1
|
Yan M, Su A, Meyer D, Sosa GR, Fritsch H, Pitters M, Fischer N, Herrler G, Becher P. Precursor of H-type II histo-blood group antigen and subterminal sialic acids on gangliosides are significantly implicated in cell entry and infection by a porcine P[11] rotavirus. Emerg Microbes Infect 2025; 14:2447608. [PMID: 39726161 PMCID: PMC11727068 DOI: 10.1080/22221751.2024.2447608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/21/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Rotaviruses, non-enveloped viruses with a double-stranded RNA genome, are the leading etiological pathogen of acute gastroenteritis in young children and animals. The P[11] genotype of rotaviruses exhibits a tropism for neonates. In the present study, a binding assay using synthetic oligosaccharides demonstrated that the VP8* protein of P[11] porcine rotavirus (PRV) strain 4555 binds to lacto-N-neotetraose (LNnT) with the sequence Galβ1,4-GlcNAcβ1,3-Galβ1,4-Glc, one of the core parts of histo-blood group antigen (HBGA) and milk glycans. However, infections were significantly inhibited by blocking of endogenous monosialoganglioside (GM) GM1a with cholera toxin B subunit and preincubation of the virus with exogenous GM1a, suggesting that GM1a is involved in the infection of P[11] PRV 4555. In addition to GM1a, preincubation of the virus with exogenous disialogangliosides (GD) GD1a, GD1b, and trisialoganglioside (GT) GT1b also prevented infection. In contrast, exogenous ganglioside GM3 only inhibited infections at an early time point, and exogenous asyalosphingolipids GA1 and LacCer did not show any inhibitory effect on infections. This indicates that P[11] PRV 4555 preferentially utilizes gangliosides containing subterminal sialic acids. Further experiments revealed that P[11] PRV 4555 infections were prevented by preincubation of the virus with Neu5Ac and Neu5Gc. These results confirmed that sialic acids are essential for P[11] PRV 4555 cell entry, despite the classification as NA-resistant strain. Overall, our results proved that P[11] rotavirus not only binds to the Gal-GlcNAc motif but also utilizes gangliosides containing subterminal sialic acids.
Collapse
Affiliation(s)
- Miaomiao Yan
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ang Su
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Denise Meyer
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gleyder Roman Sosa
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Henrik Fritsch
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Malte Pitters
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole Fischer
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Georg Herrler
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Paul Becher
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
2
|
Langel SN, Otero CE, Steppe JT, Williams CA, Travieso T, Chang J, Webster H, Williamson LE, Crowe JE, Greenberg HB, Wu H, Hornik CP, Mansouri K, Edwards RJ, Stalls V, Acharya P, Blasi M, Permar SR. Breast milk delivery of an engineered dimeric IgA protects neonates against rotavirus. Mucosal Immunol 2025; 18:441-452. [PMID: 39842610 PMCID: PMC11982437 DOI: 10.1016/j.mucimm.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/27/2024] [Accepted: 01/03/2025] [Indexed: 01/24/2025]
Abstract
Dimeric IgA (dIgA) is the dominant antibody in many mucosal tissues. It is actively transported onto mucosal surfaces as secretory IgA (sIgA) which plays an integral role in protection against enteric pathogens, particularly in young children. Therapeutic strategies that deliver engineered, potently neutralizing antibodies directly into the infant intestine through breast milk could provide enhanced antimicrobial protection for neonates. Here, we developed a murine model of maternal protective transfer against human rotavirus (RV) using systemic administration of a dimeric IgA monoclonal antibody (mAb). First, we showed that systemically administered dIgA passively transferred into breast milk and the stomach of suckling pups in a dose-dependent manner. Next, we optimized the recombinant production of a potently RV-neutralizing, VP4-specific dIgA (mAb41) antibody. We then demonstrated that systemic administration of dIgA and IgG mAb41 in lactating dams conferred protection from RV-induced diarrhea in suckling pups, with dIgA resulting in lower diarrhea incidence from IgG. Systemic delivery of engineered antimicrobial dIgA mAbs should be considered as an effective strategy for sIgA delivery to the infant gastrointestinal tract via breast milk to increase protection against enteric pathogens.
Collapse
MESH Headings
- Animals
- Mice
- Rotavirus Infections/immunology
- Rotavirus Infections/prevention & control
- Rotavirus/immunology
- Female
- Humans
- Animals, Newborn
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Immunoglobulin A, Secretory/immunology
- Immunoglobulin A, Secretory/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/genetics
- Milk, Human/immunology
- Immunity, Maternally-Acquired
- Disease Models, Animal
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/administration & dosage
- Immunoglobulin A/immunology
- Immunoglobulin A/genetics
- Mice, Inbred BALB C
- Immunization, Passive
- Capsid Proteins/immunology
Collapse
Affiliation(s)
- Stephanie N Langel
- Department of Pathology, Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Claire E Otero
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Justin T Steppe
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Caitlin A Williams
- Weill Cornell Medicine Department of Pediatrics, Division of Infectious Disease, New York, NY, USA
| | - Tatiana Travieso
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Jerry Chang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Helen Webster
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Lauren E Williamson
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt, TN, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Vanderbilt, TN, USA
| | - Harry B Greenberg
- Departments of Medicine and Microbiology and Immunology, Stanford University School of Medicine, Stanford CA, USA; The VA Palo Alto Health Care System, Department of Veterans Affairs, Palo Alto, CA, USA
| | - Huali Wu
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Christoph P Hornik
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Victoria Stalls
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA; Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Maria Blasi
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| | - Sallie R Permar
- Weill Cornell Medicine Department of Pediatrics, Division of Infectious Disease, New York, NY, USA.
| |
Collapse
|
3
|
Wang Z, Huang W, Yan G, Tian Y, Wang C, Mao X, Sun M, Zhou L, Yu C, Xia H. Isolation, Genomic Characterization, and Immunogenicity Evaluation of a G9P[23] Porcine Rotavirus Strain. Vet Sci 2025; 12:180. [PMID: 40005940 PMCID: PMC11861734 DOI: 10.3390/vetsci12020180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
(1) Background: Rotavirus (RV) is the primary pathogen causing diarrhea in infants and young children. The G9-type is an emerging genotype; however, its isolation is challenging, and its immunogenicity to piglets is poorly understood. This study aimed to isolate, characterize, and evaluate the immunogenicity of a PoRVA strain, RHeN2, obtained from the diarrhea samples of piglets from a pig farm in Henan Province, China. (2) Methods: Porcine rotavirus A (PoRVA) was isolated from diarrheal samples of piglets on a farm in Henan Province, China, through sequential passaging on MA-104 cells. Its immunogenicity and cross-neutralization potential were evaluated. Inactivated vaccines were prepared using two agents (formaldehyde and binary ethyleneimine, BEI) and administered intramuscularly to 28-day-old piglets. Dulbecco's Modified Eagle Medium (DMEM) served as the control. (3) Results: A PoRVA strain, G9P[23] type, was successfully isolated and named RHeN2. Full-genome Sanger sequencing revealed the genetic constellation of this strain as G9-P[23]-I5-R1-C1- M1-A8-N1-T1-E1-H1. In addition, the titers of neutralizing antibodies peaked at 45 days post-vaccination, and the immunogenicity of the BEI-inactivated group of vaccines was better than that of the formaldehyde-inactivated group. The RHeN2 (G9P[23]) strain demonstrated a strong cross-neutralization ability against the same G9P[23] and G4P[23] strains and a stronger cross-neutralizing ability against the P[23] strains than against the P[7] strains. (4) Conclusion: The newly isolated G9P[23] strain demonstrates favorable immunogenicity, holding potential as a G9-type vaccine candidate.
Collapse
Affiliation(s)
- Zixuan Wang
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Wen Huang
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Gengxuan Yan
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuan Tian
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Chune Wang
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- National Institutes for Food and Drug Control, Beijing 100050, China
| | - Xue Mao
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Meng Sun
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Lu Zhou
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Chong Yu
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Haihua Xia
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin 150010, China
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
4
|
Pang L, Issler T, Robson B, Sutton R, Lin S, Allmendinger J, Ariyadasa S, Premaratne A, Billington C, Prenner EJ. DNA-labeled chitosan nanoparticles: A potential new surrogate for assessing rotavirus attenuation and transport in sand filtration water treatment. ENVIRONMENTAL RESEARCH 2025; 264:120378. [PMID: 39551371 DOI: 10.1016/j.envres.2024.120378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Despite being a model in waterborne risk assessment, rotavirus attenuation and transport in sand filtration water treatment remains poorly understood due to a lack of representative surrogates. We investigated the suitability of DNA-labeled chitosan nanoparticles (DCNPs) to mimic rotavirus attenuation and transport in coastal and alluvial sands. Chitosan nanoparticles were synthesized and coupled with a DNA tracer. Compared to rotavirus, DCNPs had similar size (79 ± 7.2 nm vs. 72.5 nm) and buoyant density (1.65 ± 0.07 g/cm³ vs. 1.36-1.40 g/cm³) but a less negative zeta potential (-20.61 ± 1.94 mV vs. -29.77 ± 0.86 mV) and lower hydrophobicity (0% vs. 44%). Filtration experiments (flow rate 1.26-1.27 ml/min, pH 6.0, electrical conductivity 224-226 μs/cm) showed that DCNPs approximated rotavirus attenuation in coastal and alluvial sands (p ≥ 0.07). Repeated dosing of rotavirus and DCNPs caused removal efficiencies to decline in the sand media. Both entities displayed faster and less dispersive transport than a nonreactive solute tracer (NaCl) in sand media. This preliminary study suggested that DCNPs can approximately mimic rotavirus attenuation and transport in coastal and alluvial sands. However, further validation under diverse experimental conditions is necessary. This includes varying flow rates, pH levels, ionic strengths, and the presence of multivalent cations (e.g., Ca2+ and Mg2+) and organic matter. DCNPs, made from a nontoxic, biocompatible, and biodegradable natural biopolymer, hold promise as a safe tool for assessing rotavirus attenuation and transport in sand filtration water treatment and aquifer filtration processes.
Collapse
Affiliation(s)
- Liping Pang
- Institute of Environmental Science and Research, PO Box 29181, Christchurch, 8540, New Zealand.
| | - Travis Issler
- Department of Biological Sciences, University of Calgary, Calgary, T2N 1N4, Canada
| | - Beth Robson
- Institute of Environmental Science and Research, PO Box 29181, Christchurch, 8540, New Zealand
| | - Richard Sutton
- Institute of Environmental Science and Research, PO Box 29181, Christchurch, 8540, New Zealand
| | - Susan Lin
- Institute of Environmental Science and Research, PO Box 29181, Christchurch, 8540, New Zealand
| | - Jill Allmendinger
- Department of Biological Sciences, University of Calgary, Calgary, T2N 1N4, Canada
| | - Sujani Ariyadasa
- Institute of Environmental Science and Research, PO Box 29181, Christchurch, 8540, New Zealand
| | - Aruni Premaratne
- Institute of Environmental Science and Research, PO Box 29181, Christchurch, 8540, New Zealand
| | - Craig Billington
- Institute of Environmental Science and Research, PO Box 29181, Christchurch, 8540, New Zealand
| | - Elmar J Prenner
- Department of Biological Sciences, University of Calgary, Calgary, T2N 1N4, Canada
| |
Collapse
|
5
|
Gandini S, Conly J, Spencer EA, Evans D, Rosca EC, Brassey J, Maltoni S, Onakpoya I, Plüddemann A, Jefferson T, Heneghan C. Oro-faecal transmission of SARS-CoV-2: A systematic review of studies employing viral culture from gastrointestinal and other potential oro-faecal sources and evidence for transmission to humans. Epidemiol Infect 2024; 152:e138. [PMID: 39529596 PMCID: PMC11574600 DOI: 10.1017/s0950268824001481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/11/2024] [Accepted: 07/17/2024] [Indexed: 11/16/2024] Open
Abstract
The extent to which the oro-faecal route contributes to the transmission of SARS-CoV-2 is not established.We systematically reviewed the evidence on the presence of infectious SARS-CoV-2 in faeces and other gastrointestinal sources by examining studies that used viral culture to investigate the presence of replication-competent virus in these samples. We conducted searches in the WHO COVID-19 Database, LitCovid, medRxiv, and Google Scholar for SARS-CoV-2 using keywords and associated synonyms, with a search date up to 28 November 2023.We included 13 studies involving 229 COVID-19 subjects - providing 308 faecal or rectal swab SARS-CoV2 reverse transcription-polymerase chain reaction (RT-PCR)-positive samples tested with viral culture. The methods used for viral culture across the studies were heterogeneous. Three studies (two cohorts and one case series) reported observing replication-competent SARS-CoV-2 confirmed by quantitative RT-PCR (qPCR) and whole-genome sequencing, and qPCR including appropriate cycle threshold changes. Overall, six (1.9%) of 308 faecal samples subjected to cell culture showed replication-competent virus. One study found replication-competent samples from one immunocompromised patient. No studies were identified demonstrating direct evidence of oro-faecal transmission to humans.Our review found a relatively low frequency of replication-competent SARS-CoV-2 in faecal and other gastrointestinal sources. Although it is biologically plausible, more research is needed using standardized cell culture methods, control groups, adequate follow-up, and robust epidemiologic methods, including whether secondary infections occurred, to determine the role of the oro-faecal route in the transmission of SARS-CoV-2.
