measure economic success through cost-effectiveness analysis (CEA) based on an outcome evaluated over a certain time frame. The reimbursement price of the newly approved vaccine is then often reliant on a simulated ideal effect projection because of limited long-term data availability. This optimal cost-effectiveness result is later rarely adjusted to the observed effect measurements, barring instances of market competition-induced price erosion through the tender process. However, comprehensive and systematic monitoring of the vaccine effect (VE) for the evaluation of the real long-term economic success of vaccination is critical. It informs expectations about vaccine performance with success timelines for the investment. Here, an example is provided by a 15-year assessment of the rotavirus vaccination program in Belgium (RotaBIS study spanning 2005 to 2019 across 11 hospitals). The vaccination program started in late 2006 and yielded sub-optimal outcomes. Long-term VE surveillance data provided insights into the infection dynamics, disease progression, and vaccine performance. The presented analysis introduces novel conceptual frameworks and methodologies about the long-term economic success of vaccination programs. The CEA evaluates the initial target vaccination population, considering vaccine effectiveness compared with a historical unvaccinated group. Cost-impact analysis (CIA) covers a longer period and considers the whole vaccinated and unvaccinated population in which the vaccine has direct and indirect effects. The economic success index ratio of CIA over CEA outcomes evaluates long-term vaccination performance. Good performance is close to the optimal result, with an index value ≤1, combined with a low CEA. This measurement is a valuable aid for new vaccine introductions. It supports the establishment of robust monitoring protocols over time.
Collapse
Affiliation(s)
- Baudouin Standaert
- Department of Care & Ethics, Faculty of Medicine & Life Sciences, University of Hasselt, 3590 Diepenbeek, Belgium
| | - Marc Raes
- Department of Immunology & Infection, Faculty of Medicine & Life Sciences, University of Hasselt, 3590 Diepenbeek, Belgium;
| | - Olivier Ethgen
- Department of Public Health Sciences, Faculty of Medicine, University of Liège, 4000 Liège, Belgium;
| | | | - Mondher Toumi
- Public Health, University of Aix-Marseille, 13002 Marseille, France;
| |
Collapse
|
31
|
Liu XY, Xie W, Zhou HY, Zhang HQ, Jin YS. A comprehensive overview on antiviral effects of baicalein and its glucuronide derivative baicalin. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:621-636. [PMID: 39368944 DOI: 10.1016/j.joim.2024.09.003] [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: 09/23/2023] [Accepted: 09/07/2024] [Indexed: 10/07/2024]
Abstract
Natural product-based antiviral candidates have received significant attention. However, there is a lack of sufficient research in the field of antivirals to effectively combat patterns of drug resistance. Baicalein and its glucuronide derivative baicalin are two main components extracted from Scutellaria baicalensis Georgi. They have proven to be effective against a broad range of viruses by directly killing virus particles, protecting infected cells, and targeting viral antigens on their surface, among other mechanisms. As natural products, they both possess the advantage of lower toxicity, enhanced therapeutic efficacy, and even antagonistic effects against drug-resistant viral strains. Baicalein and baicalin exhibit promising potential as potent pharmacophore scaffolds, demonstrating their antiviral properties. However, to date, no review on the antiviral effects of baicalein and baicalin has been published. This review summarizes the recent research progress on antiviral effects of baicalein and baicalin against various types of viruses both in vitro and in vivo with a focus on the dosages and underlying mechanisms. The aim is to provide a basis for the rational development and utilization of baicalein and baicalin, as well as to promote antiviral drug research. Please cite this article as: Liu XY, Xie W, Zhou HY, Zhang HQ, Jin YS. A comprehensive overview on antiviral effects of baicalein and its glucuronide derivative baicalin. J Integr Med. 2024; 22(6): 621-636.
Collapse
Affiliation(s)
- Xin-Yang Liu
- School of Basic Medicine, Naval Medical University, Shanghai 200433, China
| | - Wei Xie
- Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200433, China
| | - He-Yang Zhou
- School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Hui-Qing Zhang
- Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200433, China.
| | - Yong-Sheng Jin
- School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
32
|
Riller Q, Schmutz M, Fourgeaud J, Fischer A, Neven B. Protective role of antibodies in enteric virus infections: Lessons from primary and secondary immune deficiencies. Immunol Rev 2024; 328:243-264. [PMID: 39340232 PMCID: PMC11659928 DOI: 10.1111/imr.13402] [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] [Indexed: 09/30/2024]
Abstract
Enteric viruses are the main cause of acute gastroenteritis worldwide with a significant morbidity and mortality, especially among children and aged adults. Some enteric viruses also cause disseminated infections and severe neurological manifestations such as poliomyelitis. Protective immunity against these viruses is not well understood in humans, with most knowledge coming from animal models, although the development of poliovirus and rotavirus vaccines has extended our knowledge. In a classical view, innate immunity involves the recognition of foreign DNA or RNA by pathogen recognition receptors leading to the production of interferons and other inflammatory cytokines. Antigen uptake and presentation to T cells and B cells then activate adaptive immunity and, in the case of the mucosal immunity, induce the secretion of dimeric IgA, the more potent immunoglobulins in viral neutralization. The study of Inborn errors of immunity (IEIs) offers a natural opportunity to study nonredundant immunity toward pathogens. In the case of enteric viruses, patients with a defective production of antibodies are at risk of developing neurological complications. Moreover, a recent description of patients with low or absent antibody production with protracted enteric viral infections associated with hepatitis reinforces the prominent role of B cells and immunoglobulins in the control of enteric virus.
Collapse
Affiliation(s)
- Quentin Riller
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine InstituteUniversité Paris Cité, INSERM UMR 1163ParisFrance
- IHU‐ImagineParisFrance
| | - Muriel Schmutz
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine InstituteUniversité Paris Cité, INSERM UMR 1163ParisFrance
- IHU‐ImagineParisFrance
| | - Jacques Fourgeaud
- Université Paris Cité, FETUSParisFrance
- Microbiology DepartmentAP‐HP, Hôpital NeckerParisFrance
| | - Alain Fischer
- Pediatric Hematology‐Immunology and Rheumatology UnitNecker‐Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
- INSERM UMRS 1163, Institut ImagineParisFrance
- Collège de FranceParisFrance
| | - Bénédicte Neven
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine InstituteUniversité Paris Cité, INSERM UMR 1163ParisFrance
- IHU‐ImagineParisFrance
- Pediatric Hematology‐Immunology and Rheumatology UnitNecker‐Children's Hospital, Assistance Publique‐Hôpitaux de ParisParisFrance
| |
Collapse
|
33
|
Sáez‐Fuertes L, Rio‐Aige K, Massot‐Cladera M, Castell M, Knipping K, Garssen J, Bourdet‐Sicard R, Rodríguez‐Lagunas MJ, Collado MC, Pérez‐Cano FJ. Bifidobacterium breve M-16 V and scGOS/lcFOS Supplementation to Dams Ameliorates Infant Rotavirus Infection in Early Life. Mol Nutr Food Res 2024; 68:e2400377. [PMID: 39468988 PMCID: PMC11605786 DOI: 10.1002/mnfr.202400377] [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: 09/29/2024] [Indexed: 10/30/2024]
Abstract
The immune system of newborns is underdeveloped, leaving them susceptible to infections like rotavirus (RV). Despite vaccines, RV remains a leading cause of child mortality, especially in developing countries. Maternal immunity is transferred during pregnancy and breastfeeding to the offspring providing protection against RV infection. This study aims to explore how the maternal diet can enhance the newborn's ability to fight early infections. Pregnant rats received orally Bifidobacterium breve M-16 V and short chain galacto-oligosaccharides (scGOS)/long chain fructo-oligosaccharides (lcFOS). At day 5 of life pups are infected with RV and at day 8, samples are collected for the infection analysis. Pups whose mothers received the synbiotic have lower RV infection severity. The levels of immunoglobulins (Ig) IgG2c and IgA are raised in pups' plasma and digested milk, respectively. Synbiotic supplementation improves intestinal maturation and increases gene expression of immune-related genes. In conclusion, the administration of this synbiotic to gestating and lactating mothers ameliorates the incidence and severity of the pup's diarrhea caused by the RV infection by improving their immunity.
