1
|
Soni S, Kennedy MA, Wang D, Li F. The role and implication of rotavirus VP8∗ in viral infection and vaccine development. Virology 2025; 609:110563. [PMID: 40378555 DOI: 10.1016/j.virol.2025.110563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/08/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
Rotaviruses (RVs) are major causative agents of diarrhea in both humans and animals worldwide. Despite the successful development of live attenuated vaccines, the efficacy of these vaccines remains low in developing countries and RV infections still result in more than 200,000 deaths in children under 5 years old globally each year. These viruses are also an enteric pathogen for agricultural animals and have caused substantial economic losses annually to the animal livestock industry. Frequent reassortment and the emergence of new RV strains continue to pose a significant challenge to human and agricultural animal health. Attachment to susceptible cells by recognizing cell surface glycans is the first step of the RV lifecycle, which is directed by the RV spike protein VP8∗. VP8∗-host glycan receptor interactions are thought to be strain-specific and play an important role in RV replication fitness, tropism, and cross-species transmission. This review will summarize the current understanding of the roles of VP8∗ in engagement of glycan receptors and its functional consequences in impacting RV replication fitness and host ranges. The current progress towards developing a VP8∗-based RV vaccine is also discussed in the review.
Collapse
Affiliation(s)
- Shalini Soni
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA
| | - Michael A Kennedy
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Dan Wang
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA
| | - Feng Li
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY, 40546, USA.
| |
Collapse
|
2
|
Ghonaim AH, Rouby SR, Nageeb WM, Elgendy AA, Xu R, Jiang C, Ghonaim NH, He Q, Li W. Insights into recent advancements in human and animal rotavirus vaccines: Exploring new frontiers. Virol Sin 2025; 40:1-14. [PMID: 39672271 PMCID: PMC11962973 DOI: 10.1016/j.virs.2024.12.001] [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: 09/05/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024] Open
Abstract
Rotavirus infections cause severe gastroenteritis and dehydration in young children and animals worldwide, leading to high rates of morbidity and mortality, predominantly in low- and middle-income countries. In the past decade, substantial progress has been made in the development and implementation of rotavirus vaccines, which have been essential in alleviating the global burden of this disease, not only in human being but also in livestock species like calves and piglets, where these infections can cause significant economic losses. By synthesizing the latest research and real-world evidence, this review article is designated to provide deep insights into the current state of rotavirus vaccine technology and its global implementation as well as the application of rotavirus vaccines in veterinary settings and their importance in controlling zoonotic transmission and maintaining food security.
Collapse
Affiliation(s)
- Ahmed H Ghonaim
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Desert Research Center, Cairo 11435, Egypt
| | - Sherin R Rouby
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Wedad M Nageeb
- Medical Microbiology and Immunology Department, Faculty of Medicine, Suez Canal University, Ismailia 41111, Egypt
| | - Ashraf Ahmed Elgendy
- Department of Immunology, Faculty of Medicine, New Kaser Al-Aini Teaching Hospital, Cairo University, 11435, Egypt
| | - Rong Xu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Changsheng Jiang
- Anhui Provincial Key Laboratory of Animal Nutritional Regulation and Health, College of Animal Science, Anhui Science and Technology University, Fengyang 233100, China
| | - Noha H Ghonaim
- Family Medicine Department, Faculty of Medicine, Suez Canal University, Ismailia 41111, Egypt
| | - Qigai He
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China
| | - Wentao Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China.
| |
Collapse
|
3
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
4
|
Wu L, Jing Z, Pan Y, Guo L, Li Z, Feng L, Tian J. Emergence of a novel pathogenic porcine G1P[7] rotavirus in China. Virology 2024; 598:110185. [PMID: 39096775 DOI: 10.1016/j.virol.2024.110185] [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: 04/14/2024] [Revised: 07/07/2024] [Accepted: 07/20/2024] [Indexed: 08/05/2024]
Abstract
Among group A rotaviruses (RVAs), the G1 genotype is the main genotype causing diarrhea in children, but it has rarely been reported in pigs. During our epidemiological investigation, we detected G1P[7] rotavirus infection in piglets across several provinces in China and then isolated a porcine G1P[7] rotavirus strain (CN1P7). Sequencing revealed that the virus constellation was G1-P[7]-I5-R1-C1-M1-A8-N1-T1-E1-H1. Phylogenetic analyses revealed that CN1P7 most likely emerged due to genetic reassortment among porcine, human, giant panda and dog rotavirus strains. In vivo experiments were conducted on two-day-old piglets, which revealed that the CN1P7 strain was pathogenic to piglets. The virus was shed through the digestive tract and respiratory tract. In addition to the intestine, the CN1P7 strain displayed extraintestinal tropisms in piglets. Histopathological analysis revealed that the lung and small intestine were the targets of CN1P7. This study is the first to explore the molecular and pathogenic characterization of a pig-origin G1P[7] rotavirus.
Collapse
Affiliation(s)
- Ling Wu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zhaoyang Jing
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Yudi Pan
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Longjun Guo
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Zixin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| | - Jin Tian
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China.
| |
Collapse
|
5
|
Chen B, Yang Y, Wang Z, Dai X, Cao Y, Zhang M, Zhang D, Ni X, Zeng Y, Pan K. Surface Display of Duck Hepatitis A Virus Type 1 VP1 Protein on Bacillus subtilis Spores Elicits Specific Systemic and Mucosal Immune Responses on Mice. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10323-2. [PMID: 39002060 DOI: 10.1007/s12602-024-10323-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Duck viral hepatitis, primarily caused by duck hepatitis A virus type 1 (DHAV-1), poses a significant threat to the global duck industry. Bacillus subtilis is commonly utilized as a safe probiotic in the development of mucosal vaccines. In this study, a recombinant strain of B. subtilis, designated as B. subtilis RV, was constructed to display the DHAV-1 capsid protein VP1 on its spore surface using the outer coat protein B as an anchoring agent. The immunogenicity of this recombinant strain was evaluated in a mouse model through mixed feeding immunization. The results indicated that B. subtilis RV could elicit specific systemic and mucosal immune responses in mice, as evidenced by the high levels of serum IgG, intestinal secretory IgA, and potent virus-neutralizing antibodies produced. Furthermore, the recombinant strain significantly upregulated the expression levels of IL-2, IL-6, IL-10, TNF-α, and IFN-γ in the intestinal mucosa. Thus, the recombinant strain maintained the balance of the Th1/Th2 immune response and demonstrated an excellent mucosal immune adjuvant function. In summary, this study suggests that B. subtilis RV can be a novel alternative for effectively controlling DHAV-1 infection as a vaccine-based feed additive.
Collapse
Affiliation(s)
- Bin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Yang Yang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Zhenhua Wang
- College of Animal Husbandry and Veterinary, Chengdu Agricultural College, Chengdu, 611130, China
| | - Xixi Dai
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
- Chongqing Three Gorges Vocational College, Chongqing, 404155, China
| | - Yuheng Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Mengwei Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Dongmei Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Xueqin Ni
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Yan Zeng
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China.
| | - Kangcheng Pan
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang District, Chengdu, 611130, China.
| |
Collapse
|
6
|
Wu C, Fu Z, Xie C, Zhao J, He F, Jiao B, Jiao B. Epidemiological Characteristics and Genotypic Features of Rotavirus and Norovirus in Jining City, 2021-2022. Viruses 2024; 16:925. [PMID: 38932216 PMCID: PMC11209223 DOI: 10.3390/v16060925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Diarrhea, often caused by viruses like rotavirus (RV) and norovirus (NV), is a global health concern. This study focuses on RV and NV in Jining City from 2021 to 2022. Between 2021 and 2022, a total of 1052 diarrhea samples were collected. Real-Time Quantitative Fluorescent Reverse Transcriptase-PCR was used to detect RV-A, NV GI, and NV GII. For RV-A-positive samples, VP7 and VP4 genes were sequenced for genotype analysis, followed by the construction of evolutionary trees. Likewise, for NV-GII-positive samples, VP1 and RdRp genes were sequenced for genotypic analysis, and evolutionary trees were subsequently constructed. Between 2021 and 2022, Jining City showed varying detection ratios: RV-A alone (excluding co-infection of RV-A and NV GII) at 7.03%, NV GI at 0.10%, NV GII alone (excluding co-infection of RV-A and NV GII) at 5.42%, and co-infection of RV-A and NV GII at 1.14%. The highest RV-A ratios were shown in children ≤1 year and 2-5 years. Jining, Jinxiang County, and Liangshan County had notably high RV-A ratios at 24.37% (excluding co-infection of RV-A and NV GII) and 18.33% (excluding co-infection of RV-A and NV GII), respectively. Jining, Qufu, and Weishan had no RV-A positives. Weishan showed the highest NV GII ratios at 35.48% (excluding co-infection of RV-A and NV GII). Genotype analysis showed that, in 2021, G9P[8] and G2P[4] were dominant at 94.44% and 5.56%, respectively. In 2022, G8P[8], G9P[8], and G1P[8] were prominent at 75.86%, 13.79%, and 10.35%, respectively. In 2021, GII.3[P12], GII.4[P16], and GII.4[P31] constituted 71.42%, 14.29%, and 14.29%, respectively. In 2022, GII.3[P12] and GII.4[P16] accounted for 55.00% and 45.00%, respectively. RV-A and NV showed varying patterns for different time frames, age groups, and regions within Jining. Genotypic shifts were also observed in prevalent RV-A and NV GII strains in Jining City from 2021 to 2022. Ongoing monitoring of RV-A and NV is recommended for effective prevention and control.
Collapse
Affiliation(s)
- Changjing Wu
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining 272000, China; (C.W.); (C.X.); (J.Z.)
| | - Zhongyan Fu
- Department of Infectious Disease Control, Shandong Center for Disease Control and Prevention, Jinan 250001, China;
| | - Cuihua Xie
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining 272000, China; (C.W.); (C.X.); (J.Z.)
| | - Jian Zhao
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining 272000, China; (C.W.); (C.X.); (J.Z.)
| | - Feifei He
- Computer Information Technology, Northern Arizona University, Flagstaff, AZ 86011, USA;
| | - Boyan Jiao
- Department of Laboratory, Jining Center for Disease Control and Prevention, Jining 272000, China; (C.W.); (C.X.); (J.Z.)
| | - Baihai Jiao
- Department of Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06032, USA
| |
Collapse
|
7
|
Graikini D, García L, Abad I, Lavilla M, Puértolas E, Pérez MD, Sánchez L. Antirotaviral activity of dairy byproducts enriched in fractions from hyperimmune bovine colostrum: the effect of thermal and high hydrostatic pressure treatments. Food Funct 2024; 15:2265-2281. [PMID: 38319687 DOI: 10.1039/d3fo05250h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Nowadays, rotaviruses remain a major health burden, especially in developing countries, and strategies complementary to vaccination are needed. In this view, dairy fractions have attracted great scientific interest, due to their high content of bioactive compounds. The objective of this study was to evaluate the antiviral activity of whey and buttermilk enriched in proteins from hyperimmune bovine colostrum (HBC) against rotavirus. The enriched fractions were spray-dried and subsequently tested for their neutralizing activity against the bovine rotavirus WC3 strain in vitro, using differentiated Caco-2/TC7 cells. The highest antirotaviral activity was observed when whey and buttermilk were enriched in purified immunoglobulin G (IgG), showing complete rotavirus neutralization at concentrations of 3 and 6 mg mL-1 for whey and buttermilk, respectively. Additionally, the use of a crude immunoglobulin fraction also gave satisfactory results. The inhibitory activities of all samples significantly decreased after the application of heat, except for the IgG-enriched buttermilk which showed a slight increase of activity following the application of short-time treatments (75 or 85 °C for 20 s). This sample also showed a significant increase of activity (13%) after the application of low-intensity high hydrostatic pressure treatment (400 MPa for 5 min). The maximum loss of bioactivity was observed at 600 MPa for 10 min (31 and 20% for whey- and buttermilk-based formulas, respectively). This study provides relevant information on the potential of whey, buttermilk, and HBC to be part of functional products as complementary strategies to combat rotavirus infections.
Collapse
Affiliation(s)
- Dimitra Graikini
- Departmento de Producción Animal y Ciencia de los Alimentos, Universidad de Zaragoza, Zaragoza, Spain.
- Instituto Agroalimentario de Aragon (IA2), Zaragoza, Spain
| | - Laura García
- Departmento de Producción Animal y Ciencia de los Alimentos, Universidad de Zaragoza, Zaragoza, Spain.
| | - Inés Abad
- Departmento de Producción Animal y Ciencia de los Alimentos, Universidad de Zaragoza, Zaragoza, Spain.
- Instituto Agroalimentario de Aragon (IA2), Zaragoza, Spain
| | - María Lavilla
- AZTI-BRTA, Food Research-Parque Tecnológico de Bizkaia, Astondo Bidea, edificio 609, 48160 Derio, Spain
| | - Eduardo Puértolas
- AZTI-BRTA, Food Research-Parque Tecnológico de Bizkaia, Astondo Bidea, edificio 609, 48160 Derio, Spain
| | - María Dolores Pérez
- Departmento de Producción Animal y Ciencia de los Alimentos, Universidad de Zaragoza, Zaragoza, Spain.
- Instituto Agroalimentario de Aragon (IA2), Zaragoza, Spain
| | - Lourdes Sánchez
- Departmento de Producción Animal y Ciencia de los Alimentos, Universidad de Zaragoza, Zaragoza, Spain.