Collapse
Affiliation(s)
- Sara Gandini
- Department of Experimental Oncology, European Institute of Oncology IRCCS, 20141Milan, Italy
| | - John Conly
- Departments of Medicine, Microbiology, Immunology & Infectious Diseases, and Pathology & Laboratory Medicine, Synder Institute for Chronic Diseases and O’Brien Institute for Public Health, Cumming School of Medicine, University of Calgary and Alberta Health Services, Calgary, Canada
| | - Elizabeth A. Spencer
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, OX2 6GG, UK
| | - David Evans
- Department of Medical Microbiology & Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Elena C Rosca
- Department of Neurology, Victor Babes University of Medicine and Pharmacy, Piata Eftimie Murgu 2, Timisoara300041, Romania
| | | | - Susanna Maltoni
- Research and Innovation Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Igho Onakpoya
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, OX2 6GG, UK
| | - Annette Plüddemann
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, OX2 6GG, UK
| | - Tom Jefferson
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, OX2 6GG, UK
| | - Carl Heneghan
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, OX2 6GG, UK
| |
Collapse
|
6
|
Ghonaim AH, Yi G, Lei M, Xie D, Ma H, Yang Z, Usama U, Wu H, Jiang Y, Li W, He Q. Isolation, characterization and whole-genome analysis of G9 group a rotaviruses in China: Evidence for possible Porcine-Human interspecies transmission. Virology 2024; 597:110129. [PMID: 38908046 DOI: 10.1016/j.virol.2024.110129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/24/2024]
Abstract
Group A rotaviruses (RVAs) are major causes of severe gastroenteritis in infants and young animals. To enhance our understanding of the relationship between human and animals RVAs, complete genome data are necessary. We screened 92 intestinal and stool samples from diarrheic piglets by RT‒PCR targeting the VP6 gene, revealing a prevalence of 10.9%. RVA was confirmed in two out of 5 calf samples. We successfully isolated two porcine samples using MA104 cell line. The full-length genetic constellation of the two isolates were determined to be G9-P[23]-I5-R1-C1-M1-A8-N1-T7-E1-H1, with close similarity to human Wa-like and porcine strains. Sequence analysis revealed the majority of genes were closely related to porcine and human RVAs. Phylogenetic analysis revealed that these isolates might have their ancestral origin from pigs, although some of their gene segments were related to human strains. This study reveals evidence of reassortment and possible interspecies transmission between pigs and humans in China.
Collapse
Affiliation(s)
- Ahmed H Ghonaim
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China; Desert Research Centre, Cairo, Egypt
| | - GuangYuan Yi
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
| | - Mingkai Lei
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
| | - Dongqi Xie
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
| | - Hailong Ma
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China; Hubei Hongshan Laboratory, Wuhan, China
| | - Zhengxin Yang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
| | - Usama Usama
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
| | - Hao Wu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
| | - Yunbo Jiang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China
| | - Wentao Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China; Hubei Hongshan Laboratory, Wuhan, China; The Animal Disease Diagnostic Centre of Huazhong Agricultural University, Wuhan, 430070, China.
| | - Qigai He
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Centre for Sustainable Pig Production, Wuhan, China; The Animal Disease Diagnostic Centre of Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
7
|
Diebold O, Zhou S, Sharp CP, Tesla B, Chook HW, Digard P, Gaunt ER. Towards the Development of a Minigenome Assay for Species A Rotaviruses. Viruses 2024; 16:1396. [PMID: 39339871 PMCID: PMC11437487 DOI: 10.3390/v16091396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
RNA virus polymerases carry out multiple functions necessary for successful genome replication and transcription. A key tool for molecular studies of viral RNA-dependent RNA polymerases (RdRps) is a 'minigenome' or 'minireplicon' assay, in which viral RdRps are reconstituted in cells in the absence of full virus infection. Typically, plasmids expressing the viral polymerase protein(s) and other co-factors are co-transfected, along with a plasmid expressing an RNA encoding a fluorescent or luminescent reporter gene flanked by viral untranslated regions containing cis-acting elements required for viral RdRp recognition. This reconstitutes the viral transcription/replication machinery and allows the viral RdRp activity to be measured as a correlate of the reporter protein signal. Here, we report on the development of a 'first-generation' plasmid-based minigenome assay for species A rotavirus using a firefly luciferase reporter gene.
Collapse
Affiliation(s)
- Ola Diebold
- Virology Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Shu Zhou
- Virology Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Colin Peter Sharp
- Virology Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Blanka Tesla
- Virology Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Hou Wei Chook
- Virology Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Paul Digard
- Virology Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Eleanor R Gaunt
- Virology Division, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| |
Collapse
|
8
|
Uprety T, Soni S, Sreenivasan C, Hause BM, Naveed A, Ni S, Graves AJ, Morrow JK, Meade N, Mellits KH, Adam E, Kennedy MA, Wang D, Li F. Genetic and antigenic characterization of two diarrhoeicdominant rotavirus A genotypes G3P[12] and G14P[12] circulating in the global equine population. J Gen Virol 2024; 105:002016. [PMID: 39163114 PMCID: PMC11335307 DOI: 10.1099/jgv.0.002016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
Equine rotavirus species A (ERVA) G3P[12] and G14P[12] are two dominant genotypes that cause foal diarrhoea with a significant economic impact on the global equine industry. ERVA can also serve as a source of novel (equine-like) rotavirus species A (RVA) reassortants with zoonotic potential as those identified previously in 2013-2019 when equine G3-like RVA was responsible for worldwide outbreaks of severe gastroenteritis and hospitalizations in children. One hurdle to ERVA research is that the standard cell culture system optimized for human rotavirus replication is not efficient for isolating ERVA. Here, using an engineered cell line defective in antiviral innate immunity, we showed that both equine G3P[12] and G14P[12] strains can be rapidly isolated from diarrhoeic foals. The genome sequence analysis revealed that both G3P[12] and G14P[12] strains share the identical genotypic constellation except for VP7 and VP6 segments in which G3P[12] possessed VP7 of genotype G3 and VP6 of genotype I6 and G14P[12] had the combination of VP7 of genotype G14 and VP6 of genotype I2. Further characterization demonstrated that two ERVA genotypes have a limited cross-neutralization. The lack of an in vitro broad cross-protection between both genotypes supported the increased recent diarrhoea outbreaks due to equine G14P[12] in foals born to dams immunized with the inactivated monovalent equine G3P[12] vaccine. Finally, using the structural modelling approach, we provided the genetic basis of the antigenic divergence between ERVA G3P[12] and G14P[12] strains. The results of this study will provide a framework for further investigation of infection biology, pathogenesis and cross-protection of equine rotaviruses.
Collapse
Affiliation(s)
- Tirth Uprety
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Shalini Soni
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Chithra Sreenivasan
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Ben M. Hause
- Department of Veterinary and Biomedical Sciences, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, 57007, USA
| | - Ahsan Naveed
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Shuisong Ni
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH 45056, USA
| | - Amy J. Graves
- Equine Diagnostic Solutions, LLC, 1501 Bull Lea Rd, Suite 104, Lexington, Kentucky 40511, USA
| | - Jennifer K. Morrow
- Equine Diagnostic Solutions, LLC, 1501 Bull Lea Rd, Suite 104, Lexington, Kentucky 40511, USA
| | - Nathan Meade
- Division of Microbiology, Brewing, and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Kenneth H. Mellits
- Division of Microbiology, Brewing, and Biotechnology, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Emma Adam
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Michael A. Kennedy
- Department of Veterinary and Biomedical Sciences, Animal Disease Research and Diagnostic Laboratory, South Dakota State University, Brookings, South Dakota, 57007, USA
| | - Dan Wang
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| | - Feng Li
- Department of Veterinary Science, Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, Kentucky 40546, USA
| |
Collapse
|
9
|
Nichols SL, Haller C, Borodavka A, Esstman SM. Rotavirus NSP2: A Master Orchestrator of Early Viral Particle Assembly. Viruses 2024; 16:814. [PMID: 38932107 PMCID: PMC11209291 DOI: 10.3390/v16060814] [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: 04/18/2024] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Rotaviruses (RVs) are 11-segmented, double-stranded (ds) RNA viruses and important causes of acute gastroenteritis in humans and other animal species. Early RV particle assembly is a multi-step process that includes the assortment, packaging and replication of the 11 genome segments in close connection with capsid morphogenesis. This process occurs inside virally induced, cytosolic, membrane-less organelles called viroplasms. While many viral and cellular proteins play roles during early RV assembly, the octameric nonstructural protein 2 (NSP2) has emerged as a master orchestrator of this key stage of the viral replication cycle. NSP2 is critical for viroplasm biogenesis as well as for the selective RNA-RNA interactions that underpin the assortment of 11 viral genome segments. Moreover, NSP2's associated enzymatic activities might serve to maintain nucleotide pools for use during viral genome replication, a process that is concurrent with early particle assembly. The goal of this review article is to summarize the available data about the structures, functions and interactions of RV NSP2 while also drawing attention to important unanswered questions in the field.
Collapse
Affiliation(s)
- Sarah L. Nichols
- Department of Biology, Wake Forest University, Wake Downtown, 455 Vine Street, Winston-Salem, NC 27106, USA;
| | - Cyril Haller
- Department of Chemical Engineering and Biotechnology, Cambridge University, Philippa Fawcett Drive, Cambridge CB3 0AS, UK;
| | - Alexander Borodavka
- Department of Chemical Engineering and Biotechnology, Cambridge University, Philippa Fawcett Drive, Cambridge CB3 0AS, UK;
| | - Sarah M. Esstman
- Department of Biology, Wake Forest University, Wake Downtown, 455 Vine Street, Winston-Salem, NC 27106, USA;
| |
Collapse
|
10
|
Raev SA, Kick MK, Chellis M, Amimo JO, Saif LJ, Vlasova AN. Histo-Blood Group Antigen-Producing Bacterial Cocktail Reduces Rotavirus A, B, and C Infection and Disease in Gnotobiotic Piglets. Viruses 2024; 16:660. [PMID: 38793542 PMCID: PMC11125826 DOI: 10.3390/v16050660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024] Open
Abstract
The suboptimal performance of rotavirus (RV) vaccines in developing countries and in animals necessitates further research on the development of novel therapeutics and control strategies. To initiate infection, RV interacts with cell-surface O-glycans, including histo-blood group antigens (HBGAs). We have previously demonstrated that certain non-pathogenic bacteria express HBGA- like substances (HBGA+) capable of binding RV particles in vitro. We hypothesized that HBGA+ bacteria can bind RV particles in the gut lumen protecting against RV species A (RVA), B (RVB), and C (RVC) infection in vivo. In this study, germ-free piglets were colonized with HBGA+ or HBGA- bacterial cocktail and infected with RVA/RVB/RVC of different genotypes. Diarrhea severity, virus shedding, immunoglobulin A (IgA) Ab titers, and cytokine levels were evaluated. Overall, colonization with HBGA+ bacteria resulted in reduced diarrhea severity and virus shedding compared to the HBGA- bacteria. Consistent with our hypothesis, the reduced severity of RV disease and infection was not associated with significant alterations in immune responses. Additionally, colonization with HBGA+ bacteria conferred beneficial effects irrespective of the piglet HBGA phenotype. These findings are the first experimental evidence that probiotic performance in vivo can be improved by including HBGA+ bacteria, providing decoy epitopes for broader/more consistent protection against diverse RVs.
Collapse
Affiliation(s)
- Sergei A. Raev
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (S.A.R.); (M.K.K.); (M.C.); (L.J.S.)
| | - Maryssa K. Kick
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (S.A.R.); (M.K.K.); (M.C.); (L.J.S.)
| | - Maria Chellis
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (S.A.R.); (M.K.K.); (M.C.); (L.J.S.)
| | | | - Linda J. Saif
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (S.A.R.); (M.K.K.); (M.C.); (L.J.S.)
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA; (S.A.R.); (M.K.K.); (M.C.); (L.J.S.)
| |
Collapse
|
11
|
Vetter J, Papa G, Tobler K, Rodriguez JM, Kley M, Myers M, Wiesendanger M, Schraner EM, Luque D, Burrone OR, Fraefel C, Eichwald C. The recruitment of TRiC chaperonin in rotavirus viroplasms correlates with virus replication. mBio 2024; 15:e0049924. [PMID: 38470055 PMCID: PMC11005421 DOI: 10.1128/mbio.00499-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Rotavirus (RV) replication takes place in the viroplasms, cytosolic inclusions that allow the synthesis of virus genome segments and their encapsidation in the core shell, followed by the addition of the second layer of the virion. The viroplasms are composed of several viral proteins, including NSP5, which serves as the main building block. Microtubules, lipid droplets, and miRNA-7 are among the host components recruited in viroplasms. We investigated the interaction between RV proteins and host components of the viroplasms by performing a pull-down assay of lysates from RV-infected cells expressing NSP5-BiolD2. Subsequent tandem mass spectrometry identified all eight subunits of the tailless complex polypeptide I ring complex (TRiC), a cellular chaperonin responsible for folding at least 10% of the cytosolic proteins. Our confirmed findings reveal that TRiC is brought into viroplasms and wraps around newly formed double-layered particles. Chemical inhibition of TRiC and silencing of its subunits drastically reduced virus progeny production. Through direct RNA sequencing, we show that TRiC is critical for RV replication by controlling dsRNA genome segment synthesis, particularly negative-sense single-stranded RNA. Importantly, cryo-electron microscopy analysis shows that TRiC inhibition results in defective virus particles lacking genome segments and polymerase complex (VP1/VP3). Moreover, TRiC associates with VP2 and NSP5 but not with VP1. Also, VP2 is shown to be essential for recruiting TRiC in viroplasms and preserving their globular morphology. This study highlights the essential role of TRiC in viroplasm formation and in facilitating virion assembly during the RV life cycle. IMPORTANCE The replication of rotavirus takes place in cytosolic inclusions termed viroplasms. In these inclusions, the distinct 11 double-stranded RNA genome segments are co-packaged to complete a genome in newly generated virus particles. In this study, we show for the first time that the tailless complex polypeptide I ring complex (TRiC), a cellular chaperonin responsible for the folding of at least 10% of the cytosolic proteins, is a component of viroplasms and is required for the synthesis of the viral negative-sense single-stranded RNA. Specifically, TRiC associates with NSP5 and VP2, the cofactor involved in RNA replication. Our study adds a new component to the current model of rotavirus replication, where TRiC is recruited to viroplasms to assist replication.