Collapse
Affiliation(s)
- Laura Sáez‐Fuertes
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| | - Karla Rio‐Aige
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| | - Malén Massot‐Cladera
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
- Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Salud Carlos IIIMadrid28029Spain
| | - Karen Knipping
- Danone Research & InnovationUtrechtthe Netherlands
- Division of Pharmacology, Faculty of ScienceUtrecht Institute for Pharmaceutical SciencesUtrechtthe Netherlands
| | - Johan Garssen
- Danone Research & InnovationUtrechtthe Netherlands
- Division of Pharmacology, Faculty of ScienceUtrecht Institute for Pharmaceutical SciencesUtrechtthe Netherlands
| | | | - María José Rodríguez‐Lagunas
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| | - María Carmen Collado
- Institute of Agrochemistry and Food Technology (IATA‐CSIC), National Research CouncilValencia46980Spain
| | - Francisco José Pérez‐Cano
- Physiology Section, Department of Biochemistry and PhysiologyFaculty of Pharmacy and Food ScienceUniversity of Barcelona (UB)Barcelona08028Spain
- Nutrition and Food Safety Research Institute (INSA‐UB)Santa Coloma de Gramenet08921Spain
| |
Collapse
|
34
|
Harima H, Qiu Y, Sasaki M, Ndebe J, Penjaninge K, Simulundu E, Kajihara M, Ohnuma A, Matsuno K, Nao N, Orba Y, Takada A, Ishihara K, Hall WW, Hang'ombe BM, Sawa H. A first report of rotavirus B from Zambian pigs leading to the discovery of a novel VP4 genotype P[9]. Virol J 2024; 21:263. [PMID: 39449113 PMCID: PMC11515359 DOI: 10.1186/s12985-024-02533-5] [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: 08/11/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Rotavirus B (RVB) causes diarrhea in humans and pigs. Although various RVB strains were identified in humans and various animals globally, little is known about the epidemiology RVB infection in Africa. In this study, we attempted to examine the prevalence of RVB infection in pig populations in Zambia. METHODS Metagenomic analyses were conducted on pig feces collected in Zambia to detect double stranded RNA viruses, including RVB. To clarify the prevalence of RVB infection in pig populations in Zambia, 147 fecal samples were screened for the RVB detection by RT-qPCR. Full genome sequence of a detected RVB was determined by Sanger sequencing and genetically analyzed. RESULTS The metagenomic analyses revealed that RVB sequence reads and contigs of RVB were detected from one fecal sample collected from pigs in Zambia. RT-qPCR screening detected RVB genomes in 36.7% (54/147) of fecal samples. Among 54 positive samples, 13 were positive in non-diarrheal samples (n = 48, 27.1%) and 41 in diarrheal samples (n = 99, 41.4%). Genetic analyses demonstrated that all the segments of ZP18-18, except for VP4, had high nucleotide sequence identities (80.6-92.6%) with all other known RVB strains detected in pigs. In contrast, the VP4 sequence of ZP18-18 was highly divergent from other RVB strains (< 64.6% identities) and formed a distinct lineage in the phylogenetic tree. Notably, the VP8 subunit of the VP4 showed remarkably low amino acid identities (33.3%) to those of known RVB strains, indicating that the VP8 subunit of ZP18-18 was unique among RVB strains. According to the whole genome classification for RVB, ZP18-18 was assigned to a genotype constellation, G18-P[9]-I12-R4-C4-M4-A8-N10-T5-E4-H7 with the newly established VP4 genotype P[9]. CONCLUSIONS This current study updates the geographical distribution and the genetic diversity of RVB. Given the lack of information regarding RVB in Africa, further RVB surveillance is required to assess the potential risk to humans and animals.
Collapse
Affiliation(s)
- Hayato Harima
- Laboratory of Veterinary Public Health, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, 183-8509, Tokyo, Japan
| | - Yongjin Qiu
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- Laboratory of Parasitology, Department of Disease Control, Graduate School of Infectious Diseases, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, 060-0818, Japan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- Institute for Vaccine Research and Development, Hokkaido University, North 21 West 11, Kita-ku, Sapporo, 001-0021, Japan
| | - Joseph Ndebe
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- Department of Disease Control, School of Veterinary Medicine, the University of Zambia, Lusaka, 10101, Zambia
| | - Kapila Penjaninge
- Department of Disease Control, School of Veterinary Medicine, the University of Zambia, Lusaka, 10101, Zambia
| | - Edgar Simulundu
- Department of Disease Control, School of Veterinary Medicine, the University of Zambia, Lusaka, 10101, Zambia
- Macha Research Trust, Choma, 20100, Zambia
| | - Masahiro Kajihara
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
| | - Aiko Ohnuma
- Technical office, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001- 0020, Japan
| | - Keita Matsuno
- Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- One Health Research Center, Hokkaido University, Sapporo, 060-0818, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
| | - Naganori Nao
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- One Health Research Center, Hokkaido University, Sapporo, 060-0818, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- Institute for Vaccine Research and Development, Hokkaido University, North 21 West 11, Kita-ku, Sapporo, 001-0021, Japan
- One Health Research Center, Hokkaido University, Sapporo, 060-0818, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
| | - Ayato Takada
- Department of Disease Control, School of Veterinary Medicine, the University of Zambia, Lusaka, 10101, Zambia
- One Health Research Center, Hokkaido University, Sapporo, 060-0818, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- Africa Center of Excellence for Infectious Diseases of Humans and Animals, the University of Zambia, Lusaka, 10101, Zambia
| | - Kanako Ishihara
- Laboratory of Veterinary Public Health, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, 183-8509, Tokyo, Japan
| | - William W Hall
- Institute for Vaccine Research and Development, Hokkaido University, North 21 West 11, Kita-ku, Sapporo, 001-0021, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
- National Virus Reference Laboratory, School of Medicine, University College Dublin, Dublin, Ireland
| | - Bernard M Hang'ombe
- Africa Center of Excellence for Infectious Diseases of Humans and Animals, the University of Zambia, Lusaka, 10101, Zambia
- Department of Para-clinical Studies, School of Veterinary Medicine, the University of Zambia, Lusaka, 10101, Zambia
| | - Hirofumi Sawa
- Institute for Vaccine Research and Development, Hokkaido University, North 21 West 11, Kita-ku, Sapporo, 001-0021, Japan.