- Instituto Agroalimentario de Aragon (IA2), Zaragoza, Spain
| |
Collapse
|
8
|
Carossino M, Vissani MA, Barrandeguy ME, Balasuriya UBR, Parreño V. Equine Rotavirus A under the One Health Lens: Potential Impacts on Public Health. Viruses 2024; 16:130. [PMID: 38257830 PMCID: PMC10819593 DOI: 10.3390/v16010130] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/29/2023] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Group A rotaviruses are a well-known cause of viral gastroenteritis in infants and children, as well as in many mammalian species and birds, affecting them at a young age. This group of viruses has a double-stranded, segmented RNA genome with high genetic diversity linked to point mutations, recombination, and, importantly, reassortment. While initial molecular investigations undertaken in the 1900s suggested host range restriction among group A rotaviruses based on the fact that different gene segments were distributed among different animal species, recent molecular surveillance and genome constellation genotyping studies conducted by the Rotavirus Classification Working Group (RCWG) have shown that animal rotaviruses serve as a source of diversification of human rotavirus A, highlighting their zoonotic potential. Rotaviruses occurring in various animal species have been linked with contributing genetic material to human rotaviruses, including horses, with the most recent identification of equine-like G3 rotavirus A infecting children. The goal of this article is to review relevant information related to rotavirus structure/genomic organization, epidemiology (with a focus on human and equine rotavirus A), evolution, inter-species transmission, and the potential zoonotic role of equine and other animal rotaviruses. Diagnostics, surveillance and the current status of human and livestock vaccines against RVA are also reviewed.
Collapse
Affiliation(s)
- Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Maria Aldana Vissani
- Escuela de Veterinaria, Facultad de Ciencias Agrarias y Veterinarias, Universidad del Salvador, Pilar, Buenos Aires B1630AHU, Argentina; (M.A.V.); (M.E.B.)
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| | - Maria E. Barrandeguy
- Escuela de Veterinaria, Facultad de Ciencias Agrarias y Veterinarias, Universidad del Salvador, Pilar, Buenos Aires B1630AHU, Argentina; (M.A.V.); (M.E.B.)
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
| | - Udeni B. R. Balasuriya
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Viviana Parreño
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| |
Collapse
|
9
|
Kwong KWY, Xin Y, Lai NCY, Sung JCC, Wu KC, Hamied YK, Sze ETP, Lam DMK. Oral Vaccines: A Better Future of Immunization. Vaccines (Basel) 2023; 11:1232. [PMID: 37515047 PMCID: PMC10383709 DOI: 10.3390/vaccines11071232] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Oral vaccines are gaining more attention due to their ease of administration, lower invasiveness, generally greater safety, and lower cost than injectable vaccines. This review introduces certified oral vaccines for adenovirus, recombinant protein-based, and transgenic plant-based oral vaccines, and their mechanisms for inducing an immune response. Procedures for regulatory approval and clinical trials of injectable and oral vaccines are also covered. Challenges such as instability and reduced efficacy in low-income countries associated with oral vaccines are discussed, as well as recent developments, such as Bacillus-subtilis-based and nanoparticle-based delivery systems that have the potential to improve the effectiveness of oral vaccines.
Collapse
Affiliation(s)
- Keith Wai-Yeung Kwong
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Ying Xin
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
| | - Nelson Cheuk-Yin Lai
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Johnny Chun-Chau Sung
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
- Oristry BioTech (HK) Limited, Hong Kong, China
- Theratide BioTech (HK) Limited, Hong Kong, China
| | - Kam-Chau Wu
- Research Department, DreamTec Cytokines Limited, Hong Kong, China
| | | | - Eric Tung-Po Sze
- School of Science and Technology, Hong Kong Metropolitan University, Hong Kong, China
| | - Dominic Man-Kit Lam
- DrD Novel Vaccines Limited, Hong Kong, China
- Torsten Wiesel International Research Institute, Sichuan University, Chengdu 610064, China
| |
Collapse
|
10
|
Tatsi EB, Koukou DM, Dellis C, Dourdouna MM, Efthymiou V, Michos A, Syriopoulou V. Epidemiological study of unusual rotavirus strains and molecular characterization of emerging P[14] strains isolated from children with acute gastroenteritis during a 15-year period. Arch Virol 2023; 168:149. [PMID: 37129790 PMCID: PMC10151219 DOI: 10.1007/s00705-023-05769-8] [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: 10/25/2022] [Accepted: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Rotavirus group A (RVA) is characterized by molecular and epidemiological diversity. To date, 42 G and 58 P RVA genotypes have been identified, some of which, like P[14], have a zoonotic origin. In this study, we describe the epidemiology of unusual RVA genotypes and the molecular characteristics of P[14] strains. Fecal samples from children ≤ 16 years of age with acute gastroenteritis (AGE) who were hospitalized during 2007-2021 in Greece were tested for RVA by immunochromatography. Positive RVA samples were G and P genotyped, and part of the VP7 and VP4 genes were sequenced by the Sanger method. Epidemiological data were also recorded. Phylogenetic analysis of P[14] was performed using MEGA 11 software. Sixty-two (1.4%) out of 4427 children with RVA AGE were infected with an unusual G (G6/G8/G10) or P (P[6]/P[9]/P[10]/P[11]/P[14]) genotype. Their median (IQR) age was 18.7 (37.3) months, and 67.7% (42/62) were males. None of the children were vaccinated against RVA. P[9] (28/62; 45.2%) was the most common unusual genotype, followed by P[14] (12/62; 19.4%). In the last two years, during the period of the COVID-19 pandemic, an emergence of P[14] was observed (5/12, 41.6%) after an 8-year absence. The highest prevalence of P[14] infection was seen in the spring (91.7%). The combinations G8P[14] (41.7%), G6P[14] (41.7%), and G4P[14] (16.6%) were also detected. Phylogenetic analysis showed a potential evolutionary relationship of three human RVA P[14] strains to a fox strain from Croatia. These findings suggest a possible zoonotic origin of P[14] and interspecies transmission between nondomestic animals and humans, which may lead to new RVA genotypes with unknown severity.
Collapse
Affiliation(s)
- Elizabeth-Barbara Tatsi
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece.
- University Research Institute of Maternal and Child Health and Precision Medicine, Athens, Greece.
| | - Dimitra-Maria Koukou
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
| | - Charilaos Dellis
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
| | - Maria-Myrto Dourdouna
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
| | - Vasiliki Efthymiou
- University Research Institute of Maternal and Child Health and Precision Medicine, Athens, Greece
| | - Athanasios Michos
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
| | - Vasiliki Syriopoulou
- First Department of Pediatrics, Infectious Diseases and Chemotherapy Research Laboratory, Medical School, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
| |
Collapse
|
11
|
Antia A, Pinski AN, Ding S. Re-Examining Rotavirus Innate Immune Evasion: Potential Applications of the Reverse Genetics System. mBio 2022; 13:e0130822. [PMID: 35699371 PMCID: PMC9426431 DOI: 10.1128/mbio.01308-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Rotaviruses represent one of the most successful pathogens in the world, with high infectivity and efficient transmission between the young of many animal species, including humans. To overcome host defenses, rotaviruses have evolved a plethora of strategies to effectively evade the innate immune response, establish initial infection in the small intestine, produce progeny, and shed into the environment. Previously, studying the roles and relative contributions of specific rotaviral factors in innate immune evasion had been challenging without a plasmid-only reverse genetics system. Although still in its infancy, current reverse genetics technology will help address important research questions regarding rotavirus innate immune evasion, host range restriction, and viral pathogenesis. In this review, we summarize the current knowledge about the antiviral host innate immune defense mechanisms, countermeasures of rotavirus-encoded factors, and strategies to better understand these interactions using the rotavirus reverse genetics system.
Collapse
Affiliation(s)
- Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Amanda N. Pinski
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
12
|
Wu Z, Li Q, Liu Y, Lv H, Mo Z, Li F, Yu Q, Jin F, Chen W, Zhang Y, Huang T, Hu X, Xia W, Gao J, Zhou H, Bai X, Liu Y, Liang Z, Jiang Z, Chen Y, Zhang J, Du J, Yang B, Xing B, Xing Y, Dong B, Yang Q, Shi C, Yan T, Ruan B, Shi H, Fan X, Feng D, Lv W, Zhang D, Kong X, Zhou L, Que D, Chen H, Chen Z, Guo X, Zhou W, Wu C, Zhou Q, Liu Y, Qiao J, Wang Y, Li X, Duan K, Zhao Y, Yang X, Xu G. Efficacy, safety and immunogenicity of hexavalent rotavirus vaccine in Chinese infants. Virol Sin 2022; 37:724-730. [PMID: 35926726 PMCID: PMC9583109 DOI: 10.1016/j.virs.2022.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 07/19/2022] [Indexed: 11/26/2022] Open
Abstract
A randomized, double-blind, placebo-controlled multicenter trial was conducted in healthy Chinese infants to assess the efficacy and safety of a hexavalent live human-bovine reassortant rotavirus vaccine (HRV) against rotavirus gastroenteritis (RVGE). A total of 6400 participants aged 6–12 weeks were enrolled and randomly assigned to either HRV (n = 3200) or placebo (n = 3200) group. All the subjects received three oral doses of vaccine four weeks apart. The vaccine efficacy (VE) against RVGE caused by rotavirus serotypes contained in HRV was evaluated from 14 days after three doses of administration up until the end of the second rotavirus season. VE against severe RVGE, VE against RVGE hospitalization caused by serotypes contained in HRV, and VE against RVGE, severe RVGE, and RVGE hospitalization caused by natural infection of any serotype of rotavirus were also investigated. All adverse events (AEs) were collected for 30 days after each dose. Serious AEs (SAEs) and intussusception cases were collected during the entire study. Our data showed that VE against RVGE caused by serotypes contained in HRV was 69.21% (95%CI: 53.31–79.69). VE against severe RVGE and RVGE hospitalization caused by serotypes contained in HRV were 91.36% (95%CI: 78.45–96.53) and 89.21% (95%CI: 64.51–96.72) respectively. VE against RVGE, severe RVGE, and RVGE hospitalization caused by natural infection of any serotype of rotavirus were 62.88% (95%CI: 49.11–72.92), 85.51% (95%CI: 72.74–92.30) and 83.68% (95%CI: 61.34–93.11). Incidences of AEs from the first dose to one month post the third dose in HRV and placebo groups were comparable. There was no significant difference in incidences of SAEs in HRV and placebo groups. This study shows that this hexavalent reassortant rotavirus vaccine is an effective, well-tolerated, and safe vaccine for Chinese infants. A multicenter, double-blind, phase III clinical trial for the efficacy and safety of hexavalent rotavirus vaccine (HRV). The vaccine efficacy against rotavirus gastroenteritis caused by serotypes contained in HRV was 69.21%. The efficacy against severe rotavirus gastroenteritis and hospitalization caused by serotypesin HRV were 91.36% and 89.21%. No significant difference between the incidences of adverse events and severe adverse events in HRV and placebo group. This hexavalent live human-bovine reassortant rotavirus vaccine iseffective, well tolerated and safe in Chinese infants.
Collapse
Affiliation(s)
- Zhiwei Wu
- Hebei Center for Disease Control and Prevention, Shijiazhuang, 050021, China
| | - Qingliang Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Yan Liu
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Huakun Lv
- Zhejiang Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Zhaojun Mo
- Guangxi Zhuang Autonomous Region Center for Disease Control and Prevention, Nanning, 530028, China
| | - Fangjun Li
- Hunan Center for Disease Control and Prevention, Changsha, 410005, China
| | - Qingchuan Yu
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Fei Jin
- Hebei Center for Disease Control and Prevention, Shijiazhuang, 050021, China
| | - Wei Chen
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Yong Zhang
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Teng Huang
- Guangxi Zhuang Autonomous Region Center for Disease Control and Prevention, Nanning, 530028, China
| | - Xiaosong Hu
- Zhejiang Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Wei Xia
- Hunan Center for Disease Control and Prevention, Changsha, 410005, China
| | - Jiamei Gao
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Haisong Zhou
- Zhengding County Center for Disease Control and Prevention, Shijiazhuang, 050800, China
| | - Xuan Bai
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Yueyue Liu
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Zhenzhen Liang
- Zhejiang Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Zhijun Jiang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Yingping Chen
- Zhejiang Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Jiuwei Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Jialiang Du
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Biao Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Bo Xing
- Zhejiang Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Yantao Xing
- Daming County Center for Disease Control and Prevention, Handan, 056900, China
| | - Ben Dong
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Qinghai Yang
- Liucheng County Center for Disease Control and Prevention, Liuzhou, 545200, China
| | - Chen Shi
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Tingdong Yan
- Xiangtan County Center for Disease Control and Prevention, Xiangtan, 411228, China
| | - Bo Ruan
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Haiyun Shi
- Yuhuan County Center for Disease Control and Prevention, Taizhou, 317600, China
| | - Xingliang Fan
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Dongyang Feng
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Weigang Lv
- Yongnian County Center for Disease Control and Prevention, Handan, 056000, China
| | - Dong Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Xiangchu Kong
- Rongshui Miao Autonomous County Center for Disease Control and Prevention, Liuzhou, 545300, China
| | - Liuyifan Zhou
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Dinghong Que
- You County Center for Disease Control and Prevention, Zhuzhou, 412315, China
| | - Hong Chen
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Zhongbing Chen
- Longyou County Center for Disease Control and Prevention, Quzhou, 324400, China
| | - Xiang Guo
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Weiwei Zhou
- Laishui County Center for Disease Control and Prevention, Baoding 074100, China
| | - Cong Wu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Qingrong Zhou
- Jiangshan County Center for Disease Control and Prevention, Quzhou, 324100, China
| | - Yuqing Liu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Jian Qiao
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Ying Wang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Xinguo Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Kai Duan
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China
| | - Yuliang Zhao
- Hebei Center for Disease Control and Prevention, Shijiazhuang, 050021, China.
| | - Xiaoming Yang
- China National Biotec Group Company Limited, National Engineering Technology Research Center for Combined Vaccines, Wuhan, 430207, China.
| | - Gelin Xu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan Institute of Biological Product Co., Ltd., Wuhan, 430207, China.