Collapse
Affiliation(s)
- Janine Vetter
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Guido Papa
- Molecular Immunology Lab, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Kurt Tobler
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Javier M. Rodriguez
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología/CSIC, Cantoblanco, Madrid, Spain
| | - Manuel Kley
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Michael Myers
- Proteomics Lab, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Mahesa Wiesendanger
- Institute of Virology, University of Zurich, Zurich, Switzerland
- Institute of Veterinary Anatomy, University of Zurich, Zurich, Switzerland
| | - Elisabeth M. Schraner
- Institute of Virology, University of Zurich, Zurich, Switzerland
- Institute of Veterinary Anatomy, University of Zurich, Zurich, Switzerland
| | - Daniel Luque
- School of Biomedical Sciences, The University of New South Wales, Sydney, New South Wales, Australia
- Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, New South Wales, Australia
| | - Oscar R. Burrone
- Molecular Immunology Lab, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Cornel Fraefel
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
12
|
Snyder AJ, Agbemabiese CA, Patton JT. Production of OSU G5P[7] Porcine Rotavirus Expressing a Fluorescent Reporter via Reverse Genetics. Viruses 2024; 16:411. [PMID: 38543776 PMCID: PMC10974435 DOI: 10.3390/v16030411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 05/23/2024] Open
Abstract
Rotaviruses are a significant cause of severe, potentially life-threatening gastroenteritis in infants and the young of many economically important animals. Although vaccines against porcine rotavirus exist, both live oral and inactivated, their effectiveness in preventing gastroenteritis is less than ideal. Thus, there is a need for the development of new generations of porcine rotavirus vaccines. The Ohio State University (OSU) rotavirus strain represents a Rotavirus A species with a G5P[7] genotype, the genotype most frequently associated with rotavirus disease in piglets. Using complete genome sequences that were determined via Nanopore sequencing, we developed a robust reverse genetics system enabling the recovery of recombinant (r)OSU rotavirus. Although rOSU grew to high titers (~107 plaque-forming units/mL), its growth kinetics were modestly decreased in comparison to the laboratory-adapted OSU virus. The reverse genetics system was used to generate the rOSU rotavirus, which served as an expression vector for a foreign protein. Specifically, by engineering a fused NSP3-2A-UnaG open reading frame into the segment 7 RNA, we produced a genetically stable rOSU virus that expressed the fluorescent UnaG protein as a functional separate product. Together, these findings raise the possibility of producing improved live oral porcine rotavirus vaccines through reverse-genetics-based modification or combination porcine rotavirus vaccines that can express neutralizing antigens for other porcine enteric diseases.
Collapse
Affiliation(s)
- Anthony J. Snyder
- Department of Biology, Indiana University, 212 S. Hawthorne Drive, Simon Hall 011, Bloomington, IN 47405, USA; (A.J.S.); (C.A.A.)
| | - Chantal A. Agbemabiese
- Department of Biology, Indiana University, 212 S. Hawthorne Drive, Simon Hall 011, Bloomington, IN 47405, USA; (A.J.S.); (C.A.A.)
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra 00233, Ghana
| | - John T. Patton
- Department of Biology, Indiana University, 212 S. Hawthorne Drive, Simon Hall 011, Bloomington, IN 47405, USA; (A.J.S.); (C.A.A.)
| |
Collapse
|
13
|
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
|
14
|
Dai J, Agbemabiese CA, Griffin AN, Patton JT. Rotavirus capping enzyme VP3 inhibits interferon expression by inducing MAVS degradation during viral replication. mBio 2023; 14:e0225523. [PMID: 37905816 PMCID: PMC10746195 DOI: 10.1128/mbio.02255-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/22/2023] [Indexed: 11/02/2023] Open
Abstract
IMPORTANCE Rotavirus is an enteric RNA virus that causes severe dehydrating gastroenteritis in infants and young children through infection of enterocytes in the small intestine. Timely clearance of the virus demands a robust innate immune response by cells associated with the small intestine, including the expression of interferon (IFN). Previous studies have shown that some rotavirus strains suppress the production of interferon, by inducing the degradation of mitochondrial antiviral signaling (MAVS) protein and interferon regulatory factor-3 (IRF3). In this study, we have used reverse genetics to generate recombinant rotaviruses expressing compromised forms of VP3 or NSP1, or both, to explore the function of these viral proteins in the degradation of MAVS and IRF3. Our results demonstrate that VP3 is responsible for MAVS depletion in rotavirus-infected cells, and through this activity, helps to suppress IFN production. Thus, VP3 functions to support the activity of rotavirus NSP1, the major interferon antagonist of the virus.
Collapse
Affiliation(s)
- Jin Dai
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| | | | - Ashley N. Griffin
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - John T. Patton
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
15
|
Casey-Moore MC, Katz E, Mijatovic-Rustempasic S, Jaimes J, Gautam R, Bowen MD. Coding-complete genome sequences of rotavirus A reference strains EDIM, Ph158, and CC425. Microbiol Resour Announc 2023; 12:e0063023. [PMID: 37823654 PMCID: PMC10652927 DOI: 10.1128/mra.00630-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023] Open
Abstract
This study reports the coding-complete genome sequences of three rotavirus A (RVA) reference strains previously adapted in tissue culture: RVA/Mouse-tc/USA/EDIM/XXXX/G16P[16] with a G16-P[16]-I7-R7-C7-M8-A7-N7-T10-E7-H9 genotype constellation, RVA/Human-tc/USA/Ph158/1998/G9P[6] with a G9-P[6]-I2-R2-C2-M2-A2-N2-T2-E2-H2 genotype constellation, and RVA/Human-tc/USA/CC425/1998/G3P[9] with a G3-P[9]-I2-R2-C2-M2-A3-N2-T1-E2-H3 genotype constellation.
Collapse
Affiliation(s)
- Mary C. Casey-Moore
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Eric Katz
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Slavica Mijatovic-Rustempasic
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jose Jaimes
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Rashi Gautam
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael D. Bowen
- Viral Gastroenteritis Branch, Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
16
|
Philip AA, Agbemabiese CA, Yi G, Patton JT. T7 expression plasmids for producing a recombinant human G1P[8] rotavirus comprising RIX4414 sequences of the RV1 (Rotarix , GSK) vaccine strain. Microbiol Resour Announc 2023; 12:e0060323. [PMID: 37819123 PMCID: PMC10652971 DOI: 10.1128/mra.00603-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/30/2023] [Indexed: 10/13/2023] Open
Abstract
The live oral rotavirus RV1 (Rotarix) vaccine is formulated from the human G1P[8] RIX4414 virus. Based on RIX4414 sequences, T7 expression plasmids were constructed that supported recovery of recombinant RIX4414-like viruses by reverse genetics. These plasmids will advance the study of the RV1 vaccine, possibly allowing improvements to its efficacy.
Collapse
Affiliation(s)
- Asha A. Philip
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Chantal A. Agbemabiese
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Guanghui Yi
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, USA
| | - John T. Patton
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, USA
| |
Collapse
|
17
|
Lin QF, Wong CXL, Eaton HE, Pang X, Shmulevitz M. Reovirus genomic diversity confers plasticity for protease utility during adaptation to intracellular uncoating. J Virol 2023; 97:e0082823. [PMID: 37747236 PMCID: PMC10617468 DOI: 10.1128/jvi.00828-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/27/2023] [Indexed: 09/26/2023] Open
Abstract
IMPORTANCE Reoviruses infect many mammals and are widely studied as a model system for enteric viruses. However, most of our reovirus knowledge comes from laboratory strains maintained on immortalized L929 cells. Herein, we asked whether naturally circulating reoviruses possess the same genetic and phenotypic characteristics as laboratory strains. Naturally circulating reoviruses obtained from sewage were extremely diverse genetically. Moreover, sewage reoviruses exhibited poor fitness on L929 cells and relied heavily on gut proteases for viral uncoating and productive infection compared to laboratory strains. We then examined how naturally circulating reoviruses might adapt to cell culture conditions. Within three passages, virus isolates from the parental sewage population were selected, displaying improved fitness and intracellular uncoating in L929 cells. Remarkably, selected progeny clones were present at 0.01% of the parental population. Altogether, using reovirus as a model, our study demonstrates how the high genetic diversity of naturally circulating viruses results in rapid adaptation to new environments.
Collapse
Affiliation(s)
- Qi Feng Lin
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Casey X. L. Wong
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Heather E. Eaton
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Xiaoli Pang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- Public Health Laboratories (ProvLab), Alberta Precision Laboratories (APL), Edmonton, Alberta, Canada
| | - Maya Shmulevitz
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
18
|
Philip AA, Hu S, Dai J, Patton JT. Recombinant rotavirus expressing the glycosylated S1 protein of SARS-CoV-2. J Gen Virol 2023; 104:001899. [PMID: 37830788 PMCID: PMC10721933 DOI: 10.1099/jgv.0.001899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Reverse genetic systems have been used to introduce heterologous sequences into the rotavirus segmented double-stranded (ds)RNA genome, enabling the generation of recombinant viruses that express foreign proteins and possibly serve as vaccine vectors. Notably, insertion of SARS-CoV-2 sequences into the segment seven (NSP3) RNA of simian SA11 rotavirus was previously shown to result in the production of recombinant viruses that efficiently expressed the N-terminal domain (NTD) and the receptor-binding domain (RBD) of the S1 region of the SARS-CoV-2 spike protein. However, efforts to generate a similar recombinant (r) SA11 virus that efficiently expressed full-length S1 were less successful. In this study, we describe modifications to the S1-coding cassette inserted in the segment seven RNA that allowed recovery of second-generation rSA11 viruses that efficiently expressed the ~120-kDa S1 protein. The ~120-kDa S1 products were shown to be glycosylated, based on treatment with endoglycosidase H, which reduced the protein to a size of ~80 kDa. Co-pulldown assays demonstrated that the ~120-kDa S1 proteins had affinity for the human ACE2 receptor. Although all the second-generation rSA11 viruses expressed glycosylated S1 with affinity for the ACE receptor, only the S1 product of one virus (rSA11/S1f) was appropriately recognized by anti-S1 antibodies, suggesting the rSA11/S1f virus expressed an authentic form of S1. Compared to the other second-generation rSA11 viruses, the design of the rSA11/S1f was unique, encoding an S1 product that did not include an N-terminal FLAG tag. Probably due to the impact of FLAG tags upstream of the S1 signal peptides, the S1 products of the other viruses (rSA11/3fS1 and rSA11/3fS1-His) may have undergone defective glycosylation, impeding antibody binding. In summary, these results indicate that recombinant rotaviruses can serve as expression vectors of foreign glycosylated proteins, raising the possibility of generating rotavirus-based vaccines that can induce protective immune responses against enteric and mucosal viruses with glycosylated capsid components, including SARS-CoV-2.
Collapse
Affiliation(s)
- Asha A. Philip
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
- Present address: CSL Seqirus, 225 Wyman Street, Waltham, MA 02452, USA
| | - Sannoong Hu
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Jin Dai
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - John T. Patton
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
19
|
Graikini D, Soro AB, Sivagnanam SP, Tiwari BK, Sánchez L. Bioactivity of Fucoidan-Rich Extracts from Fucus vesiculosus against Rotavirus and Foodborne Pathogens. Mar Drugs 2023; 21:478. [PMID: 37755091 PMCID: PMC10532486 DOI: 10.3390/md21090478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Marine algae are sources of bioactive components with defensive properties of great value against microbial infections. This study investigated the bioactivity of extracts from brown algae Fucus vesiculosus against rotavirus, the worldwide leading cause of acute gastroenteritis in infants and young children. Moreover, one of the extracts was tested against four foodborne bacteria: Campylobacter jejuni, Escherichia coli, Salmonella Typhimurium, and Listeria monocytogenes, and the non-pathogenic: E. coli K12. In vitro tests using MA104 cells revealed that both whole algae extracts and crude fucoidan precipitates neutralized rotavirus in a dose-responsive manner. The maximum neutralization activity was observed when the rotavirus was incubated with 100 μg mL-1 of the hydrochloric acid-obtained crude fucoidan (91.8%), although crude fucoidan extracted using citric acid also demonstrated high values (89.5%) at the same concentration. Furthermore, molecular weight fractionation of extracts decreased their antirotaviral activity and high molecular weight fractions exhibited higher activity compared to those of lower molecular weight. A seaweed extract with high antirotaviral activity was also found to inhibit the growth of C. jejuni, S. Typhimurium, and L. monocytogenes at a concentration of 0.2 mg mL-1. Overall, this study expands the current knowledge regarding the antimicrobial mechanisms of action of extracts from F. vesiculosus.
Collapse
Affiliation(s)
- Dimitra Graikini
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain;
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Arturo B. Soro
- Foodborne Pathogens Unit, Department of Infectious Diseases in Humans, Sciensano, 1050 Brussels, Belgium;
- Teagasc Ashtown Food Research Centre, D15 DY05 Dublin, Ireland; (S.P.S.); (B.K.T.)
| | - Saravana P. Sivagnanam
- Teagasc Ashtown Food Research Centre, D15 DY05 Dublin, Ireland; (S.P.S.); (B.K.T.)