- Department of Disease Control, School of Veterinary Medicine, the University of Zambia, Lusaka, 10101, Zambia.
- One Health Research Center, Hokkaido University, Sapporo, 060-0818, Japan.
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan.
- Africa Center of Excellence for Infectious Diseases of Humans and Animals, the University of Zambia, Lusaka, 10101, Zambia.
| |
Collapse
|
35
|
Patton JT, Desselberger U. Rotaviruses and Rotavirus Vaccines: Special Issue Editorial. Viruses 2024; 16:1665. [PMID: 39599780 PMCID: PMC11598851 DOI: 10.3390/v16111665] [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: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
Species A rotaviruses (RVA) are a major cause of acute gastroenteritis in infants and young children and in the young of various mammalian and avian species [...].
Collapse
Affiliation(s)
- John T. Patton
- Department of Biology, Indiana University, 212 S Hawthorne Drive, Simon Hall 011, Bloomington, IN 47405, USA
| | - Ulrich Desselberger
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK;
| |
Collapse
|
36
|
Gou S, Liu Y, Li Q, Yang J, Qiu L, Zhao Y. CRISPR/Cas12 System-Based Assay for Rapid, Sensitive Detection of Rotavirus in Food Samples. Foodborne Pathog Dis 2024. [PMID: 39397560 DOI: 10.1089/fpd.2024.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Foodborne viruses have become an important threat to food safety and human health. Among the foodborne viruses, group A rotavirus is the most important pathogen of diarrhea in autumn and winter. The field detection of rotavirus is crucial for the early control of infection and patient management. Quantitative real-time reverse transcription-polymerase chain reaction is the most widely used in virus detection. However, the technique relies on high-cost instruments and trained personnel, which limit its use in field detection. In this study, we developed accurate, realizable, and simple detection methods by combining optimized CRISPR (clustered regularly interspaced short palindromic repeats) Cas12 and reverse transcription loop-mediated isothermal amplification (RT-LAMP) (reverse transcription loop-mediated isothermal amplification) to reduce the requirements for temperature control and costly real-time fluorescence polymerase chain reaction instruments. We investigated two nucleic acid detection systems combining RT-LAMP with CRISPR Cas12a and RT-LAMP with CRISPR Cas12b and compared them with reverse transcription-quantitative polymerase chain reaction. The resulting detection system only needs a reaction temperature and in single tube to react for 60 min with the detection sensitivity of 38 copies/μL. Overall, this study developed an innovative method for the rapid detection of rotavirus in food samples, which will help to effectively identify food contaminated by pathogens and prevent human infections and economic losses caused by disease outbreaks.
Collapse
Affiliation(s)
- Shirui Gou
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Yan Liu
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, China
| | - Qianqian Li
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| | - Jielin Yang
- Technology Center for Animal Plant and Food Inspection and Quarantine of Shanghai Customs, Shanghai, China
| | - Long Qiu
- Shanghai Tolo Biotechnology Co., Ltd, Shanghai, China
| | - Yu Zhao
- College of Life Sciences, Shanghai Normal University, Shanghai, China
| |
Collapse
|
37
|
Wu Q, Sun B, Hou J, Hui X, Wang C, Wang W, Ying W, Liu L, Zhu L, Wang Y, Li Q, Yu M, Zhou W, Chen Y, Wu B, Sun J, Zhou Q, Qian F, Wang X. Novel Compound Heterozygous Variants in the FAS Gene Lead to Fetal Onset of Autoimmune Lymphoproliferative Syndrome (ALPS). J Clin Immunol 2024; 45:23. [PMID: 39384643 DOI: 10.1007/s10875-024-01812-8] [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: 05/20/2024] [Accepted: 09/21/2024] [Indexed: 10/11/2024]
Abstract
OBJECTIVE FAS gene defects lead to autoimmune lymphoproliferative syndrome (ALPS), which is often inherited in an autosomal dominant and rarely in an autosomal recessive manner. We report a case of a newborn girl with novel compound heterozygous variants in FAS and reveal the underlying mechanism. METHODS Whole-exome sequencing (WES) was used to identify pathogenic variants. Multiparametric flow cytometry analysis, phosflow analysis, and FAS-induced apoptosis assays were used to explore the effects of the variants on FAS expression, apoptosis, and immunophenotype. The HEK293T cells were used to assess the impact of the variants on protein expression and FAS-induced apoptosis. RESULTS The patient was born with hepatosplenomegaly, anemia, and thrombocytopenia. She also experienced COVID-19, rotavirus infection, herpes simplex virus infection, and severe pneumonia. The proportion of double-negative T cells (DNTs) was significantly elevated. Novel FAS compound heterozygous variants c.310T > A (p.C104S) and c.702_704del (p.T235del) were identified. The apoptotic ability of T cells was defective, and FAS expression on the surface of T cells was deficient. The T235del variant decreased FAS expression, and the C104S protein remained in the endoplasmic reticulum (ER) and could not translocate to the cell surface. Both mutations resulted in loss-of-function in terms of FAS-induced apoptosis in HEK293T cells. The DNTs were mainly terminally differentiated T (TEMRA) and CD45RA+HLA-DR+, with high expression of CD85j, PD-1, and CD57. The percentage of Th1, Tfh, and autoreactive B cells were significantly increased in the patient. The abnormal immunophenotyping was partially attenuated by sirolimus treatment. CONCLUSIONS We identified two variants that significantly affect FAS expression or localization, leading to early disease onset of in the fetus. Abnormalities in the mTOR pathway are associated with a favorable response to sirolimus.