| |
Collapse
|
13
|
Diebold O, Gonzalez V, Venditti L, Sharp C, Blake RA, Tan WS, Stevens J, Caddy S, Digard P, Borodavka A, Gaunt E. Using Species a Rotavirus Reverse Genetics to Engineer Chimeric Viruses Expressing SARS-CoV-2 Spike Epitopes. J Virol 2022; 96:e0048822. [PMID: 35758692 PMCID: PMC9327695 DOI: 10.1128/jvi.00488-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/31/2022] [Indexed: 02/02/2023] Open
Abstract
Species A rotavirus (RVA) vaccines based on live attenuated viruses are used worldwide in humans. The recent establishment of a reverse genetics system for rotoviruses (RVs) has opened the possibility of engineering chimeric viruses expressing heterologous peptides from other viral or microbial species in order to develop polyvalent vaccines. We tested the feasibility of this concept by two approaches. First, we inserted short SARS-CoV-2 spike peptides into the hypervariable region of the simian RV SA11 strain viral protein (VP) 4. Second, we fused the receptor binding domain (RBD) of the SARS-CoV-2 spike protein, or the shorter receptor binding motif (RBM) nested within the RBD, to the C terminus of nonstructural protein (NSP) 3 of the bovine RV RF strain, with or without an intervening Thosea asigna virus 2A (T2A) peptide. Mutating the hypervariable region of SA11 VP4 impeded viral replication, and for these mutants, no cross-reactivity with spike antibodies was detected. To rescue NSP3 mutants, we established a plasmid-based reverse genetics system for the bovine RV RF strain. Except for the RBD mutant that demonstrated a rescue defect, all NSP3 mutants delivered endpoint infectivity titers and exhibited replication kinetics comparable to that of the wild-type virus. In ELISAs, cell lysates of an NSP3 mutant expressing the RBD peptide showed cross-reactivity with a SARS-CoV-2 RBD antibody. 3D bovine gut enteroids were susceptible to infection by all NSP3 mutants, but cross-reactivity with SARS-CoV-2 RBD antibody was only detected for the RBM mutant. The tolerance of large SARS-CoV-2 peptide insertions at the C terminus of NSP3 in the presence of T2A element highlights the potential of this approach for the development of vaccine vectors targeting multiple enteric pathogens simultaneously. IMPORTANCE We explored the use of rotaviruses (RVs) to express heterologous peptides, using SARS-CoV-2 as an example. Small SARS-CoV-2 peptide insertions (<34 amino acids) into the hypervariable region of the viral protein 4 (VP4) of RV SA11 strain resulted in reduced viral titer and replication, demonstrating a limited tolerance for peptide insertions at this site. To test the RV RF strain for its tolerance for peptide insertions, we constructed a reverse genetics system. NSP3 was C-terminally tagged with SARS-CoV-2 spike peptides of up to 193 amino acids in length. With a T2A-separated 193 amino acid tag on NSP3, there was no significant effect on the viral rescue efficiency, endpoint titer, and replication kinetics. Tagged NSP3 elicited cross-reactivity with SARS-CoV-2 spike antibodies in ELISA. We highlight the potential for development of RV vaccine vectors targeting multiple enteric pathogens simultaneously.
Collapse
Affiliation(s)
- Ola Diebold
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Victoria Gonzalez
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Luca Venditti
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Colin Sharp
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Rosemary A. Blake
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Wenfang S. Tan
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Joanne Stevens
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Sarah Caddy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paul Digard
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| | - Alexander Borodavka
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Eleanor Gaunt
- Infection and Immunity Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, United Kingdom
| |
Collapse
|
14
|
Impact of Vaccination on Rotavirus Genotype Diversity: A Nearly Two-Decade-Long Epidemiological Study before and after Rotavirus Vaccine Introduction in Sicily, Italy. Pathogens 2022; 11:pathogens11040424. [PMID: 35456099 PMCID: PMC9028787 DOI: 10.3390/pathogens11040424] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Sicily was the first Italian region to introduce rotavirus (RV) vaccination with the monovalent G1P[8] vaccine Rotarix® in May 2012. In this study, the seasonal distribution and molecular characterization of RV strains detected over 19 years were compared to understand the effect of Rotarix® on the evolutionary dynamics of human RVs. A total of 7846 stool samples collected from children < 5 years of age, hospitalized with acute gastroenteritis, were tested for RV detection and genotyping. Since 2013, vaccine coverage has progressively increased, while the RV prevalence decreased from 36.1% to 13.3% with a loss of seasonality. The local distribution of RV genotypes changed over the time possibly due to vaccine introduction, with a drastic reduction in G1P[8] strains replaced by common and novel emerging RV strains, such as equine-like G3P[8] in the 2018−2019 season. Comparison of VP7 and VP4 amino acid (aa) sequences with the cognate genes of Rotarix® and RotaTeq® vaccine strains showed specific aa changes in the antigenic epitopes of VP7 and of the VP8* portion of VP4 of the Italian RV strains. Molecular epidemiological surveillance data are required to monitor the emergence of novel RV strains and ascertain if these strains may affect the efficacy of RV vaccines.
Collapse
|
15
|
Whole genome analysis of rotavirus strains circulating in Benin before vaccine introduction, 2016-2018. Virus Res 2022; 313:198715. [PMID: 35247484 DOI: 10.1016/j.virusres.2022.198715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/21/2022]
Abstract
Species A Rotaviruses (RVA) still play a major role in causing acute diarrhea in children under five years old worldwide. Currently, an 11-gene classification system is used to designate the full genotypic constellations of circulating strains. Viral proteins and non-structural proteins in the order VP7-VP4-VP6-VP1-VP2-VP3-NSP1-NSP2-NSP3-NSP4-NSP5/6 are represented by the genotypes Gx-P[x]-Ix-Rx-Cx-Mx-Ax-Nx-Tx-Ex-Hx, respectively. In Benin, ROTAVAC® vaccine was introduced into the Expanded Programme on Immunization in December 2019. To monitor circulating RVA strains for changes that may affect vaccine performance, in-depth analysis of strains prior to vaccine introduction are needed. Here we report, the whole-gene characterization (11 ORFs) for 72 randomly selected RVA strains of common and unusual genotypes collected in Benin from the 2016-2018 seasons. The sequenced strains were 15 G1P[8], 20 G2P[4], 5 G9P[8], 14 G12P[8], 9 G3P[6], 2 G1P[6], 3 G2P[6], 2 G9P[4], 1 G12P[6], and 1 G1G9P[8]/P[4]. The study strains exhibited two genetic constellations designed as Wa-like G1/G9/G12-P[6]/P[8]-I1-R1-C1-M1-A1-N1-T1-E1-H1 and DS-1-like G2/G3/G12-P[4]/P[6]-I2-R2-C2-M2-A2-N2-T2-E2-H2. Genotype G9P[4] strains possessed a DS-1-like genetic constellation with an E6 NSP4 gene, G9-P[4]-I2-R2-C2-M2-A2-N2-T2-E6-H2. The mixed genotype showed both Wa-like and DS-1-like profiles with a T6 NSP3 gene G1/G9P[8]/[4]-I1/I2-R1/R2-C1/C2-M1/M2-A1/A2-N1/N2-T1/T6-E1/E6-H1/H2. At the allelic level, the analysis of the Benin strains, reference strains (with known alleles), vaccine strains (with known alleles) identified 2-13 and 1-17 alleles for DS-1-like and Wa-like strains, respectively. Most of the study strains clustered into previously defined alleles, but we defined 3 new alleles for the VP7 (G3=1 new allele and G12=2 new alleles) and VP4 (P[4]=1 new allele and P[6]=2 new alleles) genes which formed the basis of the VP7 and VP4 gene clusters, respectively. For the remaining 9 genes, 0-6 new alleles were identified for both Wa-like and DS-1-like strains. This analysis of whole genome sequences of RVA strains circulating in Benin described genetic point mutations and reassortment events as well as novel alleles. Further detailed studies on these new alleles are needed and these data can also provide a baseline for studies on RVA in the post-vaccination period.
Collapse
|
16
|
Diller JR, Carter MH, Kanai Y, Sanchez SV, Kobayashi T, Ogden KM. Monoreassortant rotaviruses of multiple G types are differentially neutralized by sera from infants vaccinated with ROTARIX® and RotaTeq®. J Infect Dis 2021; 224:1720-1729. [PMID: 34628500 DOI: 10.1093/infdis/jiab479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/16/2021] [Indexed: 12/30/2022] Open
Abstract
Rotavirus is a leading cause of pediatric diarrheal mortality. The rotavirus outer capsid consists of VP7 and VP4 proteins, which respectively determine viral G and P type and are primary targets of neutralizing antibodies. To elucidate VP7-specific neutralizing antibody responses, we engineered monoreassortant rotaviruses each containing a human VP7 segment from a sequenced clinical specimen or a vaccine strain in an identical genetic background. We quantified replication and neutralization of engineered viruses using sera from infants vaccinated with monovalent ROTARIX® or multivalent RotaTeq® vaccines. Immunization with RotaTeq® induced broader neutralizing antibody responses than ROTARIX®. Inclusion of a single dose of RotaTeq® in the schedule enhanced G-type neutralization breadth of vaccinated infant sera. Cell type-specific differences in infectivity, replication, and neutralization were detected for some monoreassortant viruses. These findings suggest that rotavirus VP7, independent of VP4, can contribute to cell tropism and the breadth of vaccine-elicited neutralizing antibody responses.
Collapse
Affiliation(s)
- Julia R Diller
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maximilian H Carter
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yuta Kanai
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shania V Sanchez
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Takeshi Kobayashi
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kristen M Ogden
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Adeleke VT, Adeniyi AA, Adeleke MA, Okpeku M, Lokhat D. The design of multiepitope vaccines from plasmids of diarrheagenic Escherichia coli against diarrhoea infection: Immunoinformatics approach. INFECTION GENETICS AND EVOLUTION 2021; 91:104803. [PMID: 33684568 DOI: 10.1016/j.meegid.2021.104803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 01/19/2023]
Abstract
Diarrhoea infection is a major global health public problem and is caused by many organisms including diarrheagenic Escherichia coli pathotypes. The common problem with diarrhoea is the drug resistance of pathogenic bacteria, the most promising alternative means of preventing drug resistance is vaccination. However, there has not been any significant success in the prevention of diarrhoea caused by E. coli through vaccination. Epitope-based vaccine is gaining more attention due to its safety and specificity. Sequence variation of protective antigens of the pathogen has posed a new challenge in the development of epitope-based vaccines against the infection, leading to the necessity of multiepitope based design. In this study, immunoinformatics tools were used to design multiepitope vaccine candidates from plasmid genome sequences of multiple pathotypes of E. coli species involved in diarrhoea infections. The ability of the identified epitopes to be used as a cross-protect multiepitope vaccine was achieved by identifying conserved, immunogenic and antigenic peptides that can elicit CD4+ T-cell, CD8+ T-cell and B-cell and bind to MHC I and II HLA alleles. The molecular docking results of T-cell epitopes showed their well binding affinity to receptive protein and with a wider population coverage. The different multiepitope-based vaccines (MEVCs) candidates were constructed and based on the types of epitope linker they contained. The MEVCs exhibited very good binding interactions with the human immune receptor. Among multiepitope vaccines constructed, MEVC6, MEVCA and MEVCB are more promising as potential vaccine candidates for cross-protection against gastrointestinal infections according to the computational study. It is also hoped that after validation and testing, the predicted multiepitope-based vaccine candidates will probably resolve the challenge of immunological heterogeneity facing enteric vaccine development.
Collapse
Affiliation(s)
- Victoria T Adeleke
- Discipline of Chemical Engineering, University of KwaZulu-Natal, Howard Campus, Durban 4041, South Africa.
| | - Adebayo A Adeniyi
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, South Africa; Department of Industrial Chemistry, Federal University, Oye, Ekiti, Nigeria
| | - Matthew A Adeleke
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, P/Bag X54001, Durban 4000, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, P/Bag X54001, Durban 4000, South Africa
| | - David Lokhat
- Discipline of Chemical Engineering, University of KwaZulu-Natal, Howard Campus, Durban 4041, South Africa
| |
Collapse
|
18
|
Falkenhagen A, Huyzers M, van Dijk AA, Johne R. Rescue of Infectious Rotavirus Reassortants by a Reverse Genetics System Is Restricted by the Receptor-Binding Region of VP4. Viruses 2021; 13:v13030363. [PMID: 33668972 PMCID: PMC7996497 DOI: 10.3390/v13030363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/11/2021] [Accepted: 02/22/2021] [Indexed: 12/22/2022] Open
Abstract
The rotavirus species A (RVA) capsid contains the spike protein VP4, which interacts with VP6 and VP7 and is involved in cellular receptor binding. The capsid encloses the genome consisting of eleven dsRNA segments. Reassortment events can result in novel strains with changed properties. Using a plasmid-based reverse genetics system based on simian RVA strain SA11, we previously showed that the rescue of viable reassortants containing a heterologous VP4-encoding genome segment was strain-dependent. In order to unravel the reasons for the reassortment restrictions, we designed here a series of plasmids encoding chimeric VP4s. Exchange of the VP4 domains interacting with VP6 and VP7 was not sufficient for rescue of viable viruses. In contrast, the exchange of fragments encoding the receptor-binding region of VP4 resulted in virus rescue. All parent strains and the rescued reassortants replicated efficiently in MA-104 cells used for virus propagation. In contrast, replication in BSR T7/5 cells used for plasmid transfection was only efficient for the SA11 strain, whereas the rescued reassortants replicated slowly, and the parent strains failing to produce reassortants did not replicate. While future research in this area is necessary, replication in BSR T7/5 cells may be one factor that affects the rescue of RVAs.