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12P928 Cork Ireland
| | - Brijesh K. Tiwari
- Teagasc Ashtown Food Research Centre, D15 DY05 Dublin, Ireland; (S.P.S.); (B.K.T.)
| | - Lourdes Sánchez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain;
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| |
Collapse
|
20
|
Flexibility of the Rotavirus NSP2 C-Terminal Region Supports Factory Formation via Liquid-Liquid Phase Separation. J Virol 2023; 97:e0003923. [PMID: 36749077 PMCID: PMC9973012 DOI: 10.1128/jvi.00039-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Many viruses sequester the materials needed for their replication into discrete subcellular factories. For rotaviruses (RVs), these factories are called viroplasms, and they are formed in the host cell cytosol via the process of liquid-liquid phase separation (LLPS). The nonstructural protein 2 (NSP2) and its binding partner, nonstructural protein 5 (NSP5), are critical for viroplasm biogenesis. Yet it is not fully understood how NSP2 and NSP5 cooperate to form factories. The C-terminal region (CTR) of NSP2 (residues 291 to 317) is flexible, allowing it to participate in domain-swapping interactions that promote interoctamer interactions and, presumably, viroplasm formation. Molecular dynamics simulations showed that a lysine-to-glutamic acid change at position 294 (K294E) reduces NSP2 CTR flexibility in silico. To test the impact of reduced NSP2 CTR flexibility during infection, we engineered a mutant RV bearing this change (rRV-NSP2K294E). Single-cycle growth assays revealed a >1.2-log reduction in endpoint titers for rRV-NSP2K294E versus the wild-type control (rRV-WT). Using immunofluorescence assays, we found that rRV-NSP2K294E formed smaller, more numerous viroplasms than rRV-WT. Live-cell imaging experiments confirmed these results and revealed that rRV-NSP2K294E factories had delayed fusion kinetics. Moreover, NSP2K294E and several other CTR mutants formed fewer viroplasm-like structures in NSP5 coexpressing cells than did control NSP2WT. Finally, NSP2K294E exhibited defects in its capacity to induce LLPS droplet formation in vitro when incubated alongside NSP5. These results underscore the importance of NSP2 CTR flexibility in supporting the biogenesis of RV factories. IMPORTANCE Viruses often condense the materials needed for their replication into discrete intracellular factories. For rotaviruses, agents of severe gastroenteritis in children, factory formation is mediated in part by an octameric protein called NSP2. A flexible C-terminal region of NSP2 has been proposed to link several NSP2 octamers together, a feature that might be important for factory formation. Here, we created a change in NSP2 that reduced C-terminal flexibility and analyzed the impact on rotavirus factories. We found that the change caused the formation of smaller and more numerous factories that could not readily fuse together like those of the wild-type virus. The altered NSP2 protein also had a reduced capacity to form factory-like condensates in a test tube. Together, these results add to our growing understanding of how NSP2 supports rotavirus factory formation-a key step of viral replication.
Collapse
|
21
|
Strauss S, Acker J, Papa G, Desirò D, Schueder F, Borodavka A, Jungmann R. Principles of RNA recruitment to viral ribonucleoprotein condensates in a segmented dsRNA virus. eLife 2023; 12:e68670. [PMID: 36700549 PMCID: PMC9925054 DOI: 10.7554/elife.68670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 01/26/2023] [Indexed: 01/27/2023] Open
Abstract
Rotaviruses transcribe 11 distinct RNAs that must be co-packaged prior to their replication to make an infectious virion. During infection, nontranslating rotavirus transcripts accumulate in cytoplasmic protein-RNA granules known as viroplasms that support segmented genome assembly and replication via a poorly understood mechanism. Here, we analysed the RV transcriptome by combining DNA-barcoded smFISH of rotavirus-infected cells. Rotavirus RNA stoichiometry in viroplasms appears to be distinct from the cytoplasmic transcript distribution, with the largest transcript being the most enriched in viroplasms, suggesting a selective RNA enrichment mechanism. While all 11 types of transcripts accumulate in viroplasms, their stoichiometry significantly varied between individual viroplasms. Accumulation of transcripts requires the presence of 3' untranslated terminal regions and viroplasmic localisation of the viral polymerase VP1, consistent with the observed lack of polyadenylated transcripts in viroplasms. Our observations reveal similarities between viroplasms and other cytoplasmic RNP granules and identify viroplasmic proteins as drivers of viral RNA assembly during viroplasm formation.
Collapse
Affiliation(s)
| | - Julia Acker
- Department of Biochemistry, University of CambridgeCambridgeUnited Kingdom
| | - Guido Papa
- Molecular Immunology Laboratory, International Centre for Genetic Engineering and BiotechnologyTriesteItaly
| | - Daniel Desirò
- Department of Biochemistry, University of CambridgeCambridgeUnited Kingdom
| | - Florian Schueder
- Max Planck Institute of BiochemistryMunichGermany
- Department of Physics and Center for Nanoscience, Ludwig Maximilian UniversityMunichGermany
| | | | - Ralf Jungmann
- Max Planck Institute of BiochemistryMunichGermany
- Department of Physics and Center for Nanoscience, Ludwig Maximilian UniversityMunichGermany
| |
Collapse
|
22
|
Bhilegaonkar KN, Kolhe RP. Transfer of viruses implicated in human disease through food. PRESENT KNOWLEDGE IN FOOD SAFETY 2023:786-811. [DOI: 10.1016/b978-0-12-819470-6.00060-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
23
|
Dai J, Yi G, Philip AA, Patton JT. Rotavirus NSP1 Subverts the Antiviral Oligoadenylate Synthetase-RNase L Pathway by Inducing RNase L Degradation. mBio 2022; 13:e0299522. [PMID: 36413023 PMCID: PMC9765674 DOI: 10.1128/mbio.02995-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 11/23/2022] Open
Abstract
The interferon (IFN)-inducible 2',5'-oligoadenylate synthetase (OAS)-RNase L pathway plays a critical role in antiviral immunity. Group A rotaviruses, including the simian SA11 strain, inhibit this pathway through two activities: an E3-ligase related activity of NSP1 that degrades proteins necessary for IFN signaling, and a phosphodiesterase (PDE) activity of VP3 that hydrolyzes the RNase L-activator 2',5'-oligoadenylate. Unexpectedly, we found that a recombinant (r) SA11 double mutant virus deficient in both activities (rSA11-VP3H797R-NSP1ΔC17) retained the ability to prevent RNase L activation. Mass spectrometry led to the discovery that NSP1 interacts with RNase L in rSA11-infected HT29 cells. This interaction was confirmed through copulldown assay of cells transiently expressing NSP1 and RNase L. Immunoblot analysis showed that infection with wild-type rSA11 virus, rSA11-VP3H797R-NSP1ΔC17 double mutant virus, or single mutant forms of the latter virus all resulted in the depletion of endogenous RNase L. The loss of RNase L was reversed by addition of the neddylation inhibitor MLN4924, but not the proteasome inhibitor MG132. Analysis of additional mutant forms of rSA11 showed that RNase L degradation no longer occurred when either the N-terminal RING domain of NSP1 was mutated or the C-terminal 98 amino acids of NSP1 were deleted. The C-terminal RNase L degradation domain is positioned upstream and is functionally independent of the NSP1 domain necessary for inhibiting IFN expression. Our studies reveal a new role for NSP1 and its E3-ligase related activity as an antagonist of RNase L and uncover a novel virus-mediated strategy of inhibiting the OAS-RNase L pathway. IMPORTANCE For productive infection, rotavirus and other RNA viruses must suppress interferon (IFN) signaling and the expression of IFN-stimulated antiviral gene products. Particularly important is inhibiting the interferon (IFN)-inducible 2',5'-oligoadenylate synthetase (OAS)-RNase L pathway, as activated RNase L can direct the nonspecific degradation of viral and cellular RNAs, thereby blocking viral replication and triggering cell death pathways. In this study, we have discovered that the simian SA11 strain of rotavirus employs a novel strategy of inhibiting the OAS-RNase L pathway. This strategy is mediated by SA11 NSP1, a nonstructural protein that hijacks E3 cullin-RING ligases, causing the ubiquitination and degradation of host proteins essential for IFN induction. Our analysis shows that SA11 NSP1 also recognizes and causes the ubiquitination of RNase L, an activity resulting in depletion of endogenous RNase L. These data raise the possibility of using therapeutics targeting cellular E3 ligases to control rotavirus infections.
Collapse
Affiliation(s)
- Jin Dai
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - Guanghui Yi
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - Asha A. Philip
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - John T. Patton
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
24
|
Philip AA, Patton JT. Generation of Recombinant Rotaviruses Expressing Human Norovirus Capsid Proteins. J Virol 2022; 96:e0126222. [PMID: 36314817 PMCID: PMC9682992 DOI: 10.1128/jvi.01262-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022] Open
Abstract
Rotavirus, a segmented double-stranded RNA virus of the Reoviridae family, is a primary cause of acute gastroenteritis in young children. In countries where rotavirus vaccines are widely used, norovirus (NoV) has emerged as the major cause of acute gastroenteritis. Towards the goal of creating a combined rotavirus-NoV vaccine, we explored the possibility of generating recombinant rotaviruses (rRVs) expressing all or portions of the NoV GII.4 VP1 capsid protein. This was accomplished by replacing the segment 7 NSP3 open reading frame with a cassette encoding, sequentially, NSP3, a 2A stop-restart translation element, and all or portions (P, P2) of NoV VP1. In addition to successfully recovering rRVs with modified SA11 segment 7 RNAs encoding NoV capsid proteins, analogous rRVs were recovered through modification of the segment 7 RNA of the RIX4414 vaccine strain. An immunoblot assay confirmed that rRVs expressed NoV capsid proteins as independent products. Moreover, VP1 expressed by rRVs underwent dimerization and was recognized by conformational-dependent anti-VP1 antibodies. Serially passaged rRVs that expressed the NoV P and P2 were genetically stable, retaining additional sequences of up to 1.1 kbp without change. However, serially passaged rRVs containing the longer 1.6-kb VP1 sequence were less stable and gave rise to virus populations with segment 7 RNAs lacking VP1 coding sequences. Together, these studies suggest that it may be possible to develop combined rotavirus-NoV vaccines using modified segment 7 RNA to express NoV P or P2. In contrast, development of potential rotavirus-NoV vaccines expressing NoV VP1 will need additional efforts to improve genetic stability. IMPORTANCE Rotavirus (RV) and norovirus (NoV) are the two most important causes of acute viral gastroenteritis (AGE) in infants and young children. While the incidence of RV AGE has been brought under control in many countries through the introduction of universal mass vaccination with live attenuated RV vaccines, similar highly effective NoV vaccines are not available. To pursue the development of a combined RV-NoV vaccine, we examined the potential of using RV as an expression vector of all or portions of the NoV capsid protein VP1. Our results showed that by replacing the NSP3 open reading frame in RV genome segment 7 RNA with a coding cassette for NSP3, a 2A stop-restart translation element, and VP1, recombinant RVs can be generated that express NoV capsid proteins. These findings raise the possibility of developing new generations of RV-based combination vaccines that provide protection against a second enteric pathogen, such as NoV.
Collapse
Affiliation(s)
- Asha A. Philip
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - John T. Patton
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
25
|
Leblanc D, Raymond Y, Lemay MJ, Champagne CP, Brassard J. Effect of probiotic bacteria on porcine rotavirus OSU infection of porcine intestinal epithelial IPEC-J2 cells. Arch Virol 2022; 167:1999-2010. [PMID: 35794494 PMCID: PMC9402510 DOI: 10.1007/s00705-022-05510-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022]
Abstract
Rotavirus infections in nursing or post-weaning piglets are known to cause diarrhea, which can lead to commercial losses. Probiotic supplementation is used as a prophylactic or therapeutic approach to dealing with microbial infections in humans and animals. To evaluate the effect of probiotic bacteria on porcine rotavirus infections, non-transformed porcine intestinal epithelial IPEC-J2 cells were used as an in vitro model, and three different procedures were tested. When cells were exposed to seven probiotics at concentrations of 105, 106, or 107 CFU/mL for 16 h and removed before rotavirus challenge, infection reduction rates determined by flow cytometry were as follows: 15% (106) and 18% (105) for Bifidobacterium longum R0175, 15% (107) and 16% (106) for B. animalis lactis A026, and 15% (105) for Lactobacillus plantarum 299V. When cells were exposed to three selected probiotic strains for 1 h at higher concentrations, that is, 108 and 5 × 108 CFU/mL, before infection with rotavirus, no significant reduction was observed. When the probiotic bacteria were incubated with the virus before cell infection, a significant 14% decrease in the infection rate was observed for B. longum R0175. The results obtained using a cell-probiotics-virus platform combined with flow cytometry analysis suggest that probiotic bacteria can have a protective effect on IPEC-J2 cells before infection and can also prevent rotavirus infection of the cells.