Collapse
Affiliation(s)
- Qi Wu
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Bijun Sun
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Jia Hou
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Xiaoying Hui
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Chenghao Wang
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Wenjie Wang
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Wenjing Ying
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Luyao Liu
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Li Zhu
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Ying Wang
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai, China
| | - Qifan Li
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Meiping Yu
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Weitao Zhou
- Department of Respiratory Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Yao Chen
- Department of Respiratory Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Bingbing Wu
- Clinical Genetic Center, Children's Hospital of Fudan University, Shanghai, China
| | - Jinqiao Sun
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Qinhua Zhou
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China.
| | - Feng Qian
- Ministry of Education Key Laboratory of Contemporary Anthropology, Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China.
| | - Xiaochuan Wang
- Department of Allergy and Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China.
| |
Collapse
|
38
|
Azzazy EA, Amer RM, Abdellatif GM, Abd-Elmoneim HA, Abo-Alella DA. Frequency and genotyping of group A rotavirus among Egyptian children with acute gastroenteritis: a hospital-based cross-sectional study. Virol J 2024; 21:238. [PMID: 39350262 PMCID: PMC11443952 DOI: 10.1186/s12985-024-02495-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND This hospital-based cross-sectional study aims to investigate the epidemiologic and clinical characteristics of rotavirus group A (RVA) infection among children with acute gastroenteritis and to detect the most common G and P genotypes in Egypt. METHODS A total of 92 stool samples were collected from children under five who were diagnosed with acute gastroenteritis. RVA in stool samples was identified using ELISA and nested RT-PCR. Common G and P genotypes were identified utilizing multiplex nested RT-PCR assays. RESULTS RVA was detected at a rate of 24% (22 /92) using ELISA and 26.1% (24 /92) using VP6 nested RT-PCR. The ELISA test demonstrated diagnostic sensitivity, specificity, and accuracy of 91.7%, 100%, and 97.8%, respectively. G3 was the most prevalent G type (37.5%), followed by G1 (12.5%), whereas the most commonly detected P type were P[8] (41.7%) and P[6] (8.2%). RVA-positive samples were significantly associated with younger aged children (p = 0.026), and bottle-fed (p = 0.033) children. In addition, RVA-positive samples were more common during cooler seasons (p = 0.0001). Children with rotaviral gastroenteritis had significantly more frequent episodes of diarrhea (10.87 ± 3.63 times/day) and vomiting (8.79 ± 3.57 times/day) per day (p = 0.013 and p = 0.011, respectively). Moreover, they had a more severe Vesikari clinical score (p = 0.049). CONCLUSION RVA is a prevalent cause of acute gastroenteritis among Egyptian children in our locality. The discovery of various RVA genotypes in the local population, as well as the identification of common G and P untypeable strains, highlights the significance of implementing the rotavirus vaccine in Egyptian national immunization programs accompanied by continuous monitoring of strains.
Collapse
Affiliation(s)
- Ensaf A Azzazy
- Medical Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Elsharkia Governorate, Zagazig, Egypt
| | - Rania M Amer
- Medical Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Elsharkia Governorate, Zagazig, Egypt
| | - Ghada Mohammed Abdellatif
- Pediatrics Department, Faculty of Medicine, Zagazig University, Elsharkia Governorate, Zagazig, Egypt
| | - Hala Adel Abd-Elmoneim
- Medical Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Elsharkia Governorate, Zagazig, Egypt
| | - Doaa Alhussein Abo-Alella
- Medical Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Elsharkia Governorate, Zagazig, Egypt.
| |
Collapse
|
39
|
Ozogul F, Rathod N, Köse S, Alak G, Kızılyıldırım S, Bilgin Ş, Emir Çoban Ö, İnanlı AG, Ünal-Şengör GF, İzci L, Ozogul Y, Tokur B, Ucak İ, Ceylan Z, Kulawik P. Biochemical and microbial food safety hazards in seafood: A Mediterranean perspective (Part 2). ADVANCES IN FOOD AND NUTRITION RESEARCH 2024; 114:209-271. [PMID: 40155085 DOI: 10.1016/bs.afnr.2024.09.003] [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: 04/01/2025]
Abstract
The marine environment is teeming with a diverse array of algae, dinoflagellates and phytoplankton. These organisms possess the remarkable capacity to produce toxic compounds that can be passed to humans through the ingestion of seafood, resulting in potential health risks. Similarly, seafood can be susceptible to contamination from various microorganisms, viruses and parasites, thereby, potentially compromising food safety. Consuming seafood that contains toxins or pathogenic microorganisms may have serious health consequences, including the potential for severe illness or even fatality. This chapter delves into the various hazards that arise from biochemical and microbiological factors, with particular emphasis on the Mediterranean region. In addition, it provides a succinct analysis regarding the effect of COVID-19 pandemic on the safety of seafood.
Collapse
Affiliation(s)
- Fatih Ozogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Çukurova University, Adana, Türkiye; Biotechnology Research and Application Center, Çukurova University, Adana, Türkiye.
| | - Nikheel Rathod
- Department of Post Harvest Management of Meat, Poultry and Fish, Post Graduate Institute of Post Harvest Technology and Management (Dr. Balasaheb Sawant Konkan Krishi Vidyapeeth), Raigad, Maharashtra, India.
| | - Sevim Köse
- Department of Fisheries Technology Engineering, Faculty of Marine Sciences, Karadeniz Technical University, Çamburnu, Trabzon, Türkiye
| | - Gonca Alak
- Department of Seafood Processing Technology, Faculty of Fisheries, Ataturk University, Erzurum, Türkiye
| | - Suna Kızılyıldırım
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Çukurova University, Adana, Türkiye
| | - Şengül Bilgin
- Eğirdir Fisheries Faculty, Isparta University of Applied Sciences, Isparta, Türkiye
| | - Özlem Emir Çoban
- Department of Seafood Processing Technology, Faculty of Fisheries, Fırat University, Elazığ, Türkiye
| | - Ayşe Gürel İnanlı
- Department of Seafood Processing Technology, Faculty of Fisheries, Fırat University, Elazığ, Türkiye
| | - Gülgün F Ünal-Şengör
- Division of Food Safety, Department of Fisheries and Seafood Processing Technology, Faculty of Aquatic Sciences, Istanbul University, İstanbul, Türkiye
| | - Levent İzci
- Eğirdir Fisheries Faculty, Isparta University of Applied Sciences, Isparta, Türkiye
| | - Yesim Ozogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Çukurova University, Adana, Türkiye.
| | - Bahar Tokur
- Fatsa Faculty of Marine Sciences, Ordu University, Ordu, Türkiye
| | - İlknur Ucak
- Faculty of Agricultural Sciences and Technologies, Nigde Omer Halisdemir University, Nigde, Türkiye
| | - Zafer Ceylan
- Department of Molecular Biology and Genetics/Biotechnology, Science Faculty, Bartın University, Bartın, Turkiye
| | - Piotr Kulawik
- Department of Animal Products Technology, Faculty of Food Technology, University of Agriculture in Cracow, Kraków, Poland.
| |
Collapse
|
40
|
Agbemabiese CA, Dennis FE, Lartey BL, Damanka SA, Nakagomi T, Nakagomi O, Armah GE. Whole Genome Sequences of the Wildtype AU-1 Rotavirus A Strain: The Prototype of the AU-1-like Genotype Constellation. Viruses 2024; 16:1529. [PMID: 39459863 PMCID: PMC11512235 DOI: 10.3390/v16101529] [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: 08/31/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Most human rotaviruses belong to the Wa-like, DS-1-like, or AU-1-like genotype constellation. The AU-1-like constellation, albeit minor, captured attention because its prototype strain AU-1 originated from feline rotavirus, leading to the concept of interspecies transmission of rotavirus. The AU-1 genome sequence determined by various laboratories over the years has documented two conflicting VP7 sequences in the GenBank. As culture-adaptation may introduce changes in the viral genome, the original fecal (wild-type) and the seed stock of culture-adapted AU-1 genomes were sequenced using the Illumina's MiSeq platform to determine the authentic AU-1 sequence and to identify what mutational changes were selected during cell-culture adaptation. The wild-type and culture-adapted AU-1 genomes were identical except for one VP4-P475L substitution. Their VP7 gene was 99.9% identical to the previously reported AU-1 VP7 under accession number AB792641 but only 92.5% to that under accession number D86271. Thus, the wild-type sequences determined in this study (accession numbers OR727616-OR727626) should be used as the reference. The VP4-P475L mutation was more likely incidental than inevitable during cell-culture adaptation. This was the first study in which the whole genomes of both wild-type and cultured RVA strains were simultaneously determined by deep sequencing.