Collapse
Affiliation(s)
- Alexander Falkenhagen
- Department of Biological Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany;
- Correspondence:
| | - Marno Huyzers
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, 2531 Potchefstroom, South Africa; (M.H.); (A.A.v.D.)
| | - Alberdina A. van Dijk
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, 2531 Potchefstroom, South Africa; (M.H.); (A.A.v.D.)
| | - Reimar Johne
- Department of Biological Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany;
| |
Collapse
|
19
|
Tokuhara D, Hikita N. Cord Blood-Based Approach to Assess Candidate Vaccine Adjuvants Designed for Neonates and Infants. Vaccines (Basel) 2021; 9:vaccines9020095. [PMID: 33514054 PMCID: PMC7911524 DOI: 10.3390/vaccines9020095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Neonates and infants are particularly susceptible to infections, for which outcomes tend to be severe. Vaccination is a key strategy for preventing infectious diseases, but the protective immunity achieved through vaccination typically is weaker in infants than in healthy adults. One possible explanation for the poor acquisition of vaccine-induced immunity in infants is that their innate immune response, represented by toll-like receptors, is immature. The current system for developing pediatric vaccines relies on the confirmation of their safety and effectiveness in studies involving the use of mature animals or adult humans. However, creating vaccines for neonates and infants requires an understanding of their uniquely immature innate immunity. Here we review current knowledge regarding the innate immune system of neonates and infants and challenges in developing vaccine adjuvants for those children through analyses of cord blood.
Collapse
|
20
|
Kanai Y, Kobayashi T. Rotavirus reverse genetics systems: Development and application. Virus Res 2021; 295:198296. [PMID: 33440223 DOI: 10.1016/j.virusres.2021.198296] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022]
Abstract
Rotaviruses (RVs) cause acute gastroenteritis in infants and young children. Since 2006, live-attenuated vaccines have reduced the number of RV-associated deaths; however, RV is still responsible for an estimated 228,047 annual deaths worldwide. RV, a member of the family Reoviridae, has an 11-segmented double-stranded RNA genome contained within a non-enveloped, triple layered virus particle. In 2017, a long-awaited helper virus-free reverse genetics system for RV was established. Since then, numerous studies have reported the generation of recombinant RVs; these studies verify the robustness of reverse genetics systems. This review provides technical insight into current reverse genetics systems for RVs, as well as discussing basic and applied studies that have used these systems.
Collapse
Affiliation(s)
- Yuta Kanai
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
21
|
Morozova OV, Sashina TF, Novikova NA. [Phylodynamic characteristics of the Russian population of rotavirus А (Reoviridae: Sedoreovirinae: Rotavirus) based on the VP6 gene]. Vopr Virusol 2021; 65:364-372. [PMID: 33533232 DOI: 10.36233/0507-4088-2020-65-6-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Rotavirus A is one of the leading causes of acute gastroenteritis in children in the first years of life. Rotavirus infection is currently classified as a preventable infection. The most abundant rotavirion protein is VP6. MATERIAL AND METHODS Phylogenetic analysis and calculation of phylodynamic characteristics were carried out for 262 nucleotide sequences of the VP6 gene of rotavirus species A, isolated in Russia, using the BEAST v.1.10.4 software package. The derivation and analysis of amino acid sequences was performed using the MEGAX program. RESULTS This study provides phylodynamic characteristics of the rotaviruses in Russia based on the sequences coding VP6 protein. Bayesian analysis showed the circulation of rotaviruses of three sublineages of genotype I1 and three sublineages of genotype I2 in Russia. The level of accumulation of mutations was established, which turned out to be similar for genotypes I1 and I2 and amounted to 7.732E-4 and 1.008E-3 nucleotides/site/year, respectively. The effective population sizes based on nucleotide sequences of the VP6 I1 and I2 genotypes are relatively stable while after the 2000s there is a tendency of its decreasing. Comparative analysis of the amino acid sequences in the region of the intracellular neutralization sites A (231-260 aa) and B (265-292 aa) made it possible to reveal a mutation in position V252I in a proportion of Russian strains of genotype I1 some strains of genotypes I1 and I2 had mutation I281V. These substitutions were not associated with any sublineages to which the strains belong. The analysis of three T-cell epitopes revealed four amino acid differences (in aa positions 305, 315, 342, 348) that were associated with the first or second genogroup. CONCLUSION Based on the phylodynamic characteristics and amino acid composition of antigenic determinants, it was concluded that the VP6 protein is highly stable and could potentially be a good model for development of a rotavirus vaccine.
Collapse
Affiliation(s)
- O V Morozova
- FSBI «Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology» of the Federal Service for Supervision of Consumer Rights Protection and Human Welfare
| | - T F Sashina
- FSBI «Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology» of the Federal Service for Supervision of Consumer Rights Protection and Human Welfare
| | - N A Novikova
- FSBI «Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology» of the Federal Service for Supervision of Consumer Rights Protection and Human Welfare
| |
Collapse
|
22
|
Abstract
Enteric viral and bacterial infections continue to be a leading cause of mortality and morbidity in young children in low-income and middle-income countries, the elderly, and immunocompromised individuals. Vaccines are considered an effective and practical preventive approach against the predominantly fecal-to-oral transmitted gastroenteritis particularly in the resource-limited countries or regions where implementation of sanitation systems and supply of safe drinking water are not quickly achievable. While vaccines are available for a few enteric pathogens including rotavirus and cholera, there are no vaccines licensed for many other enteric viral and bacterial pathogens. Challenges in enteric vaccine development include immunological heterogeneity among pathogen strains or isolates, a lack of animal challenge models to evaluate vaccine candidacy, undefined host immune correlates to protection, and a low protective efficacy among young children in endemic regions. In this article, we briefly updated the progress and challenges in vaccines and vaccine development for the leading enteric viral and bacterial pathogens including rotavirus, human calicivirus, Shigella, enterotoxigenic Escherichia coli (ETEC), cholera, nontyphoidal Salmonella, and Campylobacter, and introduced a novel epitope- and structure-based vaccinology platform known as MEFA (multiepitope fusion antigen) and the application of MEFA for developing broadly protective multivalent vaccines against heterogenous pathogens.
Collapse
Affiliation(s)
- Hyesuk Seo
- University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA
| | - Qiangde Duan
- University of Yangzhou, Institute of Comparative Medicine, Yangzhou, PR China
| | - Weiping Zhang
- University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA,CONTACT Weiping Zhang, University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA
| |
Collapse
|
23
|
Morozova OV, Sashina TA, Epifanova NV, Kashnikov AY, Novikova NA. Increasing detection of rotavirus G2P[4] strains in Nizhny Novgorod, Russia, between 2016 and 2019. Arch Virol 2020; 166:115-124. [PMID: 33079276 DOI: 10.1007/s00705-020-04853-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/10/2020] [Indexed: 01/28/2023]
Abstract
Rotavirus infection is one of the leading causes of acute gastroenteritis in children in their first years of life. We studied the genotypic diversity of rotavirus A (RVA) strains in Nizhny Novgorod, Russia, during the period 2016-19. In total, 4714 samples of faeces from children admitted to the Nizhny Novgorod Hospital for Infectious Diseases with acute gastroenteritis were examined. The share of rotavirus-positive samples was 31.5% in 2016-17. It decreased to 21.6% in 2018-19. In Nizhny Novgorod, all six global types of RVA were detected (G1P[8], G2P[4], G3P[8], G4P[8], G9P[8] and G12P[8]), as well as sporadic samples with genotypes G9P[4], G3P[9], G9P[9], G8P[8], G2P[8], G4P[4], G3P[9]. The fraction of strains with genotype G2P[4] gradually increased from 5.9% in 2016-17 to 39.1% in 2018-19. Simultaneously, the proportion of G9P[8] strains decreased from 63.2% to 27.7% in the same period. Phylogenetic analysis showed that rotaviruses with the G2P[4] genotype carried ubiquitous alleles of the VP7 and VP4 genes during the period of their prevalence: G2-IVa-1 and G2-IVa-3; P[4]-IVa and P[4]-IVb. As rotavirus vaccination is not widely used in the region because it is not included in the national vaccination calendar in Russia so far, the increase in the number of G2P[4] RVA is likely due to natural strain fluctuations.
Collapse
Affiliation(s)
- Olga V Morozova
- I.N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, 71 Malaya Yamskaya Str., Nizhny Novgorod, Russian Federation, 603950.
| | - Tatiana A Sashina
- I.N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, 71 Malaya Yamskaya Str., Nizhny Novgorod, Russian Federation, 603950
| | - Natalia V Epifanova
- I.N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, 71 Malaya Yamskaya Str., Nizhny Novgorod, Russian Federation, 603950
| | - Alexander Yu Kashnikov
- I.N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, 71 Malaya Yamskaya Str., Nizhny Novgorod, Russian Federation, 603950
| | - Nadezhda A Novikova
- I.N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, 71 Malaya Yamskaya Str., Nizhny Novgorod, Russian Federation, 603950
| |
Collapse
|
24
|
Mwangi PN, Mogotsi MT, Seheri ML, Mphahlele MJ, Peenze I, Esona MD, Kumwenda B, Steele AD, Kirkwood CD, Ndze VN, Dennis FE, Jere KC, Nyaga MM. Whole Genome In-Silico Analysis of South African G1P[8] Rotavirus Strains Before and After Vaccine Introduction Over A Period of 14 Years. Vaccines (Basel) 2020; 8:E609. [PMID: 33066615 PMCID: PMC7712154 DOI: 10.3390/vaccines8040609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/08/2020] [Accepted: 10/13/2020] [Indexed: 12/03/2022] Open
Abstract
Rotavirus G1P[8] strains account for more than half of the group A rotavirus (RVA) infections in children under five years of age, globally. A total of 103 stool samples previously characterized as G1P[8] and collected seven years before and seven years after introducing the Rotarix® vaccine in South Africa were processed for whole-genome sequencing. All the strains analyzed had a Wa-like constellation (G1-P[8]-I1-R1-C1-M1-A1-N1-T1-E1-H1). South African pre- and post-vaccine G1 strains were clustered in G1 lineage-I and II while the majority (84.2%) of the P[8] strains were grouped in P[8] lineage-III. Several amino acid sites across ten gene segments with the exception of VP7 were under positive selective pressure. Except for the N147D substitution in the antigenic site of eight post-vaccine G1 strains when compared to both Rotarix® and pre-vaccine strains, most of the amino acid substitutions in the antigenic regions of post-vaccine G1P[8] strains were already present during the pre-vaccine period. Therefore, Rotarix® did not appear to have an impact on the amino acid differences in the antigenic regions of South African post-vaccine G1P[8] strains. However, continued whole-genome surveillance of RVA strains to decipher genetic changes in the post-vaccine period remains imperative.
Collapse
Affiliation(s)
- Peter N. Mwangi
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; (P.N.M.); (M.T.M.)
| | - Milton T. Mogotsi
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; (P.N.M.); (M.T.M.)
| | - Mapaseka L. Seheri
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa 0204, South Africa; (M.L.S.); (M.J.M.); (I.P.); (M.D.E.)
| | - M. Jeffrey Mphahlele
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa 0204, South Africa; (M.L.S.); (M.J.M.); (I.P.); (M.D.E.)
- South African Medical Research Council, Pretoria 0001, South Africa
| | - Ina Peenze
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa 0204, South Africa; (M.L.S.); (M.J.M.); (I.P.); (M.D.E.)
| | - Mathew D. Esona
- Diarrheal Pathogens Research Unit, Sefako Makgatho Health Sciences University, Medunsa 0204, South Africa; (M.L.S.); (M.J.M.); (I.P.); (M.D.E.)
| | - Benjamin Kumwenda
- College of Medicine, Department of Biomedical Sciences, Faculty of Biomedical Sciences and Health Professions, University of Malawi, Private Bag 360, Chichiri, Blantyre 3, Malawi;
| | - A. Duncan Steele
- Enteric and Diarrheal Diseases, Global Health, Bill & Melinda Gates Foundation, P.O. Box 23350, Seattle, WA 98109, USA; (A.D.S.); (C.D.K.)
| | - Carl D. Kirkwood
- Enteric and Diarrheal Diseases, Global Health, Bill & Melinda Gates Foundation, P.O. Box 23350, Seattle, WA 98109, USA; (A.D.S.); (C.D.K.)
| | - Valantine N. Ndze
- Faculty of Health Sciences, University of Buea, P.O. Box 63, Buea, Cameroon;
| | - Francis E. Dennis
- Noguchi Memorial Institute for Medical Research, University of Ghana, P.O. Box LG581, Legon, Ghana;
| | - Khuzwayo C. Jere
- Center for Global Vaccine Research, Institute of Infection, Liverpool L697BE, UK;
- Veterinary and Ecological Sciences, University of Liverpool, Liverpool L697BE, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Program, Department of Medical Laboratory Sciences, College of Medicine, University of Malawi, Blantyre 312225, Malawi
| | - Martin M. Nyaga
- Next Generation Sequencing Unit and Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa; (P.N.M.); (M.T.M.)
| |
Collapse
|
25
|
Overview of the Development, Impacts, and Challenges of Live-Attenuated Oral Rotavirus Vaccines. Vaccines (Basel) 2020; 8:vaccines8030341. [PMID: 32604982 PMCID: PMC7565912 DOI: 10.3390/vaccines8030341] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Safety, efficacy, and cost-effectiveness are paramount to vaccine development. Following the isolation of rotavirus particles in 1969 and its evidence as an aetiology of severe dehydrating diarrhoea in infants and young children worldwide, the quest to find not only an acceptable and reliable but cost-effective vaccine has continued until now. Four live-attenuated oral rotavirus vaccines (LAORoVs) (Rotarix®, RotaTeq®, Rotavac®, and RotaSIIL®) have been developed and licensed to be used against all forms of rotavirus-associated infection. The efficacy of these vaccines is more obvious in the high-income countries (HIC) compared with the low- to middle-income countries (LMICs); however, the impact is far exceeding in the low-income countries (LICs). Despite the rotavirus vaccine efficacy and effectiveness, more than 90 countries (mostly Asia, America, and Europe) are yet to implement any of these vaccines. Implementation of these vaccines has continued to suffer a setback in these countries due to the vaccine cost, policy, discharging of strategic preventive measures, and infrastructures. This review reappraises the impacts and effectiveness of the current live-attenuated oral rotavirus vaccines from many representative countries of the globe. It examines the problems associated with the low efficacy of these vaccines and the way forward. Lastly, forefront efforts put forward to develop initial procedures for oral rotavirus vaccines were examined and re-connected to today vaccines.