Collapse
Affiliation(s)
- Danielle Leblanc
- Saint-Hyacinthe Research and Development Centre, Agriculture and Agri-Food Canada, 3600 Casavant Boulevard West, Saint-Hyacinthe, QC, J2S 8E3, Canada
| | - Yves Raymond
- Saint-Hyacinthe Research and Development Centre, Agriculture and Agri-Food Canada, 3600 Casavant Boulevard West, Saint-Hyacinthe, QC, J2S 8E3, Canada
| | - Marie-Josée Lemay
- Saint-Hyacinthe Research and Development Centre, Agriculture and Agri-Food Canada, 3600 Casavant Boulevard West, Saint-Hyacinthe, QC, J2S 8E3, Canada
| | - Claude P Champagne
- Saint-Hyacinthe Research and Development Centre, Agriculture and Agri-Food Canada, 3600 Casavant Boulevard West, Saint-Hyacinthe, QC, J2S 8E3, Canada
| | - Julie Brassard
- Saint-Hyacinthe Research and Development Centre, Agriculture and Agri-Food Canada, 3600 Casavant Boulevard West, Saint-Hyacinthe, QC, J2S 8E3, Canada.
| |
Collapse
|
26
|
Gagné MJ, Savard T, Brassard J. Interactions Between Infectious Foodborne Viruses and Bacterial Biofilms Formed on Different Food Contact Surfaces. FOOD AND ENVIRONMENTAL VIROLOGY 2022; 14:267-279. [PMID: 36030359 PMCID: PMC9458689 DOI: 10.1007/s12560-022-09534-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Bacterial biofilms contribute to contamination, spoilage, persistence, and hygiene failure in the food industry, but relatively little is known about the behavior of foodborne viruses evolving in the complex communities that make up biofilm. The aim of this study was to evaluate the association between enteric viruses and biofilms on food contact surfaces. Formed biofilms of mono- and multispecies cultures were prepared on glass, stainless steel, and polystyrene coupons and 105 pfu/ml of murine norovirus, rotavirus, and hepatitis A virus were added and incubated for 15 min, 90 min, and 24 h. The data obtained clearly demonstrate that the presence of biofilms generally influences the adhesion of enteric viruses to different surfaces. Many significant increases in attachment rates were observed, particularly with rotavirus whose rate of viral infectious particles increased 7000 times in the presence of Pseudomonas fluorescens on polystyrene after 24 h of incubation and with hepatitis A virus, which seems to have an affinity for the biofilms formed by lactic acid bacteria. Murine norovirus seems to be the least influenced by the presence of biofilms with few significant increases. However, the different factors surrounding this association are unknown and seem to vary according to the viruses, the environmental conditions, and the composition of the biofilm.
Collapse
Affiliation(s)
- Marie-Josée Gagné
- Agriculture and Agri-Food Canada, Saint-Hyacinthe Research and Development Centre, 3600 Casavant Boulevard West, Saint-Hyacinthe, QC, J2S 8E3, Canada
| | - Tony Savard
- Agriculture and Agri-Food Canada, Saint-Hyacinthe Research and Development Centre, 3600 Casavant Boulevard West, Saint-Hyacinthe, QC, J2S 8E3, Canada
| | - Julie Brassard
- Agriculture and Agri-Food Canada, Saint-Hyacinthe Research and Development Centre, 3600 Casavant Boulevard West, Saint-Hyacinthe, QC, J2S 8E3, Canada.
| |
Collapse
|
27
|
Type III and Not Type I Interferons Efficiently Prevent the Spread of Rotavirus in Human Intestinal Epithelial Cells. J Virol 2022; 96:e0070622. [PMID: 36000839 PMCID: PMC9472630 DOI: 10.1128/jvi.00706-22] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Rotavirus infects intestinal epithelial cells and is the leading cause of gastroenteritis in infants worldwide. Upon viral infection, intestinal cells produce type I and type III interferons (IFNs) to alert the tissue and promote an antiviral state. These two types of IFN bind to different receptors but induce similar pathways that stimulate the activation of interferon-stimulated genes (ISGs) to combat viral infection. In this work, we studied the spread of a fluorescent wild-type (WT) SA11 rotavirus in human colorectal cancer cells lacking specific interferon receptors and compared it to that of an NSP1 mutant rotavirus that cannot interfere with the host intrinsic innate immune response. We could show that the WT rotavirus efficiently blocks the production of type I IFNs but that type III IFNs are still produced, whereas the NSP1 mutant rotavirus allows the production of both. Interestingly, while both exogenously added type I and type III IFNs could efficiently protect cells against rotavirus infection, endogenous type III IFNs were found to be key to limit infection of human intestinal cells by rotavirus. By using a fluorescent reporter cell line to highlight the cells mounting an antiviral program, we could show that paracrine signaling driven by type III IFNs efficiently controls the spread of both WT and NSP1 mutant rotavirus. Our results strongly suggest that NSP1 efficiently blocks the type I IFN-mediated antiviral response; however, its restriction of the type III IFN-mediated one is not sufficient to prevent type III IFNs from partially inhibiting viral spread in intestinal epithelial cells. Additionally, our findings further highlight the importance of type III IFNs in controlling rotavirus infection, which could be exploited as antiviral therapeutic measures. IMPORTANCE Rotavirus is one of the most common causes of gastroenteritis worldwide. In developing countries, rotavirus infections lead to more than 200,000 deaths in infants and children. The intestinal epithelial cells lining the gastrointestinal tract combat rotavirus infection by two key antiviral compounds known as type I and III interferons. However, rotavirus has developed countermeasures to block the antiviral actions of the interferons. In this work, we evaluated the arms race between rotavirus and type I and III interferons. We determined that although rotavirus could block the induction of type I interferons, it was unable to block type III interferons. The ability of infected cells to produce and release type III interferons leads to the protection of the noninfected neighboring cells and the clearance of rotavirus infection from the epithelium. This suggests that type III interferons are key antiviral agents and could be used to help control rotavirus infections in children.
Collapse
|
28
|
Abstract
Rotavirus (RV) viroplasms are cytosolic inclusions where both virus genome replication and primary steps of virus progeny assembly take place. A stabilized microtubule cytoskeleton and lipid droplets are required for the viroplasm formation, which involves several virus proteins. The viral spike protein VP4 has not previously been shown to have a direct role in viroplasm formation. However, it is involved with virus-cell attachment, endocytic internalization, and virion morphogenesis. Moreover, VP4 interacts with actin cytoskeleton components, mainly in processes involving virus entrance and egress, and thereby may have an indirect role in viroplasm formation. In this study, we used reverse genetics to construct a recombinant RV, rRV/VP4-BAP, that contains a biotin acceptor peptide (BAP) in the K145-G150 loop of the VP4 lectin domain, permitting live monitoring. The recombinant virus was replication competent but showed a reduced fitness. We demonstrate that rRV/VP4-BAP infection, as opposed to rRV/wt infection, did not lead to a reorganized actin cytoskeleton as viroplasms formed were insensitive to drugs that depolymerize actin and inhibit myosin. Moreover, wild-type (wt) VP4, but not VP4-BAP, appeared to associate with actin filaments. Similarly, VP4 in coexpression with NSP5 and NSP2 induced a significant increase in the number of viroplasm-like structures. Interestingly, a small peptide mimicking loop K145-G150 rescued the phenotype of rRV/VP4-BAP by increasing its ability to form viroplasms and hence improve virus progeny formation. Collectively, these results provide a direct link between VP4 and the actin cytoskeleton to catalyze viroplasm assembly. IMPORTANCE The spike protein VP4 participates in diverse steps of the rotavirus (RV) life cycle, including virus-cell attachment, internalization, modulation of endocytosis, virion morphogenesis, and virus egress. Using reverse genetics, we constructed for the first time a recombinant RV, rRV/VP4-BAP, harboring a heterologous peptide in the lectin domain (loop K145-G150) of VP4. The rRV/VP4-BAP was replication competent but with reduced fitness due to a defect in the ability to reorganize the actin cytoskeleton, which affected the efficiency of viroplasm assembly. This defect was rescued by adding a permeable small-peptide mimicking the wild-type VP4 loop K145-G150. In addition to revealing a new role of VP4, our findings suggest that rRV harboring an engineered VP4 could be used as a new dual vaccination platform providing immunity against RV and additional heterologous antigens.
Collapse
|
29
|
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
|
30
|
Bruno C, Paparo L, Pisapia L, Romano A, Cortese M, Punzo E, Berni Canani R. Protective effects of the postbiotic deriving from cow's milk fermentation with L. paracasei CBA L74 against Rotavirus infection in human enterocytes. Sci Rep 2022; 12:6268. [PMID: 35428750 PMCID: PMC9012738 DOI: 10.1038/s41598-022-10083-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/11/2022] [Indexed: 01/17/2023] Open
Abstract
Rotavirus (RV) is the leading cause of acute gastroenteritis-associated mortality in early childhood. Emerging clinical evidence suggest the efficacy of the postbiotic approach based on cow's milk fermentation with the probiotic Lacticaseibacillus paracasei CBAL74 (FM-CBAL74) in preventing pediatric acute gastroenteritis, but the mechanisms of action are still poorly characterized. We evaluated the protective action of FM-CBAL74 in an in vitro model of RV infection in human enterocytes. The number of infected cells together with the relevant aspects of RV infection were assessed: epithelial barrier damage (tight-junction proteins and transepithelial electrical resistance evaluation), and inflammation (reactive oxygen species, pro-inflammatory cytokines IL-6, IL-8 and TNF-α, and mitogen-activated protein kinase pathway activation). Pre-incubation with FM-CBA L74 resulted in an inhibition of epithelial barrier damage and inflammation mediated by mitogen-activated protein kinase pathway activation induced by RV infection. Modulating several protective mechanisms, the postbiotic FM-CBAL74 exerted a preventive action against RV infection. This approach could be a disrupting nutritional strategy against one of the most common killers for the pediatric age.
Collapse
Affiliation(s)
- Cristina Bruno
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Lorella Paparo
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Laura Pisapia
- Institute of Genetics and Biophysics, CNR, Naples, Italy
| | - Alessia Romano
- ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Maddalena Cortese
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Erika Punzo
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy. .,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy. .,European Laboratory for the Investigation of Food-Induced Diseases, University of Naples Federico II, Naples, Italy. .,Task Force for Microbiome Studies, University of Naples Federico II, Naples, Italy. .,Task Force for Nutraceuticals and Functional Foods, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
31
|
Skrobarczyk JW, Martin CL, Bhatia SS, Pillai SD, Berghman LR. Electron-Beam Inactivation of Human Rotavirus (HRV) for the Production of Neutralizing Egg Yolk Antibodies. Front Immunol 2022; 13:840077. [PMID: 35359996 PMCID: PMC8964080 DOI: 10.3389/fimmu.2022.840077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/21/2022] [Indexed: 12/31/2022] Open
Abstract
Electron beam (eBeam) inactivation of pathogens is a commercially proven technology in multiple industries. While commonly used in a variety of decontamination processes, this technology can be considered relatively new to the pharmaceutical industry. Rotavirus is the leading cause of severe gastroenteritis among infants, children, and at-risk adults. Infections are more severe in developing countries where access to health care, clean food, and water is limited. Passive immunization using orally administered egg yolk antibodies (chicken IgY) is proven for prophylaxis and therapy of viral diarrhea, owing to the stability of avian IgY in the harsh gut environment. Since preservation of viral antigenicity is critical for successful antibody production, the aim of this study was to demonstrate the effective use of electron beam irradiation as a method of pathogen inactivation to produce rotavirus-specific neutralizing egg yolk antibodies. White leghorn hens were immunized with the eBeam-inactivated viruses every 2 weeks until serum antibody titers peaked. The relative antigenicity of eBeam-inactivated Wa G1P[8] human rotavirus (HRV) was compared to live virus, thermally, and chemically inactivated virus preparations. Using a sandwich ELISA (with antibodies against recombinant VP8 for capture and detection of HRV), the live virus was as expected, most immunoreactive. The eBeam-inactivated HRV’s antigenicity was better preserved when compared to thermally and chemically inactivated viruses. Additionally, both egg yolk antibodies and serum-derived IgY were effective at neutralizing HRV in vitro. Electron beam inactivation is a suitable method for the inactivation of HRV and other enteric viruses for use in both passive and active immunization strategies.
Collapse
Affiliation(s)
- Jill W. Skrobarczyk
- Department of Poultry Science, Texas A&M University, College Station, TX, United States
| | - Cameron L. Martin
- Department of Poultry Science, Texas A&M University, College Station, TX, United States
| | - Sohini S. Bhatia
- Department of Poultry Science, Texas A&M University, College Station, TX, United States
- National Center for Electron Beam Research, Texas A&M University, College Station, TX, United States
| | - Suresh D. Pillai
- National Center for Electron Beam Research, Texas A&M University, College Station, TX, United States
- Department of Food Science and Technology, Texas A&M University, College Station, TX, United States
| | - Luc R. Berghman
- Department of Poultry Science, Texas A&M University, College Station, TX, United States
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- *Correspondence: Luc R. Berghman,
| |
Collapse
|
32
|
Sander WJ, Kemp G, Hugo A, Pohl CH, O’Neill HG. Rotavirus-Mediated Prostaglandin E2 Production in MA104 Cells Promotes Virus Attachment and Internalisation, Resulting in an Increased Viral Load. Front Physiol 2022; 13:805565. [PMID: 35153833 PMCID: PMC8831913 DOI: 10.3389/fphys.2022.805565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022] Open
Abstract
Rotaviruses are one of the leading causes of severe dehydrating diarrhoea in infants and children under the age of five. Despite the introduction of vaccines, disease burden remains high in sub-Saharan Africa, with no known anti-viral treatments available. During early infection rotavirus attaches to several cellular receptors and enters the cells by either clathrin-dependent or -independent endocytosis. Prostaglandin E2, an abundant eicosanoid, is produced from arachidonic acid during rotavirus infection and inhibition of prostaglandin E2 formation have a deleterious effect on rotavirus infection. In this study, MA104 cells were supplemented with γ-linolenic acid (GLA), a precursor of arachidonic acid. Infection of supplemented cells with rotavirus SA11 led to a depletion in the relative percentages of GLA and arachidonic acid which coincided with an increased production of prostaglandin E2 as monitored by ELISA. Confocal microscopy demonstrated that prostaglandin E2 co-localises with the viroplasm-forming proteins, NSP5 and NSP2. Due to the known association of viroplasms with lipid droplets and the fact that lipid droplets are sites for prostaglandin E2 production, our results indicate a possible role for viroplasms in the production of rotavirus-induced prostaglandin E2. Replication kinetics showed that inhibitors, targeting the biosynthesis of prostaglandin E2, had negative effects on rotavirus yield, especially during the early stages of infection. Using flow cytometry and prostaglandin E2 addback experiments, we show that prostaglandin E2 enhances the attachment and internalisation of rotavirus in MA104 cells indicating a possible role for prostaglandin E2 during clathrin-mediated rotavirus entry. The production of prostaglandin E2 during rotavirus infection could serve as a possible target for anti-viral treatment.