Collapse
Affiliation(s)
- Chantal Ama Agbemabiese
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon P.O. Box LG581, Ghana (G.E.A.)
- Department of Hygiene and Molecular Epidemiology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (T.N.); (O.N.)
| | - Francis Ekow Dennis
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon P.O. Box LG581, Ghana (G.E.A.)
| | - Belinda Larteley Lartey
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon P.O. Box LG581, Ghana (G.E.A.)
| | - Susan Afua Damanka
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon P.O. Box LG581, Ghana (G.E.A.)
| | - Toyoko Nakagomi
- Department of Hygiene and Molecular Epidemiology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (T.N.); (O.N.)
| | - Osamu Nakagomi
- Department of Hygiene and Molecular Epidemiology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (T.N.); (O.N.)
| | - George Enyimah Armah
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon P.O. Box LG581, Ghana (G.E.A.)
| |
Collapse
|
41
|
Morgan B, Lyons EA, Handley A, Bogdanovic-Sakran N, Pavlic D, Witte D, Mandolo J, Turner A, Jere KC, Justice F, Ong DS, Bonnici R, Boniface K, Donato CM, Mpakiza A, Meyer A, Bar-Zeev N, Iturriza-Gomara M, Cunliffe NA, Danchin M, Bines JE. Rotavirus-Specific Maternal Serum Antibodies and Vaccine Responses to RV3-BB Rotavirus Vaccine Administered in a Neonatal or Infant Schedule in Malawi. Viruses 2024; 16:1488. [PMID: 39339964 PMCID: PMC11437397 DOI: 10.3390/v16091488] [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: 08/16/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
High titres of rotavirus-specific maternal antibodies may contribute to lower rotavirus vaccine efficacy in low- and middle-income countries (LMICs). RV3-BB vaccine (G3P[6]) is based on a neonatal rotavirus strain that replicates well in the newborn gut in the presence of breast milk. This study investigated the association between maternal serum antibodies and vaccine response in infants administered the RV3-BB vaccine. Serum was collected antenatally from mothers of 561 infants enrolled in the RV3-BB Phase II study conducted in Blantyre, Malawi, and analysed for rotavirus-specific serum IgA and IgG antibodies using enzyme-linked immunosorbent assay. Infant vaccine take was defined as cumulative IgA seroconversion (≥3 fold increase) and/or stool vaccine shedding. Maternal IgA or IgG antibody titres did not have a negative impact on vaccine-like stool shedding at any timepoint. Maternal IgG (but not IgA) titres were associated with reduced take post dose 1 (p < 0.005) and 3 (p < 0.05) in the neonatal vaccine schedule group but not at study completion (week 18). In LMICs where high maternal antibodies are associated with low rotavirus vaccine efficacy, RV3-BB in a neonatal or infant vaccine schedule has the potential to provide protection against severe rotavirus disease.
Collapse
Affiliation(s)
- Benjamin Morgan
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Eleanor A. Lyons
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Amanda Handley
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
- Medicines Development for Global Health, Melbourne, VIC 3001, Australia
| | - Nada Bogdanovic-Sakran
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Daniel Pavlic
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Desiree Witte
- Malawi Liverpool Welcome Trust Programme, Blantyre P.O. Box 30096, Chichi, Malawi; (D.W.); (J.M.); (K.C.J.)
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7ZX, UK; (A.T.); (M.I.-G.); (N.A.C.)
| | - Jonathan Mandolo
- Malawi Liverpool Welcome Trust Programme, Blantyre P.O. Box 30096, Chichi, Malawi; (D.W.); (J.M.); (K.C.J.)
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Ann Turner
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7ZX, UK; (A.T.); (M.I.-G.); (N.A.C.)
| | - Khuzwayo C. Jere
- Malawi Liverpool Welcome Trust Programme, Blantyre P.O. Box 30096, Chichi, Malawi; (D.W.); (J.M.); (K.C.J.)
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7ZX, UK; (A.T.); (M.I.-G.); (N.A.C.)
| | - Frances Justice
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Darren Suryawijaya Ong
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Rhian Bonnici
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Karen Boniface
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Celeste M. Donato
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
| | - Ashley Mpakiza
- Malawi Liverpool Welcome Trust Programme, Blantyre P.O. Box 30096, Chichi, Malawi; (D.W.); (J.M.); (K.C.J.)
| | - Anell Meyer
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
- Department of Gastroenterology and Clinical Nutrition, Royal Children’s Hospital, Parkville, VIC 3052, Australia
| | - Naor Bar-Zeev
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7ZX, UK; (A.T.); (M.I.-G.); (N.A.C.)
| | - Miren Iturriza-Gomara
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7ZX, UK; (A.T.); (M.I.-G.); (N.A.C.)
- GSK Vaccines for Global Health Institute, 53100 Sienna, Italy
| | - Nigel A. Cunliffe
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7ZX, UK; (A.T.); (M.I.-G.); (N.A.C.)
| | - Margaret Danchin
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
- GSK Vaccines for Global Health Institute, 53100 Sienna, Italy
- Department of General Medicine, Royal Children’s Hospital, Parkville, VIC 3052, Australia
| | - Julie E. Bines
- Enteric Diseases, Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia (A.H.); (N.B.-S.); (D.P.); (F.J.); (D.S.O.); (R.B.); (C.M.D.); (M.D.)
- Department of Gastroenterology and Clinical Nutrition, Royal Children’s Hospital, Parkville, VIC 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
| |
Collapse
|
42
|
Gleason A, Kumar CK, Klein E, Laxminarayan R, Nandi A. Effect of rotavirus vaccination on the burden of rotavirus disease and associated antibiotic use in India: A dynamic agent-based simulation analysis. Vaccine 2024; 42:126211. [PMID: 39137492 PMCID: PMC11385704 DOI: 10.1016/j.vaccine.2024.126211] [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: 05/21/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Rotavirus is a leading cause of diarrhea in infants and young children in many low- and middle-income countries. India launched a childhood immunization program for rotavirus in 2016, starting with four states and expanding it to cover all states by 2019. The objective of this study was to estimate the effects of the rotavirus vaccination program in India on disease burden and antibiotic misuse. METHODS We built a dynamic agent-based model of rotavirus progression in children under five within each district in India. Simulations were run for various scenarios of vaccination coverage in the context of India's Universal Immunization Programme. Population data were obtained from the National Family Household Surveys and used to calibrate the models. Disease parameters were obtained from published studies. We estimated past and projected future reduction of disease burden and antibiotic misuse due to full vaccination nationwide, by state, and by wealth quintile. RESULTS We estimate that rotavirus vaccination in India has reduced the prevalence of rotavirus cases by 33.7% (prediction interval: 30.7-36.0%), total antibiotic misuse due to rotavirus by 21.8% (18.6-25.1%), and total deaths due to rotavirus by 38.3% (31.3-44.4%) for children under five. We estimate total antibiotic misuse due to rotavirus infection to be 7.6% (7.5-7.9%) of total antibiotic consumption in this demographic versus 9.6% (9.4-9.9%) in the absence of vaccination. We project rotaviral prevalence to drop to below one case for every 100,000 individuals in those below five if vaccination coverage is increased by 50.3% (45.2-58.5%) to 68.1% (63.1-76.4) nationwide. CONCLUSION Universal coverage of childhood rotavirus vaccination can substantially reduce inappropriate antibiotic use in India.