Collapse
|
26
|
Song Y, Feng N, Sanchez-Tacuba L, Yasukawa LL, Ren L, Silverman RH, Ding S, Greenberg HB. Reverse Genetics Reveals a Role of Rotavirus VP3 Phosphodiesterase Activity in Inhibiting RNase L Signaling and Contributing to Intestinal Viral Replication In Vivo. J Virol 2020; 94:e01952-19. [PMID: 32051268 PMCID: PMC7163120 DOI: 10.1128/jvi.01952-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/07/2020] [Indexed: 12/13/2022] Open
Abstract
Our understanding of how rotavirus (RV) subverts host innate immune signaling has greatly increased over the past decade. However, the relative contribution of each virus-encoded innate immune antagonist has not been fully studied in the context of RV infection in vivo Here, we present both in vitro and in vivo evidence that the host interferon (IFN)-inducible 2'-5'-oligoadenylate synthetase (OAS) and RNase L pathway effectively suppresses the replication of heterologous RV strains. VP3 from homologous RVs relies on its 2'-5'-phosphodiesterase (PDE) domain to counteract RNase L-mediated antiviral signaling. Using an RV reverse-genetics system, we show that compared to the parental strain, VP3 PDE mutant RVs replicated at low levels in the small intestine and were shed less in the feces of wild-type mice, and such defects were rescued in Rnasel-/- suckling mice. Collectively, these findings highlight an important role of VP3 in promoting viral replication and pathogenesis in vivo in addition to its well-characterized function as the viral RNA-capping enzyme.IMPORTANCE Rotaviruses are significant human pathogens that result in diarrhea, dehydration, and deaths in many children around the world. Rotavirus vaccines have suboptimal efficacy in low- to middle-income countries, where the burden of the diseases is the most severe. With the ultimate goal of improving current vaccines, we aim to better understand how rotavirus interacts with the host innate immune system in the small intestine. Here, we demonstrate that interferon-activated RNase L signaling blocks rotavirus replication in a strain-specific manner. In addition, virus-encoded VP3 antagonizes RNase L activity both in vitro and in vivo These studies highlight an ever-evolving arms race between antiviral factors and viral pathogens and provide a new means of targeted attenuation for next-generation rotavirus vaccine design.
Collapse
Affiliation(s)
- Yanhua Song
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, California, USA
- Palo Alto Veterans Institute of Research, VA Palo Alto Health Care System, Palo Alto, California, USA
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Ningguo Feng
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, California, USA
- Palo Alto Veterans Institute of Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Liliana Sanchez-Tacuba
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, California, USA
- Palo Alto Veterans Institute of Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Linda L Yasukawa
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, California, USA
- Palo Alto Veterans Institute of Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Lili Ren
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | - Robert H Silverman
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Harry B Greenberg
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, California, USA
- Palo Alto Veterans Institute of Research, VA Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
27
|
Kanchan V, Zaman K, Aziz AB, Zaman SF, Zaman F, Haque W, Khanam M, Karim MM, Kale S, Ali SK, Goveia MG, Kaplan SS, Gill D, Khan WA, Yunus M, Singh A, Clemens JD. A randomized Phase I/II study to evaluate safety and reactogenicity of a heat-stable rotavirus vaccine in healthy adults followed by evaluation of the safety, reactogenicity, and immunogenicity in infants. Hum Vaccin Immunother 2020; 16:693-702. [PMID: 31526218 PMCID: PMC7227685 DOI: 10.1080/21645515.2019.1664239] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Objectives: To assess the safety and reactogenicity of single oral dose of heat-stable rotavirus vaccine (HSRV) in healthy adults aged 18–45 years followed by assessment of safety, reactogenicity, and immunogenicity of three doses of HSRV in healthy infants aged 6–8 weeks at enrollment. Trial Design: Single-center randomized controlled, sequential, blinded (adults) and open-label (infants). Setting: Single site at International Center for Diarrheal Disease Research, Bangladesh (icddr,b). Participants: Fifty eligible adults randomized in 1:1 ratio (HSRV: Placebo) followed by 50 eligible infants randomized in 1:1 ratio (HSRV: Comparator (RotaTeq®, pentavalent human-bovine (WC3) reassortant live-attenuated, rotavirus vaccine)). Intervention: Adults received either a single dose of HSRV or placebo and followed for 14 days. Infants received three doses of either HSRV or comparator with a follow-up for 28 days after each dose. Main Outcome Measures: Solicited and unsolicited adverse events (AEs) along with any serious adverse events (SAEs) were part of the safety and reactogenicity assessment in adults and infants whereas serum anti-rotavirus IgA response rates were part of immunogenicity assessment in infants only. Post-vaccination fecal shedding of vaccine-virus rotavirus strains was also determined in adults and infants. Results: In this study, HSRV, when compared with placebo, did not result in increase in solicited adverse events (solicited AEs) in adults. In infants, HSRV had a safety profile similar to comparator vis-à-vis solicited AEs. In infants, fecal shedding of vaccine-virus strains was not detected in HSRV recipients but was observed in two comparator recipients. Percentage of infants exhibiting threefold rise in serum anti-rotavirus IgA titers from baseline to 1-month post-dose 3 in HSRV group was 88% (22/25) and 84% (21/25) in comparator group. Conclusion: HSRV was found to be generally well-tolerated in both adults and infants and immunogenic in infants.
Collapse
Affiliation(s)
- Vibhu Kanchan
- MSD Wellcome Trust Hilleman Laboratories Pvt. Ltd., New Delhi, India
| | | | | | | | | | | | | | | | - Sachin Kale
- MSD Wellcome Trust Hilleman Laboratories Pvt. Ltd., New Delhi, India
| | - Syed Khalid Ali
- MSD Wellcome Trust Hilleman Laboratories Pvt. Ltd., New Delhi, India
| | | | | | - Davinder Gill
- MSD Wellcome Trust Hilleman Laboratories Pvt. Ltd., New Delhi, India
| | | | | | - Ajitpal Singh
- MSD Wellcome Trust Hilleman Laboratories Pvt. Ltd., New Delhi, India
| | | |
Collapse
|
28
|
Smith MR, Bugada LF, Wen F. Rapid microsphere-assisted peptide screening (MAPS) of promiscuous MHCII-binding peptides in Zika virus envelope protein. AIChE J 2020; 66:e16697. [PMID: 33343002 PMCID: PMC7747769 DOI: 10.1002/aic.16697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/06/2019] [Indexed: 12/31/2022]
Abstract
Despite promising developments in computational tools, peptide-class II MHC (MHCII) binding predictors continue to lag behind their peptide-class I MHC counterparts. Consequently, peptide-MHCII binding is often evaluated experimentally using competitive binding assays, which tend to sacrifice throughput for quantitative binding detail. Here, we developed a high-throughput semiquantitative peptide-MHCII screening strategy termed microsphere-assisted peptide screening (MAPS) that aims to balance the accuracy of competitive binding assays with the throughput of computational tools. Using MAPS, we screened a peptide library from Zika virus envelope (E) protein for binding to four common MHCII alleles (DR1, DR4, DR7, DR15). Interestingly, MAPS revealed a significant overlap between peptides that promiscuously bind multiple MHCII alleles and antibody neutralization sites. This overlap was also observed for rotavirus outer capsid glycoprotein VP7, suggesting a deeper relationship between B cell and CD4+ T cell specificity which can facilitate the design of broadly protective vaccines to Zika and other viruses.
Collapse
Affiliation(s)
- Mason R. Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Luke F. Bugada
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
- Catalysis Science and Technology Institute, University of Michigan, Ann Arbor, Michigan
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
- Catalysis Science and Technology Institute, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
29
|
Haidara FC, Tapia MD, Sow SO, Doumbia M, Coulibaly F, Diallo F, Traoré A, Kodio M, Kelly CL, Fitzpatrick M, Kotloff K, Victor JC, Neuzil K. Evaluation of a Booster Dose of Pentavalent Rotavirus Vaccine Coadministered With Measles, Yellow Fever, and Meningitis A Vaccines in 9-Month-Old Malian Infants. J Infect Dis 2019; 218:606-613. [PMID: 29659924 PMCID: PMC6047426 DOI: 10.1093/infdis/jiy215] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/10/2018] [Indexed: 12/03/2022] Open
Abstract
Background Rotavirus vaccines given to infants are safe and efficacious. A booster dose of rotavirus vaccine could extend protection into the second year of life in low-resource countries. Methods We conducted an open-label, individual-randomized trial in Bamako, Mali. We assigned 600 infants aged 9–11 months to receive measles vaccine (MV), yellow fever vaccine (YFV), and meningococcal A conjugate vaccine (MenAV) with or without pentavalent rotavirus vaccine (PRV). We assessed the noninferiority (defined as a difference of ≤10%) of seroconversion and seroresponse rates to MV, YFV, and MenAV. We compared the seroresponse to PRV. Results Seroconversion to MV occurred in 255 of 261 PRV recipients (97.7%) and 246 of 252 control infants (97.6%; difference, 0.1% [95% confidence interval {CI}, −4.0%–4.2%]). Seroresponse to YFV occurred in 48.1% of PRV recipients (141 of 293), compared with 52.2% of controls (153 of 293; difference, −4.1% [95% CI, −12.2%–4.0%]). A 4-fold rise in meningococcus A bactericidal titer was observed in 273 of 292 PRV recipients (93.5%) and 276 of 293 controls (94.2%; difference, −0.7% [95% CI, −5.2%–3.8%]). Rises in geometric mean concentrations of immunoglobulin A and immunoglobulin G antibodies to rotavirus were higher among PRV recipients (118 [95% CI, 91–154] and 364 [95% CI, 294–450], respectively), compared with controls (68 [95% CI, 50–92] and 153 [95% CI, 114–207], respectively). Conclusions PRV did not interfere with MV and MenAV; this study could not rule out interference with YFV. PRV increased serum rotavirus antibody levels. Clinical Trials Registration NCT02286895.
Collapse
Affiliation(s)
| | - Milagritos D Tapia
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Samba O Sow
- Centre pour le Développement des Vaccins-Mali, Bamako.,Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | | | | | | | - Awa Traoré
- Centre pour le Développement des Vaccins-Mali, Bamako
| | | | | | - Meagan Fitzpatrick
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | - Karen Kotloff
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | | | - Kathleen Neuzil
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| |
Collapse
|
30
|
Dual Recognition of Sialic Acid and αGal Epitopes by the VP8* Domains of the Bovine Rotavirus G6P[5] WC3 and of Its Mono-reassortant G4P[5] RotaTeq Vaccine Strains. J Virol 2019; 93:JVI.00941-19. [PMID: 31243129 PMCID: PMC6714814 DOI: 10.1128/jvi.00941-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 06/18/2019] [Indexed: 01/02/2023] Open
Abstract
Group A rotaviruses initiate infection through the binding of the VP8* domain of the VP4 protein to sialic acids (SAs) or histo-blood group antigens (HBGAs). Although the bovine G6P[5] WC3 strain is an important animal pathogen and is used as the backbone in the bovine-human reassortant RotaTeq vaccine, the receptor(s) for their P[5] VP8* domain has remained elusive. Using a variety of approaches, we demonstrated that the WC3 and bovine-human mono-reassortant G4P[5] vaccine strains recognize both α2,6-linked SA and αGal HBGA as ligands. Neither ligand is expressed on human small intestinal epithelial cells, explaining the absence of natural human infection by P[5]-bearing strains. However, we observed that the P[5]-bearing WC3 and G4P[5] RotaTeq vaccine strains could still infect human intestinal epithelial cells. Thus, the four P[5] RotaTeq vaccine strains potentially binding to additional alternative receptors may be efficient and effective in providing protection against severe rotavirus disease in human. Group A rotaviruses, an important cause of severe diarrhea in children and young animals, initiate infection via interactions of the VP8* domain of the VP4 spike protein with cell surface sialic acids (SAs) or histo-blood group antigens (HBGAs). Although the bovine G6P[5] WC3 strain is an important animal pathogen and is also used in the bovine-human reassortant RotaTeq vaccine, the receptor(s) for the VP8* domain of WC3 and its reassortant strains have not yet been identified. In the present study, HBGA- and saliva-binding assays showed that both G6P[5] WC3 and mono-reassortant G4P[5] strains recognized the αGal HBGA. The infectivity of both P[5]-bearing strains was significantly reduced in αGal-free MA-104 cells by pretreatment with a broadly specific neuraminidase or by coincubation with the α2,6-linked SA-specific Sambucus nigra lectin, but not by the α2,3-linked specific sialidase or by Maackia amurensis lectin. Free NeuAc and the αGal trisaccharide also prevented the infectivity of both strains. This indicated that both P[5]-bearing strains utilize α2,6-linked SA as a ligand on MA104 cells. However, the two strains replicated in differentiated bovine small intestinal enteroids and in their human counterparts that lack α2,6-linked SA or αGal HBGA, suggesting that additional or alternative receptors such as integrins, hsp70, and tight-junction proteins bound directly to the VP5* domain can be used by the P[5]-bearing strains to initiate the infection of human cells. In addition, these data also suggested that P[5]-bearing strains have potential for cross-species transmission. IMPORTANCE Group A rotaviruses initiate infection through the binding of the VP8* domain of the VP4 protein to sialic acids (SAs) or histo-blood group antigens (HBGAs). Although the bovine G6P[5] WC3 strain is an important animal pathogen and is used as the backbone in the bovine-human reassortant RotaTeq vaccine, the receptor(s) for their P[5] VP8* domain has remained elusive. Using a variety of approaches, we demonstrated that the WC3 and bovine-human mono-reassortant G4P[5] vaccine strains recognize both α2,6-linked SA and αGal HBGA as ligands. Neither ligand is expressed on human small intestinal epithelial cells, explaining the absence of natural human infection by P[5]-bearing strains. However, we observed that the P[5]-bearing WC3 and G4P[5] RotaTeq vaccine strains could still infect human intestinal epithelial cells. Thus, the four P[5] RotaTeq vaccine strains potentially binding to additional alternative receptors may be efficient and effective in providing protection against severe rotavirus disease in human.