Collapse
Affiliation(s)
- Willem J. Sander
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Gabré Kemp
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Arnold Hugo
- Department of Animal Science, University of the Free State, Bloemfontein, South Africa
| | - Carolina H. Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Hester G. O’Neill
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
- *Correspondence: Hester G. O’Neill,
| |
Collapse
|
33
|
Esona MD, Gautam R, Chhabra P, Vinjé J, Bowen MD, Burke RM. Gastrointestinal Tract Infections: Viruses. ENCYCLOPEDIA OF INFECTION AND IMMUNITY 2022:82-106. [DOI: 10.1016/b978-0-12-818731-9.00217-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
34
|
Geiger F, Acker J, Papa G, Wang X, Arter WE, Saar KL, Erkamp NA, Qi R, Bravo JPK, Strauss S, Krainer G, Burrone OR, Jungmann R, Knowles TPJ, Engelke H, Borodavka A. Liquid-liquid phase separation underpins the formation of replication factories in rotaviruses. EMBO J 2021; 40:e107711. [PMID: 34524703 PMCID: PMC8561643 DOI: 10.15252/embj.2021107711] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 12/29/2022] Open
Abstract
RNA viruses induce the formation of subcellular organelles that provide microenvironments conducive to their replication. Here we show that replication factories of rotaviruses represent protein-RNA condensates that are formed via liquid-liquid phase separation of the viroplasm-forming proteins NSP5 and rotavirus RNA chaperone NSP2. Upon mixing, these proteins readily form condensates at physiologically relevant low micromolar concentrations achieved in the cytoplasm of virus-infected cells. Early infection stage condensates could be reversibly dissolved by 1,6-hexanediol, as well as propylene glycol that released rotavirus transcripts from these condensates. During the early stages of infection, propylene glycol treatments reduced viral replication and phosphorylation of the condensate-forming protein NSP5. During late infection, these condensates exhibited altered material properties and became resistant to propylene glycol, coinciding with hyperphosphorylation of NSP5. Some aspects of the assembly of cytoplasmic rotavirus replication factories mirror the formation of other ribonucleoprotein granules. Such viral RNA-rich condensates that support replication of multi-segmented genomes represent an attractive target for developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Florian Geiger
- Department of ChemistryLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Julia Acker
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Guido Papa
- International Center for Genetic Engineering and BiotechnologyTriesteItaly
- Present address:
Medical Research Council Laboratory of Molecular Biology (MRC LMB)CambridgeUK
| | - Xinyu Wang
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | | | - Kadi L Saar
- Department of ChemistryUniversity of CambridgeCambridgeUK
| | - Nadia A Erkamp
- Department of ChemistryUniversity of CambridgeCambridgeUK
| | - Runzhang Qi
- Department of ChemistryUniversity of CambridgeCambridgeUK
| | - Jack PK Bravo
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Present address:
Department of Molecular BiosciencesUniversity of Texas at AustinAustinTXUSA
| | - Sebastian Strauss
- Department of Physics and Center for NanoscienceMax Planck Institute of BiochemistryMunichGermany
| | - Georg Krainer
- Department of ChemistryUniversity of CambridgeCambridgeUK
| | - Oscar R Burrone
- International Center for Genetic Engineering and BiotechnologyTriesteItaly
| | - Ralf Jungmann
- Department of Physics and Center for NanoscienceMax Planck Institute of BiochemistryMunichGermany
| | | | - Hanna Engelke
- Department of ChemistryLudwig‐Maximilians‐Universität MünchenMunichGermany
- Institute of Pharmaceutical SciencesKarl‐Franzens‐Universität GrazGrazAustria
| | - Alexander Borodavka
- Department of ChemistryLudwig‐Maximilians‐Universität MünchenMunichGermany
- Department of BiochemistryUniversity of CambridgeCambridgeUK
- Department of Physics and Center for NanoscienceMax Planck Institute of BiochemistryMunichGermany
| |
Collapse
|
35
|
Bravo JPK, Bartnik K, Venditti L, Acker J, Gail EH, Colyer A, Davidovich C, Lamb DC, Tuma R, Calabrese AN, Borodavka A. Structural basis of rotavirus RNA chaperone displacement and RNA annealing. Proc Natl Acad Sci U S A 2021; 118:e2100198118. [PMID: 34615715 PMCID: PMC8521686 DOI: 10.1073/pnas.2100198118] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 01/13/2023] Open
Abstract
Rotavirus genomes are distributed between 11 distinct RNA molecules, all of which must be selectively copackaged during virus assembly. This likely occurs through sequence-specific RNA interactions facilitated by the RNA chaperone NSP2. Here, we report that NSP2 autoregulates its chaperone activity through its C-terminal region (CTR) that promotes RNA-RNA interactions by limiting its helix-unwinding activity. Unexpectedly, structural proteomics data revealed that the CTR does not directly interact with RNA, while accelerating RNA release from NSP2. Cryo-electron microscopy reconstructions of an NSP2-RNA complex reveal a highly conserved acidic patch on the CTR, which is poised toward the bound RNA. Virus replication was abrogated by charge-disrupting mutations within the acidic patch but completely restored by charge-preserving mutations. Mechanistic similarities between NSP2 and the unrelated bacterial RNA chaperone Hfq suggest that accelerating RNA dissociation while promoting intermolecular RNA interactions may be a widespread strategy of RNA chaperone recycling.
Collapse
Affiliation(s)
- Jack P K Bravo
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Kira Bartnik
- Department of Chemistry, Center for NanoScience, Nanosystems Initiative Munich, Centre for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, D-81377 Munich, Germany
| | - Luca Venditti
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - Julia Acker
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom
| | - Emma H Gail
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
- Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, European Molecular Biology Laboratory (EMBL) Australia, Clayton, VIC 3800, Australia
| | - Alice Colyer
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Chen Davidovich
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia
- Australian Research Council (ARC) Centre of Excellence in Advanced Molecular Imaging, European Molecular Biology Laboratory (EMBL) Australia, Clayton, VIC 3800, Australia
| | - Don C Lamb
- Department of Chemistry, Center for NanoScience, Nanosystems Initiative Munich, Centre for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, D-81377 Munich, Germany
| | - Roman Tuma
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
- Faculty of Science, University of South Bohemia, 370 05 Ceske Budejovice, Czech Republic
| | - Antonio N Calabrese
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Alexander Borodavka
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom;
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, United Kingdom
- Department of Chemistry, Center for NanoScience, Nanosystems Initiative Munich, Centre for Integrated Protein Science Munich, Ludwig Maximilian University of Munich, D-81377 Munich, Germany
| |
Collapse
|
36
|
Detection of group A rotavirus in oyster tissues by in situ capture RT-qPCR. Food Control 2021. [DOI: 10.1016/j.foodcont.2021.108161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
37
|
Effectiveness of water and sanitizer washing solutions for removing enteric viruses from blueberries. Food Control 2021. [DOI: 10.1016/j.foodcont.2021.108043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
38
|
Gozalbo-Rovira R, Santiso-Bellón C, Buesa J, Rubio-del-Campo A, Vila-Vicent S, Muñoz C, Yebra MJ, Monedero V, Rodríguez-Díaz J. Microbiota Depletion Promotes Human Rotavirus Replication in an Adult Mouse Model. Biomedicines 2021; 9:846. [PMID: 34356911 PMCID: PMC8301474 DOI: 10.3390/biomedicines9070846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
Intestinal microbiota-virus-host interaction has emerged as a key factor in mediating enteric virus pathogenicity. With the aim of analyzing whether human gut bacteria improve the inefficient replication of human rotavirus in mice, we performed fecal microbiota transplant (FMT) with healthy infants as donors in antibiotic-treated mice. We showed that a simple antibiotic treatment, irrespective of FMT, resulted in viral shedding for 6 days after challenge with the human rotavirus G1P[8] genotype Wa strain (RVwa). Rotavirus titers in feces were also significantly higher in antibiotic-treated animals with or without FMT but they were decreased in animals subject to self-FMT, where a partial re-establishment of specific bacterial taxons was evidenced. Microbial composition analysis revealed profound changes in the intestinal microbiota of antibiotic-treated animals, whereas some bacterial groups, including members of Lactobacillus, Bilophila, Mucispirillum, and Oscillospira, recovered after self-FMT. In antibiotic-treated and FMT animals where the virus replicated more efficiently, differences were observed in gene expression of immune mediators, such as IL1β and CXCL15, as well as in the fucosyltransferase FUT2, responsible for H-type antigen synthesis in the small intestine. Collectively, our results suggest that antibiotic-induced microbiota depletion eradicates the microbial taxa that restrict human rotavirus infectivity in mice.
Collapse
Affiliation(s)
- Roberto Gozalbo-Rovira
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, 46010 Valencia, Spain; (R.G.-R.); (C.S.-B.); (J.B.); (S.V.-V.); (C.M.)
| | - Cristina Santiso-Bellón
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, 46010 Valencia, Spain; (R.G.-R.); (C.S.-B.); (J.B.); (S.V.-V.); (C.M.)
| | - Javier Buesa
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, 46010 Valencia, Spain; (R.G.-R.); (C.S.-B.); (J.B.); (S.V.-V.); (C.M.)
- Hospital Clínico Universitario de Valencia, Instituto de Investigación INCLIVA, 46010 Valencia, Spain
| | - Antonio Rubio-del-Campo
- Department of Biotechnology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, 46980 Valencia, Spain; (A.R.-d.-C.); (M.J.Y.)
| | - Susana Vila-Vicent
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, 46010 Valencia, Spain; (R.G.-R.); (C.S.-B.); (J.B.); (S.V.-V.); (C.M.)
| | - Carlos Muñoz
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, 46010 Valencia, Spain; (R.G.-R.); (C.S.-B.); (J.B.); (S.V.-V.); (C.M.)
| | - María J. Yebra
- Department of Biotechnology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, 46980 Valencia, Spain; (A.R.-d.-C.); (M.J.Y.)
| | - Vicente Monedero
- Department of Biotechnology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, 46980 Valencia, Spain; (A.R.-d.-C.); (M.J.Y.)
| | - Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, 46010 Valencia, Spain; (R.G.-R.); (C.S.-B.); (J.B.); (S.V.-V.); (C.M.)
- Hospital Clínico Universitario de Valencia, Instituto de Investigación INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
39
|
Seasonal and Age-Associated Pathogen Distribution in Newborn Calves with Diarrhea Admitted to ICU. Vet Sci 2021; 8:vetsci8070128. [PMID: 34357920 PMCID: PMC8310227 DOI: 10.3390/vetsci8070128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 11/17/2022] Open
Abstract
Calf mortality constitutes a substantial loss for agriculture economy-based countries and is also a significant herd problem in developed countries. However, the occurrence and frequency of responsible gastro-intestinal (GI) pathogens in severe newborn diarrhea is still not well known. We aimed to determine the seasonal and age-associated pathogen distribution of severe diarrhea in newborn calves admitted to the intensive care unit (ICU) of Erciyes University animal hospital over a year. Fecal samples were collected during the ICU admissions, and specimens were subjected to a diarrheal pathogen screening panel that included bovine coronavirus (BCoV), Cryptosporidium spp., ETEC K99+, and bovine rotavirus, using RT-PCR and conventional PCR methods. Further isolation experiments were performed with permissive cell cultures and bacterial enrichment methods to identify the clinical importance of infectious pathogen shedding in the ICU. Among the hospitalized calves aged less than 45 days old, the majority of calves originated from small farms (85.9%). The pathogen that most frequently occurred was Cryptosporidium spp. (61.5%) followed by rotavirus (56.4%). The frequency of animal admission to ICU and GI pathogen identification was higher during the winter season (44.9%) when compared to other seasons. Most calves included in the study were 1-6 days old (44.9%). Lastly, co-infection with rotavirus and Cryptosporidium spp. occurred more frequently than other dual or multi-infection events. This study was the first to define severe diarrhea-causing GI pathogens from ICU admitted newborn calves in Turkey.