Collapse
Affiliation(s)
- Alec Gleason
- High Meadows Environmental Institute, Princeton University, Princeton, NJ, USA
| | | | - Eili Klein
- One Health Trust, Washington, DC, USA; Department of Emergency Medicine, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ramanan Laxminarayan
- High Meadows Environmental Institute, Princeton University, Princeton, NJ, USA; One Health Trust, Bengaluru, India
| | - Arindam Nandi
- One Health Trust, Washington, DC, USA; Population Council, 1 Dag Hammarskjold Plaza, New York, NY 10017, United States.
| |
Collapse
|
43
|
Suzuki Y, Yaeshiro M, Uehara D, Ishihara R. Shared clusters between phylogenetic trees for genomic segments of Rotavirus A with distinct genotype constellations. GENE REPORTS 2024; 36:101956. [DOI: 10.1016/j.genrep.2024.101956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
44
|
Oluwaseun S, Cagnan L, Xausa I, Nachbar RB, Levy Bachelot L, Chen YH, Carias C. Projected Public Health Impact of a Universal Rotavirus Vaccination Program in France. Pediatr Infect Dis J 2024; 43:902-908. [PMID: 39163534 PMCID: PMC11319077 DOI: 10.1097/inf.0000000000004448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/27/2024]
Abstract
OBJECTIVE In June 2022, French health authorities issued a universal recommendation for routine administration and reimbursement of rotavirus vaccines in infants. Given this recent recommendation by French health authorities, we sought to understand the public health impact of a universal rotavirus vaccination strategy compared with no vaccination. MATERIALS AND METHODS A deterministic, age-structured, nonlinear dynamic transmission model, accounting for herd immunity, was developed. We considered 3 vaccination coverage scenarios: high (95%), medium (75%) and low (55%). Model parameter values were based on published modeling and epidemiological literature. Model outcomes included rotavirus gastroenteritis (RVGE) cases and healthcare resource utilization due to RVGE (hospitalizations, general practitioner or emergency department visits), as well as the number needed to vaccinate to prevent 1 RVGE case (mild or severe) and 1 RVGE-related hospitalization. Model calibration and analyses were conducted using Mathematica 11.3. RESULTS Over 5 years following implementation, RVGE cases for children under 5 years are estimated to be reduced by 84% under a high vaccination coverage scenario, by 72% under a medium vaccination coverage scenario and by 47% under a low vaccination coverage scenario. Across all scenarios, the number needed to vaccinate to avert 1 RVGE case and hospitalization varied between 1.86-2.04 and 24.15-27.44, respectively. CONCLUSIONS Rotavirus vaccination with high vaccination coverage in France is expected to substantially reduce the number of RVGE cases and associated healthcare resource utilization.
Collapse
Affiliation(s)
| | - Lauren Cagnan
- Market Access Department, MSD France, Courbevoie Cedex, France
| | - Ilaria Xausa
- Wolfram Solutions Department, Wolfram Research Inc, Champaign, IL
| | | | | | | | | |
Collapse
|
45
|
Uprety T, Soni S, Sreenivasan C, Hause BM, Naveed A, Ni S, Graves AJ, Morrow JK, Meade N, Mellits KH, Adam E, Kennedy MA, Wang D, Li F. Genetic and antigenic characterization of two diarrhoeicdominant rotavirus A genotypes G3P[12] and G14P[12] circulating in the global equine population. J Gen Virol 2024; 105:002016. [PMID: 39163114 PMCID: PMC11335307 DOI: 10.1099/jgv.0.002016] [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: 05/31/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
Equine rotavirus species A (ERVA) G3P[12] and G14P[12] are two dominant genotypes that cause foal diarrhoea with a significant economic impact on the global equine industry. ERVA can also serve as a source of novel (equine-like) rotavirus species A (RVA) reassortants with zoonotic potential as those identified previously in 2013-2019 when equine G3-like RVA was responsible for worldwide outbreaks of severe gastroenteritis and hospitalizations in children. One hurdle to ERVA research is that the standard cell culture system optimized for human rotavirus replication is not efficient for isolating ERVA. Here, using an engineered cell line defective in antiviral innate immunity, we showed that both equine G3P[12] and G14P[12] strains can be rapidly isolated from diarrhoeic foals. The genome sequence analysis revealed that both G3P[12] and G14P[12] strains share the identical genotypic constellation except for VP7 and VP6 segments in which G3P[12] possessed VP7 of genotype G3 and VP6 of genotype I6 and G14P[12] had the combination of VP7 of genotype G14 and VP6 of genotype I2. Further characterization demonstrated that two ERVA genotypes have a limited cross-neutralization. The lack of an in vitro broad cross-protection between both genotypes supported the increased recent diarrhoea outbreaks due to equine G14P[12] in foals born to dams immunized with the inactivated monovalent equine G3P[12] vaccine. Finally, using the structural modelling approach, we provided the genetic basis of the antigenic divergence between ERVA G3P[12] and G14P[12] strains. The results of this study will provide a framework for further investigation of infection biology, pathogenesis and cross-protection of equine rotaviruses.