Collapse
|
31
|
Mo Z, Ma X, Luo P, Mo Y, Kaplan SS, Shou Q, Zheng M, Hille DA, Arnold BA, Liao X. Immunogenicity of pentavalent rotavirus vaccine in Chinese infants. Vaccine 2019; 37:1836-1843. [PMID: 30808567 DOI: 10.1016/j.vaccine.2019.02.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 12/27/2018] [Accepted: 02/03/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND A phase III, randomized, double-blind, placebo-controlled clinical study was conducted in China to assess the efficacy, safety, and immunogenicity of the pentavalent rotavirus vaccine (RotaTeqTM, RV5) among Chinese infants. The efficacy and safety data have been previously reported. This report presents the immunogenicity data of the study. METHODS 4,040 infants aged 6-12 weeks were randomly assigned in a 1:1 ratio to receive 3 oral doses of RV5 or placebo. Trivalent oral poliovirus vaccine (tOPV) and diphtheria, tetanus, and acellular pertussis vaccine (DTaP) were administered in a staggered-use (N = 3,240) or concomitant-use (N = 800) schedule. Immunogenicity of RV5 was evaluated in 800 participants (400 participants from each staggered- and concomitant-use immunogenicity subgroup). Geometric mean titers (GMTs) and seroresponse rates (≥3-fold rise from baseline to PD3) were measured for anti-rotavirus IgA in the staggered- and concomitant-use subgroups and measured for serum neutralizing antibodies (SNAs) to human rotavirus serotypes G1, G2, G3, G4, P1A[8] in the staggered-use subgroup. Immune responses to tOPV and DTaP co-administered with RV5 were also evaluated in the concomitant-use immunogenicity subgroup. (ClinicalTrials.gov registry: NCT02062385) RESULTS: The PD3 GMT and seroresponse rate of anti-rotavirus IgA were higher in the RV5 group (82.42 units/mL, 89.4%) compared to the placebo group (0.33 units/mL, 10.1%). Rotavirus type-specific SNA responses were also higher in the RV5 group compared to the placebo group. In the concomitant-use subgroup, the seroprotection rates of anti-poliovirus type 1, 2, 3 in the participants who received RV5 were non-inferior to those who received placebo, and the antibody responses to DTaP antigens were comparable between the two vaccination groups. CONCLUSIONS RV5 was immunogenic in Chinese infants. Immune responses induced by tOPV and DTaP were not affected by the concomitant use of RV5.
Collapse
Affiliation(s)
- Zhaojun Mo
- Guangxi Center for Disease Control and Prevention, Guangxi, PR China
| | - Xiao Ma
- National Institutes for Food and Drug Control, Beijing, PR China
| | - Peng Luo
- National Institutes for Food and Drug Control, Beijing, PR China
| | - Yi Mo
- Guangxi Center for Disease Control and Prevention, Guangxi, PR China
| | | | - Qiong Shou
- MSD R&D (China) Co., Ltd., Beijing, PR China
| | | | | | | | | | - Xueyan Liao
- MSD R&D (China) Co., Ltd., Beijing, PR China.
| |
Collapse
|
32
|
Zafari E, Soleimanjahi H, Mohammadi A, Teimoori A, Mahravani H. Molecular and biological characterization of the human-bovine rotavirus-based reassortant rotavirus. Microb Pathog 2018; 121:65-69. [PMID: 29753872 DOI: 10.1016/j.micpath.2018.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/09/2018] [Accepted: 05/09/2018] [Indexed: 11/19/2022]
Abstract
Rotaviruses (RV) are the leading cause of acute infantile gastroenteritis, associated with elevated mortality in low-income countries. Morbidity and mortality, length and rates of hospitalization due to RV gastroenteritis are dropping. Improving the quality of newborns life is an ongoing challenge for health-care providers. In this study, homemade reassortant human-bovine rotavirus was developed and biological activity and molecular characterization of candidate vaccine were evaluated for the vaccine stability. Virus titration and purification of reassortant rotavirus strains were evaluated by plaque assays, electropherotyping. The genetic stability after first, third and sixth passage was by sequencing. Due to WHO recommendation, developingment of national capacity for vaccine production in appropriate quantities and at affordable prices is the cornerstone of developing global vaccination policies. Such studies are critical to producing national vaccines and modeling herd protection.
Collapse
Affiliation(s)
- Ehsan Zafari
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Ashraf Mohammadi
- Razi Vaccine and Serum Research Institute (RVSRI) Hessark Karadj, Agricultural Research, Education and Extension Organization (AREEO), Tehran, Iran
| | - Ali Teimoori
- Department of Virology, Ahwaz Joundishpor University of Medical Sciences, Ahvaz, IR Iran
| | - Homayon Mahravani
- Razi Vaccine and Serum Research Institute (RVSRI) Hessark Karadj, Agricultural Research, Education and Extension Organization (AREEO), Tehran, Iran
| |
Collapse
|
33
|
Race-related differences in antibody responses to the inactivated influenza vaccine are linked to distinct pre-vaccination gene expression profiles in blood. Oncotarget 2018; 7:62898-62911. [PMID: 27588486 PMCID: PMC5325335 DOI: 10.18632/oncotarget.11704] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/25/2016] [Indexed: 01/22/2023] Open
Abstract
We conducted a 5-year study analyzing antibody and B cell responses to the influenza A virus components of the inactivated influenza vaccine, trivalent (IIV3) or quadrivalent (IIV4) in younger (aged 35-45) and aged (≥65 years of age) Caucasian and African American individuals. Antibody titers to the two influenza A virus strains, distribution of circulating B cell subsets and the blood transcriptome were tested at baseline and after vaccination while expression of immunoregulatory markers on B cells were analyzed at baseline. African Americans mounted higher virus neutralizing and IgG antibody responses to the H1N1 component of IIV3 or 4 compared to Caucasians. African Americans had higher levels of circulating B cell subsets compared to Caucasians. Expression of two co-regulators, i.e., programmed death (PD)-1 and the B and T cell attenuator (BTLA) were differentially expressed in the two cohorts. Race-related differences were caused by samples from younger African Americans, while results obtained with samples of aged African Americans were similar to those of aged Caucasians. Gene expression profiling by Illumina arrays revealed highly significant differences in 1368 probes at baseline between Caucasians and African Americans although samples from both cohorts showed comparable changes in transcriptome following vaccination. Genes differently expressed between samples from African Americans and Caucasians regardless of age were enriched for myeloid genes, while the transcripts that differed in expression between younger African Americans and younger Caucasians were enriched for those specific for B-cells.
Collapse
|
34
|
Kozyra I, Rzeżutka A. Farmed and companion animals as reservoirs of zoonotic rotavirus strains. POSTĘPY MIKROBIOLOGII - ADVANCEMENTS OF MICROBIOLOGY 2018; 57:156-166. [DOI: 10.21307/pm-2018.57.2.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Abstract
Rotavirus (RV) infections are a major epidemiological problem in humans and farm animals. So far, a number of human and animal RV strains have been identified. Based on the antigenic properties of the VP6 capsid protein, they have been classified into eight serogroups (A-H). The most important of them are viruses from group A (RVA), which are responsible for more than 90% of cases of rotaviral diarrhoea. The segmented structure of the virus genome and the presence of animals in human neighbourhood favour genetic reassortment between RV strains originating from different hosts. This could result in an emergence of zoonotic virus strains. The increasing number of human infections caused by virus strains having genotypes which have only been identified in animals indicates the need for epidemiological surveillance of infections. Additionally, the identification of epidemic virus strains in the outbreaks of disease in humans should be conducted. The identification of RVA strains circulating in humans and animals will allow the assessment of the impact of vaccination on the selection and emergence of zoonotic RVA strains.
1. Introduction. 2. General characteristics and classification of rotaviruses. 3. Group A rotavirus infection in humans. 4. Group A rotavirus infection in animals. 5. Genetic changes and reassortment as factors leading to the formation of zoonotic rotavirus strains. 6. Impact of human immunization on changes in genotype profile of circulating rotavirus strains. 7. Conclusions
Collapse
Affiliation(s)
- Iwona Kozyra
- Zakład Wirusologii Żywności i Środowiska , Państwowy Instytut Weterynaryjny – Państwowy Instytut Badawczy , Poland , Poland
| | - Artur Rzeżutka
- Zakład Wirusologii Żywności i Środowiska , Państwowy Instytut Weterynaryjny – Państwowy Instytut Badawczy , Poland , Poland
| |
Collapse
|
35
|
Morozova OV, Sashina TA, Epifanova NV, Zverev VV, Kashnikov AU, Novikova NA. Phylogenetic comparison of the VP7, VP4, VP6, and NSP4 genes of rotaviruses isolated from children in Nizhny Novgorod, Russia, 2015-2016, with cogent genes of the Rotarix and RotaTeq vaccine strains. Virus Genes 2017; 54:225-235. [PMID: 29236215 DOI: 10.1007/s11262-017-1529-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 12/07/2017] [Indexed: 02/01/2023]
Abstract
Group A rotaviruses (RVA) are one of the leading causes of gastroenteritis in young children worldwide. The introduction of universal mass vaccination around the world has contributed to a reduction in hospitalizations and outpatient visits associated with rotavirus infection. Continued surveillance of RVA strains is needed to determine long-term effects of vaccine introduction. In the present work, we carried out the analysis of the genotypic diversity of RVA strains isolated in Nizhny Novgorod (Russia) during the 2015-2016 epidemic season. Also we conducted a comparative analysis of the amino acid sequences of T-cell epitopes of wild-type and vaccine (RotaTeq and Rotarix) strains. In total, 1461 samples were examined. RVAs were detected in 30.4% of cases. Rotaviruses with genotype G9P[8] (40.5%) dominated in the 2015-16 epidemic season. Additionally, RVAs with the following genotypes were detected: G4P[8] (25.4%), G1P[8] (13%), G2P[4] (3.2%). Rotaviruses with genotypes G3P[9], G6P[9], and G1P[9] totaled 3%. The number of partially typed and untyped RVA samples was 66 (14.9%). The findings of a RVA of G6P[9] genotype in Russia were an original observation. Our analysis of VP6 and NSP4 T-cell epitopes showed highly conserved amino acid sequences. The found differences seem not to be caused by the immune pressure but were rather related to the genotypic affiliations of the proteins. Vaccination against rotavirus infection is not included in the national vaccination schedule in Russia. Monitoring of the genotypic and antigenic diversity of contemporary RVA will allow providing a comparative analysis of wild-type strains in areas with and without vaccine campaign.
Collapse
Affiliation(s)
- O V Morozova
- I. N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, Nizhny Novgorod, Russian Federation. .,Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation.
| | - T A Sashina
- I. N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, Nizhny Novgorod, Russian Federation
| | - N V Epifanova
- I. N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, Nizhny Novgorod, Russian Federation
| | - V V Zverev
- I. N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, Nizhny Novgorod, Russian Federation
| | - A U Kashnikov
- I. N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, Nizhny Novgorod, Russian Federation
| | - N A Novikova
- I. N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology, Nizhny Novgorod, Russian Federation.,Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russian Federation
| |
Collapse
|
36
|
Liu GF, Hille D, Kaplan SS, Goveia MG. Postdose 3 G1 serum neutralizing antibody as correlate of protection for pentavalent rotavirus vaccine. Hum Vaccin Immunother 2017; 13:2357-2363. [PMID: 28836489 PMCID: PMC5647971 DOI: 10.1080/21645515.2017.1356522] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/16/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023] Open
Abstract
Although clinical trials of the pentavalent rotavirus vaccine (RotaTeq®, RV5) have demonstrated efficacy against RV gastroenteritis (RGE) in low and high-income settings, a clear correlate of protection or a measure of immune response that could predict efficacy has yet to be identified. This is the first time that immunogenicity data with both serum neutralized antibody (SNA) titers and anti-RV IgA titers from several clinical efficacy trials were pooled to provide a unique context for evaluating the correlation between immunogenicity and RGE risk or efficacy of RV5. The correlation between immunogenicity and RGE risk is evaluated with data at the individual subject level. The analyses show that higher Postdose 3 (PD3) G1 SNA titers are associated with lower odds of contracting any RGE. The correlation between immunogenicity and efficacy is assessed using aggregated population level data, which shows higher efficacy associated with higher PD3 G1 SNA geometric mean titer (GMT) ratio (between RV5 and placebo) and PD3 serum anti-RV IgA GMT ratio. Among high-income countries, efficacy plateaus over the range of PD3 G1 SNA GMT ratios and PD3 serum anti-RV IgA GMT ratios. From both individual- and population-level analyses, PD3 G1 SNA titers correlated most closely with the RGE risk or efficacy for RV5.
Collapse
|
37
|
Morozova OV, Sashina TA, Novikova NA. Detection and molecular characterization of reassortant DS-1-like G1P [8] strains of rotavirus A. Vopr Virusol 2017; 62:91-96. [PMID: 36494934 DOI: 10.18821/0507-4088-2017-62-2-91-96] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Indexed: 12/13/2022]
Abstract
Group A rotaviruses (RVA) are the main cause of viral gastroenteritis in children worldwide. In this study we provide the molecular characteristics of reassortant DS-1-like G1P[8] RVA strains detected in Russia for the first time. Previously, such reassortant strains were detected in Japan and Thailand. The G1P[8] RVAs with DS-1-like short electropherotype RNA-PAGE were isolated from children hospitalised with an acute gastroenteritis during the 2013-2014 period. The DS-1-like G1P[8] strains accounted for 2.6% of all RVA strains detected continuously throughout the season. A phylogenetic analysis was made on the basis of the established nucleotide sequences of genes VP7, VP8* (VP4), VP6 and NSP4. The Nizhny Novgorod strains belong to G1-I and G1-II alleles of VP7 gene and to P[8]-3 allele of VP4. According to their VP6 sequences, two Russian samples clustered with the reassortant strains isolated in Japan, Thailand and Australia and two other strains were phylogenetically close to the typical G2P[4] DS-1-like RVA. Nucleotide sequences of G1P[8] strains that belong to NSP4 gene form a separate cluster from G3P[8] DS-1-like rotaviruses detected in Thailand and Australia. The RVA alleles included in Rotarix and RotaTeq vaccine strains were clustered separately from the studied reassortant RVAs. On the grounds of phylogenetic analysis we assume a polyphyletic origin of reassortants between Wa- and DS-1-like strains. Mutation rates evaluated by Bayesian inference in clusters with reassortant RVA strains were 1.004Е-3 (VP7), 1.227E-3 (VP4), 3.909E-4 (VP6), and 4.014Е-4 (NSP4). Analysis of tMRCA showed relatively contemporary origin of alleles DS-1-like G1P[8] rotaviruses: VP7 - 1998 (G1-I) and 1981 (G1-II), VP4 - 1998, VP6 - 1994, NSP4 - 1979.