Collapse
|
40
|
Kumar P, Shukla RS, Patel A, Pullagurla SR, Bird C, Ogun O, Kumru OS, Hamidi A, Hoeksema F, Yallop C, Bines JE, Joshi SB, Volkin DB. Formulation development of a live attenuated human rotavirus (RV3-BB) vaccine candidate for use in low- and middle-income countries. Hum Vaccin Immunother 2021; 17:2298-2310. [PMID: 33861183 PMCID: PMC8189091 DOI: 10.1080/21645515.2021.1885279] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/29/2021] [Indexed: 01/05/2023] Open
Abstract
Formulation development was performed with the live, attenuated, human neonatal rotavirus vaccine candidate (RV3-BB) with three main objectives to facilitate use in low- and middle- income countries including (1) a liquid, 2-8°C stable vaccine, (2) no necessity for pre-neutralization of gastric acid prior to oral administration of a small-volume dose, and (3) a low-cost vaccine dosage form. Implementation of a high-throughput RT-qPCR viral infectivity assay for RV3-BB, which correlated well with traditional FFA assays in terms of monitoring RV3-BB stability profiles, enabled more rapid and comprehensive formulation development studies. A wide variety of different classes and types of pharmaceutical excipients were screened for their ability to stabilize RV3-BB during exposure to elevated temperatures, freeze-thaw and agitation stresses. Sucrose (50-60% w/v), PEG-3350, and a solution pH of 7.8 were selected as promising stabilizers. Using a combination of an in vitro gastric digestion model (to mimic oral delivery conditions) and accelerated storage stability studies, several buffering agents (e.g., succinate, adipate and acetate at ~200 to 400 mM) were shown to protect RV3-BB under acidic conditions, and at the same time, minimize virus destabilization during storage. Several optimized RV3-BB candidate formulations were identified based on negligible viral infectivity losses during storage at 2-8°C and -20°C for up to 12 months, as well as by relative stability comparisons at 15°C and 25°C (up to 12 and 3 months, respectively). These RV3-BB stability results are discussed in the context of stability profiles of other rotavirus serotypes as well as future RV3-BB formulation development activities.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Ravi S. Shukla
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Ashaben Patel
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Swathi R. Pullagurla
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Christopher Bird
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Oluwadara Ogun
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Ozan S. Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Ahd Hamidi
- Batavia Biosciences B.V., Bioscience Park Leiden, Leiden, The Netherlands
| | - Femke Hoeksema
- Batavia Biosciences B.V., Bioscience Park Leiden, Leiden, The Netherlands
| | - Christopher Yallop
- Batavia Biosciences B.V., Bioscience Park Leiden, Leiden, The Netherlands
| | - Julie E. Bines
- Murdoch Children’s Research Institute, Department of Paediatrics, University of Melbourne, Parkville, Australia
- Department of Gastroenterology and Clinical Nutrition, Royal Children’s Hospital, Parkville, Australia
| | - Sangeeta B. Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
41
|
Kocher J, Castellucci TB, Wen K, Li G, Yang X, Lei S, Jiang X, Yuan L. Simvastatin Reduces Protection and Intestinal T Cell Responses Induced by a Norovirus P Particle Vaccine in Gnotobiotic Pigs. Pathogens 2021; 10:pathogens10070829. [PMID: 34357979 PMCID: PMC8308729 DOI: 10.3390/pathogens10070829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/20/2021] [Accepted: 06/28/2021] [Indexed: 11/24/2022] Open
Abstract
Noroviruses (NoVs) are a leading cause of acute gastroenteritis worldwide. P particles are a potential vaccine candidate against NoV. Simvastatin is a cholesterol-reducing drug that is known to increase NoV infectivity. In this study, we examined simvastatin’s effects on P particle-induced protective efficacy and T-cell immunogenicity using the gnotobiotic pig model of human NoV infection and diarrhea. Pigs were intranasally inoculated with three doses (100 µg/dose) of GII.4/VA387-derived P particles together with monophosphoryl lipid A and chitosan adjuvants. Simvastatin-fed pigs received 8 mg/day orally for 11 days prior to challenge. A subset of pigs was orally challenged with 10 ID50 of a NoV GII.4/2006b variant at post-inoculation day (PID) 28 and monitored for 7 days post-challenge. Intestinal and systemic T cell responses were determined pre- and postchallenge. Simvastatin abolished the P particle’s protection and significantly increased diarrhea severity after NoV infection. Simvastatin decreased proliferation of virus-specific and non-specific CD8 T cells in duodenum and virus-specific CD4 and CD8 T cells in spleen and significantly reduced numbers of intestinal mononuclear cells in vaccinated pigs. Furthermore, simvastatin significantly decreased numbers of duodenal CD4+IFN-γ+, CD8+IFN-γ+ and regulatory T cells and total duodenal activated CD4+ and CD8+ T cells in vaccinated pigs pre-challenge at PID 28. Following challenge, simvastatin prevented the IFN-γ+ T cell response in spleen of vaccinated pigs. These results indicate that simvastatin abolished P particle vaccine-induced partial protection through, at least in part, impairing T cell immunity. The findings have specific implications for the development of preventive and therapeutic strategies against NoV gastroenteritis, especially for the elderly population who takes statin-type drugs.
Collapse
Affiliation(s)
- Jacob Kocher
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (J.K.); (T.B.C.); (K.W.); (G.L.); (X.Y.); (S.L.)
| | - Tammy Bui Castellucci
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (J.K.); (T.B.C.); (K.W.); (G.L.); (X.Y.); (S.L.)
| | - Ke Wen
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (J.K.); (T.B.C.); (K.W.); (G.L.); (X.Y.); (S.L.)
| | - Guohua Li
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (J.K.); (T.B.C.); (K.W.); (G.L.); (X.Y.); (S.L.)
| | - Xingdong Yang
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (J.K.); (T.B.C.); (K.W.); (G.L.); (X.Y.); (S.L.)
| | - Shaohua Lei
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (J.K.); (T.B.C.); (K.W.); (G.L.); (X.Y.); (S.L.)
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Lijuan Yuan
- Center for Emerging, Zoonotic, and Arthropod-Borne Pathogens, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (J.K.); (T.B.C.); (K.W.); (G.L.); (X.Y.); (S.L.)
- Correspondence:
| |
Collapse
|
42
|
Nilsson EM, Sullivan OM, Anderson ML, Argobright HM, Shue TM, Fedowitz FR, LaConte LEW, Esstman SM. Reverse genetic engineering of simian rotaviruses with temperature-sensitive lesions in VP1, VP2, and VP6. Virus Res 2021; 302:198488. [PMID: 34146610 DOI: 10.1016/j.virusres.2021.198488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/24/2021] [Accepted: 06/10/2021] [Indexed: 10/21/2022]
Abstract
Rotaviruses are 11-segmented double-stranded RNA viruses and important causes of acute gastroenteritis in young children. To investigate the functions of specific viral proteins during the rotavirus lifecycle, temperature-sensitive (ts) mutants were previously created using a cultivatable simian strain (SA11) and chemical mutagenesis. These ts SA11 mutants replicate more efficiently at the permissive temperature of 31 °C than at the non-permissive temperature of 39 °C. Prototype strains SA11-tsC, SA11-tsF, and SA11-tsG were mapped to the genes encoding structural proteins VP1, VP2, and VP6, respectively, and putative ts lesions were identified using Sanger sequencing. However, additional background mutations in their genomes had hampered validation of the ts lesions and confounded their use in mechanistic studies. Here, we employed plasmid only-based reverse genetics to engineer recombinant (r) SA11 rotaviruses containing only the putative ts lesions of SA11-tsC (L138P change in VP1), SA11-tsF (A387D change in VP2) or SA11-tsG (S10T, D13H, and A121G changes in VP6). For simplicity, we refer to these newly-engineered, isogenic viruses as rSA11-tsVP1, rSA11-tsVP2, and rSA11-tsVP6. Single-cycle growth assays revealed that these mutants indeed exhibit ts phenotypes with significantly diminished titers (>1.5-logs) at 39 °C versus 31 °C. The rSA11 ts mutants proved genetically stable at the population-level following 3 sequential passages at 39 °C, but individual revertant clones were detected in plaque assays. Heat sensitivity experiments showed that pre-incubation of rSA11-tsVP1 or rSA11-tsVP2, but not rSA11-tsVP6, at 39 °C diminished replication at 31 °C. This result indicates that the ts lesions in VP1 and VP2 affect the incoming virion but those in VP6 affect a later stage of the viral lifecycle. In silico molecular dynamics simulations predicted temperature-dependent, long-range effects of the S10T, D13H, and/or A121G changes on the VP6 structure. Altogether, our results confirm the ts lesions of the original SA11-tsC, SA11-tsF, and SA11-tsG mutants, provide a new set of isogenic strains for investigating aspects of rotavirus replication, and shed light on how the ts lesions might impact VP1, VP2, or VP6 functions.
Collapse
Affiliation(s)
- Emil M Nilsson
- Department of Biology, Wake Forest University, Winston-Salem, NC, USA
| | - Owen M Sullivan
- Department of Biology, Wake Forest University, Winston-Salem, NC, USA
| | | | | | - Taylor M Shue
- Department of Biology, Wake Forest University, Winston-Salem, NC, USA
| | | | - Leslie E W LaConte
- Fralin Biomedical Research Institute, Roanoke, VA, USA; Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | | |
Collapse
|
43
|
Aggarwal S, Hassan E, Baldridge MT. Experimental Methods to Study the Pathogenesis of Human Enteric RNA Viruses. Viruses 2021; 13:975. [PMID: 34070283 PMCID: PMC8225081 DOI: 10.3390/v13060975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
Every year, millions of children are infected with viruses that target the gastrointestinal tract, causing acute gastroenteritis and diarrheal illness. Indeed, approximately 700 million episodes of diarrhea occur in children under five annually, with RNA viruses norovirus, rotavirus, and astrovirus serving as major causative pathogens. Numerous methodological advancements in recent years, including the establishment of novel cultivation systems using enteroids as well as the development of murine and other animal models of infection, have helped provide insight into many features of viral pathogenesis. However, many aspects of enteric viral infections remain elusive, demanding further study. Here, we describe the different in vitro and in vivo tools available to explore different pathophysiological attributes of human enteric RNA viruses, highlighting their advantages and limitations depending upon the question being explored. In addition, we discuss key areas and opportunities that would benefit from further methodological progress.
Collapse
Affiliation(s)
- Somya Aggarwal
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.A.); (E.H.)
| | - Ebrahim Hassan
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.A.); (E.H.)
| | - Megan T. Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; (S.A.); (E.H.)
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
44
|
Philip AA, Patton JT. Rotavirus as an Expression Platform of Domains of the SARS-CoV-2 Spike Protein. Vaccines (Basel) 2021; 9:449. [PMID: 34063562 PMCID: PMC8147602 DOI: 10.3390/vaccines9050449] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
Among vaccines administered to children are those targeting rotavirus, a segmented double-stranded RNA virus that represents a major cause of severe gastroenteritis. To explore the feasibility of establishing a combined rotavirus-SARS-CoV-2 vaccine, we generated recombinant (r)SA11 rotaviruses with modified segment 7 RNAs that contained coding cassettes for NSP3, a translational 2A stop-restart signal, and a FLAG-tagged portion of the SARS-CoV-2 spike (S) protein: S1 fragment, N-terminal domain (NTD), receptor-binding domain (RBD), extended RBD (ExRBD), or S2 core (CR) domain. Generation of rSA11 containing the S1 coding sequence required a sequence insertion of 2.2 kbp, the largest such insertion yet introduced into the rotavirus genome. Immunoblotting showed that rSA11 viruses containing the smaller NTD, RBD, ExRBD, and CR coding sequences expressed S-protein products of expected size, with ExRBD expressed at highest levels. These rSA11 viruses were genetically stable during serial passage. In contrast, the rSA11 virus containing the full-length S coding sequence (rSA11/NSP3-fS1) failed to express its expected 80 kDa fS1 product, for unexplained reasons. Moreover, rSA11/NSP3-fS1 was genetically unstable, with variants lacking the S1 insertion appearing during serial passage. Nonetheless, these results emphasize the potential usefulness of rotavirus vaccines as expression vectors of immunogenic portions of the SARS-CoV-2 S protein, including NTD, RBD, ExRBD, and CR, that have sizes smaller than the S1 fragment.
Collapse
|
45
|
Sylvestre É, Prévost M, Burnet JB, Pang X, Qiu Y, Smeets P, Medema G, Hachad M, Dorner S. Demonstrating the reduction of enteric viruses by drinking water treatment during snowmelt episodes in urban areas. WATER RESEARCH X 2021; 11:100091. [PMID: 33598650 PMCID: PMC7868990 DOI: 10.1016/j.wroa.2021.100091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/21/2020] [Accepted: 01/20/2021] [Indexed: 05/25/2023]
Abstract
This study investigates short-term fluctuations in virus concentrations in source water and their removal by full-scale drinking water treatment processes under different source water conditions. Transient peaks in raw water faecal contamination were identified using in situ online β-d-glucuronidase activity monitoring at two urban drinking water treatment plants. During these peaks, sequential grab samples were collected at the source and throughout the treatment train to evaluate concentrations of rotavirus, adenovirus, norovirus, enterovirus, JC virus, reovirus, astrovirus and sapovirus by reverse transcription and real-time quantitative PCR. Virus infectivity was assessed through viral culture by measurement of cytopathic effect and integrated cell culture qPCR. Virus concentrations increased by approximately 0.5-log during two snowmelt/rainfall episodes and approximately 1.0-log following a planned wastewater discharge upstream of the drinking water intake and during a β-d-glucuronidase activity peak in dry weather conditions. Increases in the removal of adenovirus and rotavirus by coagulation/flocculation processes were observed during peak virus concentrations in source water, suggesting that these processes do not operate under steady-state conditions but dynamic conditions in response to source water conditions. Rotavirus and enterovirus detected in raw and treated water samples were predominantly negative in viral culture. At one site, infectious adenoviruses were detected in raw water and water treated by a combination of ballasted clarification, ozonation, GAC filtration, and UV disinfection operated at a dose of 40 mJ cm-2. The proposed sampling strategy can inform the understanding of the dynamics associated with virus concentrations at drinking water treatment plants susceptible to de facto wastewater reuse.