Collapse
Affiliation(s)
- Tirth Uprety
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Shalini Soni
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Chithra Sreenivasan
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Ben M. Hause
- Department of Veterinary and Biomedical Sciences, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, 57007, USA
| | - Ahsan Naveed
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Shuisong Ni
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA
| | - Amy J. Graves
- Equine Diagnostic Solutions, LLC, 1501 Bull Lea Rd, Suite 104, Lexington, Kentucky 40511, USA
| | - Jennifer K. Morrow
- Equine Diagnostic Solutions, LLC, 1501 Bull Lea Rd, Suite 104, Lexington, Kentucky 40511, USA
| | - Nathan Meade
- Division of Microbiology, Brewing, and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Kenneth H. Mellits
- Division of Microbiology, Brewing, and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Emma Adam
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Michael A. Kennedy
- Department of Veterinary and Biomedical Sciences, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, 57007, USA
| | - Dan Wang
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Feng Li
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| |
Collapse
|
46
|
Tariq A, Piontkivska H. Reovirus infection induces transcriptome-wide unique A-to-I editing changes in the murine fibroblasts. Virus Res 2024; 346:199413. [PMID: 38848818 PMCID: PMC11225029 DOI: 10.1016/j.virusres.2024.199413] [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: 02/20/2024] [Revised: 05/26/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024]
Abstract
The conversion of Adenosine (A) to Inosine (I), by Adenosine Deaminases Acting on RNA or ADARs, is an essential post-transcriptional modification that contributes to proteome diversity and regulation in metazoans including humans. In addition to its transcriptome-regulating role, ADARs also play a major part in immune response to viral infection, where an interferon response activates interferon-stimulated genes, such as ADARp150, in turn dynamically regulating host-virus interactions. A previous report has shown that infection from reoviruses, despite strong activation of ADARp150, does not influence the editing of some of the major known editing targets, while likely editing others, suggesting a potentially nuanced editing pattern that may depend on different factors. However, the results were based on a handful of selected editing sites and did not cover the entire transcriptome. Thus, to determine whether and how reovirus infection specifically affects host ADAR editing patterns, we analyzed a publicly available deep-sequenced RNA-seq dataset, from murine fibroblasts infected with wild-type and mutant reovirus strains that allowed us to examine changes in editing patterns on a transcriptome-wide scale. To the best of our knowledge, this is the first transcriptome-wide report on host editing changes after reovirus infection. Our results demonstrate that reovirus infection induces unique nuanced editing changes in the host, including introducing sites uniquely edited in infected samples. Genes with edited sites are overrepresented in pathways related to immune regulation, cellular signaling, metabolism, and growth. Moreover, a shift in editing targets has also been observed, where the same genes are edited in infection and control conditions but at different sites, or where the editing rate is increased for some and decreased for other differential targets, supporting the hypothesis of dynamic and condition-specific editing by ADARs.
Collapse
Affiliation(s)
- Ayesha Tariq
- Department of Biological Sciences, Kent State University, Kent, OH, USA
| | - Helen Piontkivska
- Department of Biological Sciences, Kent State University, Kent, OH, USA; Brain Health Research Institute, Kent State University, Kent, OH, USA; Healthy Communities Research Institute, Kent State University, Kent, OH, USA.
| |
Collapse
|
47
|
Sadiq A, Khan T, Bostan N, Yinda CK, Matthijnssens J. Antigenic epitope analysis of Pakistani G3 and G9 rotavirus strains compared to vaccine strains revealed multiple amino acid differences. Diagn Microbiol Infect Dis 2024; 109:116346. [PMID: 38759540 DOI: 10.1016/j.diagmicrobio.2024.116346] [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: 05/27/2022] [Revised: 08/13/2023] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
Rotaviruses belong to genotype VP4-P[8] are a significant cause of severe loose diarrhea in infants and young children. In the present study, we characterised the complete genome of three of the Pakistani P[8]b RVA strains by Illumina HiSeq sequencing technology to determine the complete genotype constellation providing insight into the evolutionary dynamics of their genes using maximum likelihood analysis. The maximum genomic sequences of our study strains were similar to more recent human Wa-Like G1P[8]a, G3P[8]a, G4P[6], G4P[8], G9P[4], G9P[8]a, G11P[25],G12P[8]a and G12P[6] strains circulating around the world. Therefore, strains PAK274, PAK439 and PAK624 carry natively distinctive VP4 gene with universally common human Wa-Like genetic backbone. Comparing our study P[8]b strains with vaccines strains RotarixTM and RotaTeqTM, multiple amino acid differences were examined between vaccine virus antigenic epitopes and Pakistani isolates. Over time, these differences may result in the selection for strains that will escape the vaccine-induced RVA-neutralizing-antibody effect.
Collapse
Affiliation(s)
- Asma Sadiq
- Department of Microbiology, University of Jhang, Jhang, Pakistan
| | - Tariq Khan
- Department of Biosciences, COMSATS University (CUI), Park Road, Tarlai Kalan, Chak Shahzad, Islamabad,45550, Pakistan
| | - Nazish Bostan
- Department of Biosciences, COMSATS University (CUI), Park Road, Tarlai Kalan, Chak Shahzad, Islamabad,45550, Pakistan.
| | - Claude Kwe Yinda
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Viral Metagenomics, Leuven, Belgium
| | - Jelle Matthijnssens
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Viral Metagenomics, Leuven, Belgium
| |
Collapse
|
48
|
Munlela B, João ED, Strydom A, Bauhofer AFL, Chissaque A, Chilaúle JJ, Maurício IL, Donato CM, O’Neill HG, de Deus N. Whole-Genome Characterization of Rotavirus G9P[6] and G9P[4] Strains That Emerged after Rotavirus Vaccine Introduction in Mozambique. Viruses 2024; 16:1140. [PMID: 39066302 PMCID: PMC11281483 DOI: 10.3390/v16071140] [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: 05/15/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Mozambique introduced the Rotarix® vaccine into the National Immunization Program in September 2015. Following vaccine introduction, rotavirus A (RVA) genotypes, G9P[4] and G9P[6], were detected for the first time since rotavirus surveillance programs were implemented in the country. To understand the emergence of these strains, the whole genomes of 47 ELISA RVA positive strains detected between 2015 and 2018 were characterized using an Illumina MiSeq-based sequencing pipeline. Of the 29 G9 strains characterized, 14 exhibited a typical Wa-like genome constellation and 15 a DS-1-like genome constellation. Mostly, the G9P[4] and G9P[6] strains clustered consistently for most of the genome segments, except the G- and P-genotypes. For the G9 genotype, the strains formed three different conserved clades, separated by the P type (P[4], P[6] and P[8]), suggesting different origins for this genotype. Analysis of the VP6-encoding gene revealed that seven G9P[6] strains clustered close to antelope and bovine strains. A rare E6 NSP4 genotype was detected for strain RVA/Human-wt/MOZ/HCN1595/2017/G9P[4] and a genetically distinct lineage IV or OP354-like P[8] was identified for RVA/Human-wt/MOZ/HGJM0644/2015/G9P[8] strain. These results highlight the need for genomic surveillance of RVA strains detected in Mozambique and the importance of following a One Health approach to identify and characterize potential zoonotic strains causing acute gastroenteritis in Mozambican children.
Collapse
Affiliation(s)
- Benilde Munlela
- Instituto Nacional de Saúde (INS), Parcela 3943, Vila de Marracuene, Maputo 0205-02, Mozambique; (E.D.J.); (A.F.L.B.); (A.C.); (J.J.C.); (N.d.D.)
- Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - Eva D. João
- Instituto Nacional de Saúde (INS), Parcela 3943, Vila de Marracuene, Maputo 0205-02, Mozambique; (E.D.J.); (A.F.L.B.); (A.C.); (J.J.C.); (N.d.D.)
| | - Amy Strydom
- Department of Microbiology and Biochemistry, University of the Free State, 205 Nelson Mandela Avenue, Bloemfontein 9301, South Africa; (A.S.); (H.G.O.)
| | - Adilson Fernando Loforte Bauhofer
- Instituto Nacional de Saúde (INS), Parcela 3943, Vila de Marracuene, Maputo 0205-02, Mozambique; (E.D.J.); (A.F.L.B.); (A.C.); (J.J.C.); (N.d.D.)
- Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - Assucênio Chissaque
- Instituto Nacional de Saúde (INS), Parcela 3943, Vila de Marracuene, Maputo 0205-02, Mozambique; (E.D.J.); (A.F.L.B.); (A.C.); (J.J.C.); (N.d.D.)
- Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - Jorfélia J. Chilaúle
- Instituto Nacional de Saúde (INS), Parcela 3943, Vila de Marracuene, Maputo 0205-02, Mozambique; (E.D.J.); (A.F.L.B.); (A.C.); (J.J.C.); (N.d.D.)
| | - Isabel L. Maurício
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, 1349-008 Lisboa, Portugal;
| | - Celeste M. Donato
- The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, VIC 3000, Australia;
| | - Hester G. O’Neill
- Department of Microbiology and Biochemistry, University of the Free State, 205 Nelson Mandela Avenue, Bloemfontein 9301, South Africa; (A.S.); (H.G.O.)
| | - Nilsa de Deus
- Instituto Nacional de Saúde (INS), Parcela 3943, Vila de Marracuene, Maputo 0205-02, Mozambique; (E.D.J.); (A.F.L.B.); (A.C.); (J.J.C.); (N.d.D.)
- Departamento de Ciências Biológicas, Universidade Eduardo Mondlane, Julius Nyerere Avenue, Maputo 3453, Mozambique
| |
Collapse
|
49
|
Woodyear S, Chandler TL, Kawagishi T, Lonergan TM, Patel VA, Williams CA, Permar SR, Ding S, Caddy SL. Chimeric Viruses Enable Study of Antibody Responses to Human Rotaviruses in Mice. Viruses 2024; 16:1145. [PMID: 39066309 PMCID: PMC11281508 DOI: 10.3390/v16071145] [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: 05/14/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
The leading cause of gastroenteritis in children under the age of five is rotavirus infection, accounting for 37% of diarrhoeal deaths in infants and young children globally. Oral rotavirus vaccines have been widely incorporated into national immunisation programs, but whilst these vaccines have excellent efficacy in high-income countries, they protect less than 50% of vaccinated individuals in low- and middle-income countries. In order to facilitate the development of improved vaccine strategies, a greater understanding of the immune response to existing vaccines is urgently needed. However, the use of mouse models to study immune responses to human rotavirus strains is currently limited as rotaviruses are highly species-specific and replication of human rotaviruses is minimal in mice. To enable characterisation of immune responses to human rotavirus in mice, we have generated chimeric viruses that combat the issue of rotavirus host range restriction. Using reverse genetics, the rotavirus outer capsid proteins (VP4 and VP7) from either human or murine rotavirus strains were encoded in a murine rotavirus backbone. Neonatal mice were infected with chimeric viruses and monitored daily for development of diarrhoea. Stool samples were collected to quantify viral shedding, and antibody responses were comprehensively evaluated. We demonstrated that chimeric rotaviruses were able to efficiently replicate in mice. Moreover, the chimeric rotavirus containing human rotavirus outer capsid proteins elicited a robust antibody response to human rotavirus antigens, whilst the control chimeric murine rotavirus did not. This chimeric human rotavirus therefore provides a new strategy for studying human-rotavirus-specific immunity to the outer capsid, and could be used to investigate factors causing variability in rotavirus vaccine efficacy. This small animal platform therefore has the potential to test the efficacy of new vaccines and antibody-based therapeutics.
Collapse
Affiliation(s)
- Sarah Woodyear
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| | - Tawny L. Chandler
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| | - Takahiro Kawagishi
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63101, USA
| | - Tom M. Lonergan
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| | - Vanshika A. Patel
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| | | | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY 10001, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63101, USA
| | - Sarah L. Caddy
- Baker Institute for Animal Health, Cornell University, Ithaca, NY 14850, USA; (S.W.)
| |
Collapse
|
50
|
Fallah T, Mansour Ghanaiee R, Karimi A, Zahraei SM, Mahmoudi S, Alebouyeh M. Comparative analysis of the RVA VP7 and VP4 antigenic epitopes circulating in Iran and the Rotarix and RotaTeq vaccines. Heliyon 2024; 10:e33887. [PMID: 39071626 PMCID: PMC11282978 DOI: 10.1016/j.heliyon.2024.e33887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/10/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
Analyzing the lineages and detecting antigenic variation in immunogenic motifs of Group A Rotavirus (RVA) variants is crucial because it can impact vaccine efficacy. This study investigated the circulating lineages of VP4 and VP7 proteins of human RVA isolates and their phylogeny in ≤24-month-old symptomatic, rotavirus-positive children with transudative diarrhea within 48 h of admission to Mofid Children's Hospital between December 2020 and March 2022 in Tehran, Iran. Antigen detection was performed by ELISA, RNA extraction, and semi-nested multiplex PCR for G/P genotypes, followed by sequencing and bioinformatic analysis using multiple sequence alignments in MEGA and phylogenetic analysis by Geneious Prime. The similarity of VP7 and VP4 amino acids with the RotaTeq and Rotarix vaccine strains for cytotoxic T cell and antigenic epitopes was evaluated using the UCSF Chimera Molecular Modeling System. Overall, 27.3 % of the samples were RVA positive, showing untypeable (2.5 %), single (76.9 %), and mixed (20.5 %) genotypic characteristics. The strains clustered in the G1/II, G2/IV, G3/I, G4/I, G9/III, P (Kachooei et al., 2023) [8]/III, P (Howley et al., 2020) [4]/V, and P (Wahyuni et al., 2021) [6]/I lineages. Comparative analysis of VP7 antigenic epitopes showed that the G1/II strains were completely conserved, while the G2/IV, G3/I, G4/I, G6, G9/III strains contained 2, 3-5, 2, 4 and 9 amino acid substitutions, respectively. The P (Kachooei et al., 2023) [8]/III genotypes differed by 3 amino acids, while the P (Wahyuni et al., 2021) [6]/I genotype had the most substitutions. CTL epitopes were completely conserved in G3/I strains, but other genotypes differed by 1-4 amino acids compared to the vaccine strains. Given the diversity of circulating RVA genotypes and the observed mutations in neutralizing and CTL epitopes, immune escape by some of the strains is likely in Iran. This finding underscores the importance of evaluating the effect of rotavirus vaccines on local genotypes and related lineages before implementing a vaccination program.
Collapse
Affiliation(s)
- Tina Fallah
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Roxana Mansour Ghanaiee
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdollah Karimi
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohsen Zahraei
- Center for Communicable Diseases Control, Ministry of Health and Medical Education, Tehran, Iran
| | - Sussan Mahmoudi
- Center for Communicable Diseases Control, Ministry of Health and Medical Education, Tehran, Iran
| | - Masoud Alebouyeh
- Pediatric Infections Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|