Collapse
Affiliation(s)
- O V Morozova
- I.N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology.,Lobachevsky State University of Nizhny Novgorod
| | - T A Sashina
- I.N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology
| | - N A Novikova
- I.N. Blokhina Nizhny Novgorod Research Institute of Epidemiology and Microbiology.,Lobachevsky State University of Nizhny Novgorod
| |
Collapse
|
38
|
Moussa A, Fredj MBH, BenHamida-Rebaï M, Fodha I, Boujaafar N, Trabelsi A. Phylogenetic analysis of partial VP7 gene of the emerging human group A rotavirus G12 strains circulating in Tunisia. J Med Microbiol 2017; 66:112-118. [DOI: 10.1099/jmm.0.000420] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Amal Moussa
- Faculty of Pharmacy, University of Monastir, 5000 Monastir, Tunisia
- LR14SP02, Epidemiology and Immunogenetics of Human Viral Infections, Laboratory of Microbiology, Sahloul University Hospital, 4054 Sousse, Tunisia
| | - Mouna Ben Hadj Fredj
- Faculty of Sciences and Techniques, University of Kairouan, 9100 Kairouan, Tunisia
- LR14SP02, Epidemiology and Immunogenetics of Human Viral Infections, Laboratory of Microbiology, Sahloul University Hospital, 4054 Sousse, Tunisia
| | - Meriam BenHamida-Rebaï
- Faculty of Pharmacy, University of Monastir, 5000 Monastir, Tunisia
- LR14SP02, Epidemiology and Immunogenetics of Human Viral Infections, Laboratory of Microbiology, Sahloul University Hospital, 4054 Sousse, Tunisia
| | - Imene Fodha
- Faculty of Pharmacy, University of Monastir, 5000 Monastir, Tunisia
- LR14SP02, Epidemiology and Immunogenetics of Human Viral Infections, Laboratory of Microbiology, Sahloul University Hospital, 4054 Sousse, Tunisia
| | - Noureddine Boujaafar
- Laboratory of Microbiology, Sahloul University Hospital, 4054 Sousse, Tunisia
- Faculty of Pharmacy, University of Monastir, 5000 Monastir, Tunisia
| | - Abdelhalim Trabelsi
- Faculty of Pharmacy, University of Monastir, 5000 Monastir, Tunisia
- LR14SP02, Epidemiology and Immunogenetics of Human Viral Infections, Laboratory of Microbiology, Sahloul University Hospital, 4054 Sousse, Tunisia
| |
Collapse
|
39
|
Decrease of Rotavirus Gastroenteritis to a Low Level Without Resurgence for Five Years After Universal RotaTeq Vaccination in Finland. Pediatr Infect Dis J 2016; 35:1304-1308. [PMID: 27455440 DOI: 10.1097/inf.0000000000001305] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Universal rotavirus (RV) vaccination with RotaTeq was introduced into National Immunization Programme (NIP) of Finland in September 2009. We have previously reported the reduction of RV gastroenteritis (GE) cases in the first 2 years after RV vaccination in NIP in Finland. METHODS In Tampere University Hospital, a 2-year survey of acute GE (AGE) in children was conducted before NIP in the years 2006 to 2008. This was followed by a similar prospective survey in years 2009 to 2011 and now extended to years 2012 to 2014. Stool samples from children examined in the hospital for AGE were analyzed by real-time polymerase chain reaction assays for RV and norovirus, and positive samples were typed by sequencing. RESULTS The proportion of RVGE of all AGE cases decreased from 52% (421 of 809 cases) in pre-NIP years to 26% (86 of 330 cases) in post-NIP years 2009 to 2011 falling to 12% (40 of 347 cases) in 2012 and 2014. The hospitalizations for RVGE were reduced by 90% and the outpatient clinic visits also by 90% in 2012 to 2014, compared with pre-NIP year; all AGE cases were reduced by 59%. Norovirus was a major causative agent of AGE in the post-NIP period, accounting for 34% of the cases in 2009 to 2011 and 29% in 2012 to 2014. CONCLUSIONS RV vaccination in NIP has led to a major reduction of RVGE cases seen in hospital with no resurgence in 5 years after NIP. A high coverage of RV vaccination will maintain RV activity at a low level but not eliminate wild-type RV circulation.
Collapse
|
40
|
Molecular Analysis of VP7 Gene of Rotavirus G1 Strains Isolated from North India. Curr Microbiol 2016; 73:781-789. [DOI: 10.1007/s00284-016-1129-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/20/2016] [Indexed: 10/21/2022]
|
41
|
Unusual rotavirus genotypes in humans and animals with acute diarrhoea in Northeast India. Epidemiol Infect 2016; 144:2780-9. [PMID: 27113208 DOI: 10.1017/s0950268816000807] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Rotavirus (RV) infection causes acute infantile diarrhoea in humans and animals and remains a major concern for vaccine development. The close proximity of humans to animals may foster cross-species infection resulting in the emergence of novel/unusual strains by genetic reassortment. In this study, we characterized 500 diarrhoeal samples for group A rotaviruses (RVA) from children (n = 290), piglets (n = 95) and calves (n = 115) in Northeast India during 2012-2013. The data showed that 142/500 (28·4%) faecal samples were positive for RVA with the highest level of infection detected in piglets (57/142, 40·1%) followed by children (51/142, 35·9%) and calves (34/142, 23·9%). Sequence-based G- and P-typing showed G1P[8] (25%) and G1P[7] (35%) were the prevailing genotypes in both humans and animals. Single cases of unusual genotypes, i.e. G9P[8], G5P[8] in humans and G1P[13], G1P[23] and G3P[7] in animals were also identified. Cluster analyses of the sequences showed regional strains were genetically closer to their homologous strains. However, human G5P[8] and porcine G1P[8] strains showed homology to heterologous hosts of their prototype strains. The subsequent global spread of unusual RV strains may result in their establishment over time, presenting challenges to future vaccine evaluation programmes. More studies on emerging genotypes are required to elucidate how RVA strains evolve post-vaccination. This study supports the need for continuous surveillance of RVA infections after detecting from diverse hosts in a common setting.
Collapse
|
42
|
Tanmoy AM, Ahmed ASMNU, Arumugam R, Hossain B, Marzan M, Saha S, Arifeen SE, Baqui AH, Black RE, Kang G, Saha SK. Rotavirus Surveillance at a WHO-Coordinated Invasive Bacterial Disease Surveillance Site in Bangladesh: A Feasibility Study to Integrate Two Surveillance Systems. PLoS One 2016; 11:e0153582. [PMID: 27096958 PMCID: PMC4838211 DOI: 10.1371/journal.pone.0153582] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/31/2016] [Indexed: 01/12/2023] Open
Abstract
The World Health Organization (WHO) currently coordinates rotavirus diarrhea and invasive bacterial disease (IBD) surveillance at 178 sentinel sites in 60 countries. However, only 78 sites participate in both surveillance systems using a common sentinel site. Here, we explored the feasibility of extending a WHO-IBD surveillance platform to generate data on the burden of rotaviral diarrhea and its epidemiological characteristics to prepare the countries to measure the impact of rotaviral vaccine. A six-month (July to December, 2012) surveillance, managed by IBD team, collected stool samples and clinical data from under-five children with acute watery diarrhea at an IBD sentinel site. Samples were tested for rotavirus antigen by ELISA and genotyped by PCR at the regional reference laboratory (RRL). Specimens were collected from 79% (n = 297) of eligible cases (n = 375); 100% of which were tested for rotavirus by ELISA and 54% (159/297) of them were positive. At RRL, all the cases were confirmed by PCR and genotyped (99%; 158/159). The typing results revealed the predominance of G12 (40%; 64/159) genotype, followed by G1 (31%; 50/159) and G9 (19%; 31/159). All in all, this exploratory surveillance collected the desired demographic and epidemiological data and achieved almost all the benchmark indicators of WHO, starting from enrollment number to quality assurance through a number of case detection, collection, and testing of specimens and genotyping of strains at RRL. The success of this WHO-IBD site in achieving these benchmark indicators of WHO can be used by WHO as a proof-of-concept for considering integration of rotavirus surveillance with WHO-IBD platforms, specifically in countries with well performing IBD site and no ongoing rotavirus surveillance.
Collapse
Affiliation(s)
- Arif Mohammad Tanmoy
- Child Health Research Foundation, Department of Microbiology, Dhaka Shishu Hospital, Dhaka, Bangladesh
| | - ASM Nawshad Uddin Ahmed
- Child Health Research Foundation, Department of Microbiology, Dhaka Shishu Hospital, Dhaka, Bangladesh
- Department of Pediatrics, Dhaka Shishu Hospital, Bangladesh Institute of Child Health, Dhaka, Bangladesh
| | - Rajesh Arumugam
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Belal Hossain
- Child Health Research Foundation, Department of Microbiology, Dhaka Shishu Hospital, Dhaka, Bangladesh
| | - Mahfuza Marzan
- Child Health Research Foundation, Department of Microbiology, Dhaka Shishu Hospital, Dhaka, Bangladesh
| | - Shampa Saha
- Child Health Research Foundation, Department of Microbiology, Dhaka Shishu Hospital, Dhaka, Bangladesh
| | - Shams El Arifeen
- Child Health Research Foundation, Department of Microbiology, Dhaka Shishu Hospital, Dhaka, Bangladesh
- Department of Child and Adolescent Health, International Centre for Diarrhoeal Disease Research, Mohakhali, Dhaka, Bangladesh
| | - Abdullah H. Baqui
- Child Health Research Foundation, Department of Microbiology, Dhaka Shishu Hospital, Dhaka, Bangladesh
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Robert E. Black
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Gagandeep Kang
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, India
| | - Samir Kumar Saha
- Child Health Research Foundation, Department of Microbiology, Dhaka Shishu Hospital, Dhaka, Bangladesh
- Department of Microbiology, Dhaka Shishu Hospital, Bangladesh Institute of Child Health, Dhaka, Bangladesh
- * E-mail:
| |
Collapse
|
43
|
Marshall S, Sahm LJ, Moore AC. The success of microneedle-mediated vaccine delivery into skin. Hum Vaccin Immunother 2016; 12:2975-2983. [PMID: 27050528 DOI: 10.1080/21645515.2016.1171440] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Microneedles (MNs) are designed to specifically target the outermost, skin barrier layer, the stratum corneum, creating transient pathways for minimally invasive transcutaneous delivery. It is reported that MNs can facilitate delivery without stimulating the pain receptors or damaging blood vessels that lie beneath, thus being perceived as painless and associated with reduced bleeding. This immunocompetence of the skin, coupled with its ease of access, makes this organ an attractive vaccination site. The purpose of this review was to collate primary scientific literature pertaining to MN-mediated in vivo vaccination programmes. A total of 62 original research articles are presented, compiling vaccination strategies in 6 different models (mouse, rat, guinea pig, rabbit, pig, macaque and human). Vaccines tested span a wide range of viral, bacterial and protozoan pathogens and includes 7 of the 13 vaccine-preventable diseases, as defined by the WHO. This review highlights the paucity of available clinical trial data. MN-delivered vaccines have demonstrated safety and immunogenicity in pre-clinical models and boast desirable attributes such as painless administration, thermostability, dose-sparing capacity and the potential for self-administration. These advantages should contribute to enhanced global vaccine access.
Collapse
Affiliation(s)
- Sarah Marshall
- a School of Pharmacy, University College Cork , Cork , Ireland
| | - Laura J Sahm
- a School of Pharmacy, University College Cork , Cork , Ireland.,b Department of Pharmacy , Mercy University Hospital , Cork , Ireland
| | - Anne C Moore
- a School of Pharmacy, University College Cork , Cork , Ireland.,c Department of Pharmacology and Therapeutics , University College Cork , Cork , Ireland
| |
Collapse
|
44
|
O'Ryan M, Vidal R, del Canto F, Salazar JC, Montero D. Vaccines for viral and bacterial pathogens causing acute gastroenteritis: Part I: Overview, vaccines for enteric viruses and Vibrio cholerae. Hum Vaccin Immunother 2015; 11:584-600. [PMID: 25715048 DOI: 10.1080/21645515.2015.1011019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Efforts to develop vaccines for prevention of acute diarrhea have been going on for more than 40 y with partial success. The myriad of pathogens, more than 20, that have been identified as a cause of acute diarrhea throughout the years pose a significant challenge for selecting and further developing the most relevant vaccine candidates. Based on pathogen distribution as identified in epidemiological studies performed mostly in low-resource countries, rotavirus, Cryptosporidium, Shigella, diarrheogenic E. coli and V. cholerae are predominant, and thus the main targets for vaccine development and implementation. Vaccination against norovirus is most relevant in middle/high-income countries and possibly in resource-deprived countries, pending a more precise characterization of disease impact. Only a few licensed vaccines are currently available, of which rotavirus vaccines have been the most outstanding in demonstrating a significant impact in a short time period. This is a comprehensive review, divided into 2 articles, of nearly 50 vaccine candidates against the most relevant viral and bacterial pathogens that cause acute gastroenteritis. In order to facilitate reading, sections for each pathogen are organized as follows: i) a discussion of the main epidemiological and pathogenic features; and ii) a discussion of vaccines based on their stage of development, moving from current licensed vaccines to vaccines in advanced stage of development (in phase IIb or III trials) to vaccines in early stages of clinical development (in phase I/II) or preclinical development in animal models. In this first article we discuss rotavirus, norovirus and Vibrio cholerae. In the following article we will discuss Shigella, Salmonella (non-typhoidal), diarrheogenic E. coli (enterotoxigenic and enterohemorragic), and Campylobacter jejuni.
Collapse
Key Words
- ALA, aminolevulenic acid
- ASC, antibody secreting cell
- Ace, accessory cholera enterotoxin
- CT, cholera toxin
- CT-A cholera toxin A subunit
- CT-B cholera toxin B subunit
- Cep, core encoded pilus
- E. coli
- ETEC
- ETEC, enterotoxigenic E. coli
- GEMS, global enteric multi-center study
- HA/P, hemaglutinin protease
- HBGA, histo-blood group antibodies
- IS, intussusception
- IgA, immunoglobulin A
- IgG, immunoglobulin G
- IgM, immunoglobulin M
- LB, lower boundary
- LLR, Lanzhou Lamb Rotavirus vaccine
- LPS, lipopolysaccharide
- MPL, monophosphoril lipid A
- MSH, mannose-sensitive hemaglutinin pilus
- REST, rotavirus efficacy and safety trial
- RITARD
- RR, relative risk, CI, confidence interval
- RecA, recombinase A
- SAES, serious adverse events
- SRSV, small round virus, ORF, open reading frame
- STEC
- STEC, shigatoxin producing E. coli
- TCP, toxin co-regulated pilus
- V. cholerae
- VA1.3, vaccine attempt 1.3
- VLP, virus like particle
- VLPs, virus like particles, VRPs, virus replicon particles
- VP, viral proteins
- WHO, World Health Organization
- Zot, zonula occludens toxin
- acute diarrhea
- campylobacter
- enteric pathogens
- gastroenteritis
- norovirus
- removable intestinal tie-adult rabbit diarrhea
- rotavirus
- salmonella
- shigella
- vaccines
Collapse
Affiliation(s)
- Miguel O'Ryan
- a Microbiology and Mycology Program; Institute of Biomedical Sciences; Universidad de Chile ; Santiago , Chile
| | | | | | | | | |
Collapse
|
45
|
Controlling Rotavirus-associated diarrhea: Could single-domain antibody fragments make the difference? Rev Argent Microbiol 2015; 47:368-79. [PMID: 26654700 DOI: 10.1016/j.ram.2015.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/15/2015] [Accepted: 09/21/2015] [Indexed: 12/30/2022] Open
Abstract
Group A Rotavirus (RVA) remains a leading cause of severe diarrhea and child mortality. The variable domain of camelid heavy chain antibodies (VHH) display potent antigen-binding capacity, have low production costs and are suitable for oral therapies. Two sets of anti-RVA VHHs have been developed: ARP1-ARP3; 2KD1-3B2. Here, we explore the potential of both sets as a prevention strategy complementary to vaccination and a treatment option against RVA-associated diarrhea in endangered populations. Both sets have been expressed in multiple production systems, showing extensive neutralizing capacity against strains of RVA in vitro. They were also tested in the neonatal mouse model with various degrees of success in preventing or treating RVA-induced diarrhea. Interestingly, mitigation of the symptoms was also achieved with freeze-dried ARP1, so that it could be applied in areas where cold chains are difficult to maintain. 3B2 was tested in a pre-clinical trial involving gnotobiotic piglets where it conferred complete protection against RVA-induced diarrhea. ARP1 was used in the first clinical trial for anti-RVA VHHs, successfully reducing stool output in infants with RVA diarrhea, with no detected side effects.
Collapse
|
46
|
Bucardo F, Nordgren J. Impact of vaccination on the molecular epidemiology and evolution of group A rotaviruses in Latin America and factors affecting vaccine efficacy. INFECTION GENETICS AND EVOLUTION 2015; 34:106-13. [PMID: 26079278 DOI: 10.1016/j.meegid.2015.06.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/10/2015] [Accepted: 06/11/2015] [Indexed: 12/15/2022]
Abstract
Despite high rotavirus (RV) vaccine coverage (∼83%) and good effectiveness (∼77%) against RV-diarrhea hospitalization, RV is still contributing to the burden of diarrhea that persists in hospital settings in several Latin American countries, where RV vaccination is being implemented. Due to the extensive genomic and antigenic diversity, among co-circulating human RV, a major concern has been that the introduction of RV vaccination could exert selection pressure leading to higher prevalence of strains not included in the vaccines and/or emergence of new strains, thus, reducing the efficacy of vaccination. Here we review the molecular epidemiology of RV in Latin America and explore issues of RV evolution and selection in light of vaccination. We further explore etiologies behind the large burden of diarrhea remaining after vaccination in some countries and discuss plausible reasons for vaccine failures.
Collapse
Affiliation(s)
- Filemón Bucardo
- Department of Microbiology, National Autonomous University of León, Nicaragua (UNAN-León), Nicaragua.
| | - Johan Nordgren
- Division of Molecular Virology, Clinical and Experimental Medicine, Medical Faculty University of Linköping, 581 85 Linköping, Sweden
| |
Collapse
|
47
|
Comparative characteristics of the VP7 and VP4 antigenic epitopes of the rotaviruses circulating in Russia (Nizhny Novgorod) and the Rotarix and RotaTeq vaccines. Arch Virol 2015; 160:1693-703. [DOI: 10.1007/s00705-015-2439-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/24/2015] [Indexed: 11/26/2022]
|
48
|
Fisher JD, Freeman K, Clarke A, Spurgeon P, Smyth M, Perkins GD, Sujan MA, Cooke MW. Patient safety in ambulance services: a scoping review. HEALTH SERVICES AND DELIVERY RESEARCH 2015. [DOI: 10.3310/hsdr03210] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BackgroundThe role of ambulance services has changed dramatically over the last few decades with the introduction of paramedics able to provide life-saving interventions, thanks to sophisticated equipment and treatments available. The number of 999 calls continues to increase, with adverse events theoretically possible with each one. Most patient safety research is based on hospital data, but little is known concerning patient safety when using ambulance services, when things can be very different. There is an urgent need to characterise the evidence base for patient safety in NHS ambulance services.ObjectiveTo identify and map available evidence relating to patient safety when using ambulance services.DesignMixed-methods design including systematic review and review of ambulance service documentation, with areas for future research prioritised using a Delphi process.Setting and participantsAmbulance services, their staff and service users in UK.Data sourcesA wide range of data sources were explored. Multiple databases, reference lists from key papers and citations, Google and the NHS Confederation website were searched, and experts contacted to ensure that new data were included in the review. The databases MEDLINE, EMBASE, Cumulative Index to Nursing and Allied Health Literature (CINAHL), Web of Science, Science Direct, Emerald, Education Resources Information Center (ERIC), Applied Social Sciences Index and Abstracts, Social Services Abstracts, Sociological Abstracts, International Bibliography of the Social Sciences (IBSS), PsycINFO, PsycARTICLES, Health Management Information Consortium (HMIC), NHS Evidence, Cochrane Database of Systematic Reviews, Database of Abstracts of Reviews of Effects (DARE), NHS Economic Evaluation Database (NHS EED),Health Technology Assessment, the FADE library, Current Awareness Service for Health (CASH), OpenDOAR (Directory of Open Access Repositories) and Open System for Information on Grey Literature in Europe (OpenSIGLE) and Zetoc (The British Library's Electronic Table of Contents) were searched from 1 January 1980 to 12 October 2011. Publicly available documents and issues identified by National Patient Safety Agency (NPSA), NHS Litigation Authority (NHSLA) and coroners’ reports were considered. Opinions and perceptions of senior managers, ambulance staff and service users were solicited.Review methodsData were extracted from annual reports using two-stage thematic analysis, data from quality accounts were collated with safety priorities tabulated and considered using thematic analysis, NPSA incident report data were collated and displayed comparatively using descriptive statistics, claims reported to NHSLA were analysed to identify number and cost of claims from mistakes and/or poor service, and summaries of coroners’ reports were assessed using thematic analysis to identify underlying safety issues. The depth of analysis is limited by the remit of a scoping exercise and availability of data.ResultsWe identified studies exploring different aspects of safety, which were of variable quality and with little evidence to support activities currently undertaken by ambulance services. Adequately powered studies are required to address issues of patient safety in this service, and it appeared that national priorities were what determined safety activities, rather than patient need. There was inconsistency of information on attitudes and approaches to patient safety, exacerbated by a lack of common terminology.ConclusionPatient safety needs to become a more prominent consideration for ambulance services, rather than operational pressures, including targets and driving the service. Development of new models of working must include adequate training and monitoring of clinical risks. Providers and commissioners need a full understanding of the safety implications of introducing new models of care, particularly to a mobile workforce often isolated from colleagues, which requires a body of supportive evidence and an inherent critical evaluation culture. It is difficult to extrapolate findings of clinical studies undertaken in secondary care to ambulance service practice and current national guidelines often rely on consensus opinion regarding applicability to the pre-hospital environment. Areas requiring further work include the safety surrounding discharging patients, patient accidents, equipment and treatment, delays in transfer/admission to hospital, and treatment and diagnosis, with a clear need for increased reliability and training for improving handover to hospital.FundingThe National Institute for Health Research Health Services and Delivery Research programme.
Collapse
Affiliation(s)
- Joanne D Fisher
- Department of Health Sciences, Warwick Medical School, Coventry, UK
| | - Karoline Freeman
- Department of Health Sciences, Warwick Medical School, Coventry, UK
| | - Aileen Clarke
- Department of Health Sciences, Warwick Medical School, Coventry, UK
| | - Peter Spurgeon
- Department of Health Sciences, Warwick Medical School, Coventry, UK
| | - Mike Smyth
- West Midlands Ambulance Service, Millennium Point, Waterfront Business Park, Brierley Hill, West Midlands, UK
| | - Gavin D Perkins
- Department of Health Sciences, Warwick Medical School, Coventry, UK
| | | | - Matthew W Cooke
- Department of Health Sciences, Warwick Medical School, Coventry, UK
| |
Collapse
|
49
|
Poles J, Alvarez Y, Hioe CE. Induction of intestinal immunity by mucosal vaccines as a means of controlling HIV infection. AIDS Res Hum Retroviruses 2014; 30:1027-40. [PMID: 25354023 DOI: 10.1089/aid.2014.0233] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
CD4(+) T cells in the mucosa of the gastrointestinal (GI) tract are preferentially targeted and depleted by HIV. As such, the induction of an effective anti-HIV immune response in the mucosa of the GI tract-through vaccination-could protect this vulnerable population of cells. Mucosal vaccination provides a promising means of inducing robust humoral and cellular responses in the GI tract. Here we review data from the literature about the effectiveness of various mucosal vaccination routes--oral (intraintestinal/tonsilar/sublingual), intranasal, and intrarectal--with regard to the induction of immune responses mediated by cytotoxic T cells and antibodies in the GI mucosa, as well as protective efficacy in challenge models. We present data from the literature indicating that mucosal routes have the potential to effectively elicit GI mucosal immunity and protect against challenge. Given their capacity for the induction of anti-HIV immune responses in the GI mucosa, we propose that mucosal routes, including the nonconventional sublingual, tonsilar, and intrarectal routes, be considered for the delivery of the next generation HIV vaccines. However, further studies are necessary to determine the ideal vectors and vaccination regimens for these routes of immunization and to validate their efficacy in controlling HIV infection.
Collapse
Affiliation(s)
- Jordan Poles
- Department of Microbiology, New York University School of Medicine, New York, New York
| | - Yelina Alvarez
- VA New York Harbor Healthcare System–Manhattan Campus and Department of Pathology, New York University School of Medicine, New York, New York
| | - Catarina E. Hioe
- VA New York Harbor Healthcare System–Manhattan Campus and Department of Pathology, New York University School of Medicine, New York, New York
| |
Collapse
|
50
|
Degiuseppe JI, Parra GI, Stupka JA. Genetic diversity of G3 rotavirus strains circulating in Argentina during 1998-2012 assessed by full genome analyses. PLoS One 2014; 9:e110341. [PMID: 25337915 PMCID: PMC4206407 DOI: 10.1371/journal.pone.0110341] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 09/19/2014] [Indexed: 12/28/2022] Open
Abstract
Seasonal shifts in the predominant strains and the periodic emergence of new strains are epidemiological features of human rotaviruses. After the sporadic detection in two samples in 1998, G3P[8] strains reemerged as the predominant rotavirus during 2008-2009 in Argentina. Notably, in 2011 6.3% (37/587) of samples presented the G3P[6] genotypes, which coincided with the recent detection of G3P[6] and G2P[6] strains in South America and Europe. Analyses of the 11 gene segments of four G3P[8] and two G3P[6] strains revealed that G3P[8] strains detected a decade apart (1998 and 2009) presented minor differences, while the G3P[6] strains presented a complete different genomic constellation albeit showing a similar VP7 gene. This study provides insights in the dynamics and evolution of one of the genotypes with the wider range of hosts and inter-species transmission potential.
Collapse
Affiliation(s)
- Juan Ignacio Degiuseppe
- Laboratorio de Gastroenteritis Virales, Instituto Nacional de Enfermedades Infecciosas, Administración Nacional de Laboratorios e Institutos de Salud (ANLIS) “Dr. Carlos G. Malbrán”, Buenos Aires, Argentina
| | - Gabriel Ignacio Parra
- Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Asunción, Asunción, Paraguay
| | - Juan Andrés Stupka
- Laboratorio de Gastroenteritis Virales, Instituto Nacional de Enfermedades Infecciosas, Administración Nacional de Laboratorios e Institutos de Salud (ANLIS) “Dr. Carlos G. Malbrán”, Buenos Aires, Argentina
| |
Collapse
|