Collapse
Affiliation(s)
- Émile Sylvestre
- NSERC Industrial Chair on Drinking Water, Department of Civil, Geological, and Mining Engineering, Polytechnique Montreal, Montreal, Quebec, H3C 3A7, Canada
- Canada Research Chair in Source Water Protection, Department of Civil, Geological, and Mining Engineering, Polytechnique Montreal, Montreal, Quebec, H3C 3A7, Canada
| | - Michèle Prévost
- NSERC Industrial Chair on Drinking Water, Department of Civil, Geological, and Mining Engineering, Polytechnique Montreal, Montreal, Quebec, H3C 3A7, Canada
| | - Jean-Baptiste Burnet
- NSERC Industrial Chair on Drinking Water, Department of Civil, Geological, and Mining Engineering, Polytechnique Montreal, Montreal, Quebec, H3C 3A7, Canada
- Canada Research Chair in Source Water Protection, Department of Civil, Geological, and Mining Engineering, Polytechnique Montreal, Montreal, Quebec, H3C 3A7, Canada
| | - Xiaoli Pang
- Department of Laboratory Medicine and Pathology, University of Alberta, 116th & 85 Ave, Edmonton, AB, T6G 2R3, Canada
- Public Health Laboratory, Alberta Precision Laboratories, 8440-112nd, Edmonton, AB, T6G 2J2, Canada
| | - Yuanyuan Qiu
- Department of Laboratory Medicine and Pathology, University of Alberta, 116th & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| | - Patrick Smeets
- KWR Water Research Institute, Groningenhaven 7, 3433 PE, Nieuwegein, the Netherlands
| | - Gertjan Medema
- KWR Water Research Institute, Groningenhaven 7, 3433 PE, Nieuwegein, the Netherlands
- Sanitary Engineering, Department of Water Management, Faculty of Civil Engineering and Geosciences, Delft University of Technology, P.O. Box 5048, 2600GA, Delft, the Netherlands
| | - Mounia Hachad
- NSERC Industrial Chair on Drinking Water, Department of Civil, Geological, and Mining Engineering, Polytechnique Montreal, Montreal, Quebec, H3C 3A7, Canada
- Canada Research Chair in Source Water Protection, Department of Civil, Geological, and Mining Engineering, Polytechnique Montreal, Montreal, Quebec, H3C 3A7, Canada
| | - Sarah Dorner
- Canada Research Chair in Source Water Protection, Department of Civil, Geological, and Mining Engineering, Polytechnique Montreal, Montreal, Quebec, H3C 3A7, Canada
| |
Collapse
|
46
|
Cárcamo-Calvo R, Muñoz C, Buesa J, Rodríguez-Díaz J, Gozalbo-Rovira R. The Rotavirus Vaccine Landscape, an Update. Pathogens 2021; 10:520. [PMID: 33925924 PMCID: PMC8145439 DOI: 10.3390/pathogens10050520] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 11/17/2022] Open
Abstract
Rotavirus is the leading cause of severe acute childhood gastroenteritis, responsible for more than 128,500 deaths per year, mainly in low-income countries. Although the mortality rate has dropped significantly since the introduction of the first vaccines around 2006, an estimated 83,158 deaths are still preventable. The two main vaccines currently deployed, Rotarix and RotaTeq, both live oral vaccines, have been shown to be less effective in developing countries. In addition, they have been associated with a slight risk of intussusception, and the need for cold chain maintenance limits the accessibility of these vaccines to certain areas, leaving 65% of children worldwide unvaccinated and therefore unprotected. Against this backdrop, here we review the main vaccines under development and the state of the art on potential alternatives.
Collapse
Affiliation(s)
- Roberto Cárcamo-Calvo
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
| | - Carlos Muñoz
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
| | - Javier Buesa
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Roberto Gozalbo-Rovira
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; (R.C.-C.); (C.M.); (J.B.)
- Instituto de Investigación INCLIVA, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| |
Collapse
|
47
|
Smith SC, Gribble J, Diller JR, Wiebe MA, Thoner TW, Denison MR, Ogden KM. Reovirus RNA recombination is sequence directed and generates internally deleted defective genome segments during passage. J Virol 2021; 95:JVI.02181-20. [PMID: 33472930 PMCID: PMC8103698 DOI: 10.1128/jvi.02181-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
For viruses with segmented genomes, genetic diversity is generated by genetic drift, reassortment, and recombination. Recombination produces RNA populations distinct from full-length gene segments and can influence viral population dynamics, persistence, and host immune responses. Viruses in the Reoviridae family, including rotavirus and mammalian orthoreovirus (reovirus), have been reported to package segments containing rearrangements or internal deletions. Rotaviruses with RNA segments containing rearrangements have been isolated from immunocompromised and immunocompetent children and in vitro following serial passage at relatively high multiplicity. Reoviruses that package small, defective RNA segments have established chronic infections in cells and in mice. However, the mechanism and extent of Reoviridae RNA recombination are undefined. Towards filling this gap in knowledge, we determined the titers and RNA segment profiles for reovirus and rotavirus following serial passage in cultured cells. The viruses exhibited occasional titer reductions characteristic of interference. Reovirus strains frequently accumulated segments that retained 5' and 3' terminal sequences and featured large internal deletions, while similarly fragmented segments were rarely detected in rotavirus populations. Using next-generation RNA-sequencing to analyze RNA molecules packaged in purified reovirus particles, we identified distinct recombination sites within individual viral genome segments. Recombination junctions were frequently but not always characterized by short direct sequence repeats upstream and downstream that spanned junction sites. Taken together, these findings suggest that reovirus accumulates defective gene segments featuring internal deletions during passage and undergoes sequence-directed recombination at distinct sites.IMPORTANCE Viruses in the Reoviridae family include important pathogens of humans and other animals and have segmented RNA genomes. Recombination in RNA virus populations can facilitate novel host exploration and increased disease severity. The extent, patterns, and mechanisms of Reoviridae recombination and the functions and effects of recombined RNA products are poorly understood. Here, we provide evidence that mammalian orthoreovirus regularly synthesizes RNA recombination products that retain terminal sequences but contain internal deletions, while rotavirus rarely synthesizes such products. Recombination occurs more frequently at specific sites in the mammalian orthoreovirus genome, and short regions of identical sequence are often detected at junction sites. These findings suggest that mammalian orthoreovirus recombination events are directed in part by RNA sequences. An improved understanding of recombined viral RNA synthesis may enhance our capacity to engineer improved vaccines and virotherapies in the future.
Collapse
Affiliation(s)
- Sydni Caet Smith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | - Jennifer Gribble
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | - Julia R Diller
- Department of Pediatrics, Vanderbilt University Medical Center
| | - Michelle A Wiebe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | - Timothy W Thoner
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | - Mark R Denison
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
- Department of Pediatrics, Vanderbilt University Medical Center
| | - Kristen M Ogden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
- Department of Pediatrics, Vanderbilt University Medical Center
| |
Collapse
|
48
|
Faizuloev E, Mintaev R, Petrusha O, Marova A, Smirnova D, Ammour Y, Meskina E, Sergeev O, Zhavoronok S, Karaulov A, Svitich O, Zverev V. New approach of genetic characterization of group A rotaviruses by the nanopore sequencing method. J Virol Methods 2021; 292:114114. [PMID: 33662411 DOI: 10.1016/j.jviromet.2021.114114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 02/17/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023]
Abstract
Nanopore sequencing of virus genomes represented by segmented RNA (e.g. rotaviruses) requires the development of specific approaches. Due to the massive use of rotavirus vaccines, the relevance of monitoring the genetic diversity of circulating strains of group A rotaviruses (RVA) increased. The WHO recommended method of multiplex type-specific PCR does not allow genotyping of all clinically significant strains of RVA and identifying inter-strain differences within the genotype. We have described a new principle of amplification of RVA gene segments using six primers for reverse transcription and one universal primer for PCR for nanopore sequencing. The amplification of RVA genome was tested on clinical samples and three phylogenetically distant laboratory RVA strains, Wa (G1P[8]), DS-1 (G2P[4]) and 568 (G3P[3]). The developed protocol of sample preparation and nanopore sequencing allowed obtaining full-length sequences for gene segments of RVA, including the diagnostically significant segments 9 (VP7), 4 (VP4) and 6 (VP6) with high accuracy and coverage. The accuracy of sequencing of the rotavirus genome exceeded 99.5 %, and the genome coverage varied for different strains from 59.0 to 99.6 % (on average 86 %). The developed approach of nanopore sequencing of RVA genome could be a prospective tool for epidemiological studies and surveillance of rotavirus infection.
Collapse
Affiliation(s)
- Evgeny Faizuloev
- I. Mechnikov Research Institute of Vaccines and Sera, Department of Virology, Moscow, Russia.
| | - Ramil Mintaev
- I. Mechnikov Research Institute of Vaccines and Sera, Department of Virology, Moscow, Russia; FSBI «Center for Strategic Planning and Management of Medical and Biological Health Risks», Laboratory of Gene Therapy, Moscow, Russia
| | - Olga Petrusha
- I. Mechnikov Research Institute of Vaccines and Sera, Department of Virology, Moscow, Russia
| | - Anna Marova
- I. Mechnikov Research Institute of Vaccines and Sera, Department of Virology, Moscow, Russia
| | - Daria Smirnova
- I. Mechnikov Research Institute of Vaccines and Sera, Department of Virology, Moscow, Russia
| | - Yulia Ammour
- I. Mechnikov Research Institute of Vaccines and Sera, Department of Virology, Moscow, Russia
| | - Elena Meskina
- M. Vladimirsky Moscow Regional Research Clinical Institute (MONIKI), Department of Children's Infections, Moscow, Russia
| | - Oleg Sergeev
- Sechenov First Moscow State Medical University, Faculty of Preventive Medicine, Moscow, Russia
| | - Sergey Zhavoronok
- Belarusian State Medical University, Department of Infectious Diseases, Minsk, Belarus
| | - Alexander Karaulov
- Sechenov First Moscow State Medical University, Department of Clinical Immunology and Allergy, Moscow, Russia
| | - Oxana Svitich
- I. Mechnikov Research Institute of Vaccines and Sera, Department of Virology, Moscow, Russia; Sechenov First Moscow State Medical University, Faculty of Preventive Medicine, Moscow, Russia
| | - Vitaly Zverev
- I. Mechnikov Research Institute of Vaccines and Sera, Department of Virology, Moscow, Russia; Sechenov First Moscow State Medical University, Faculty of Preventive Medicine, Moscow, Russia
| |
Collapse
|
49
|
Elkady G, Zhu J, Peng Q, Chen M, Liu X, Chen Y, Hu C, Chen H, Guo A. Isolation and whole protein characterization of species A and B bovine rotaviruses from Chinese calves. INFECTION GENETICS AND EVOLUTION 2021; 89:104715. [PMID: 33434703 DOI: 10.1016/j.meegid.2021.104715] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/14/2020] [Accepted: 01/06/2021] [Indexed: 10/22/2022]
Abstract
Rotaviruses (RVs) account for severe diarrhea in children and young animals globally. In the current study, the fecal samples of diarrheic calves from a beef farm in Inner Mongolia were screened for RVA by ELISA and RT-PCR, followed by culture of three positive RVA samples in the MA-104 cell line. After 10 blind passages, cytopathic effects (CPE) appeared as detachment, granulation, and clustering of the inoculated cells. The virus isolates were identified by RT-PCR (VP6 gene RVA) and ESI-LC-MS/MS for whole protein sequencing. The protein sequences demonstrated the presence of two strains from species A rotavirus and one RVB strain; RVA/Cow-tc/CHN/35333/2019/G6P[5] was mixed with one RVB strain (RVB/Cow-tc/CHN/35334/2019/G5P[3]) in two samples, and RVA/Cow-tc/CHN/10927/2019/G8P[7] was found in one sample. They are of genotype constellations (G6-P[5]-I2-R2-C2-M2-A3-N2-T6-E2-H3), (G8-P[7]-I5-R1-C1- M2-A1-N1-T1-E1-H1), and (G5-P[3]-I3-R5-C5-A5-N4-H5), respectively. Besides, phylogenetic analysis of the obtained sequences demonstrated viral evolution.
Collapse
Affiliation(s)
- Gehad Elkady
- The State Key Laboratory of Agricultural Microbiology, Wuhan 430070, China; College of Veterinary Medicine, Cooperative Innovation Centre of Substantial Pig Production, Hubei Province China, Huazhong Agricultural University, Wuhan 430070, China
| | - Jie Zhu
- The State Key Laboratory of Agricultural Microbiology, Wuhan 430070, China; College of Veterinary Medicine, Cooperative Innovation Centre of Substantial Pig Production, Hubei Province China, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingjie Peng
- Wuhan Keqian Biology Co. Ltd, Wuhan 430023, China
| | - Ming Chen
- The State Key Laboratory of Agricultural Microbiology, Wuhan 430070, China; College of Veterinary Medicine, Cooperative Innovation Centre of Substantial Pig Production, Hubei Province China, Huazhong Agricultural University, Wuhan 430070, China
| | - Xin Liu
- College of Veterinary Medicine, Cooperative Innovation Centre of Substantial Pig Production, Hubei Province China, Huazhong Agricultural University, Wuhan 430070, China
| | - Yingyu Chen
- The State Key Laboratory of Agricultural Microbiology, Wuhan 430070, China; College of Veterinary Medicine, Cooperative Innovation Centre of Substantial Pig Production, Hubei Province China, Huazhong Agricultural University, Wuhan 430070, China; National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
| | - Changmin Hu
- The State Key Laboratory of Agricultural Microbiology, Wuhan 430070, China
| | - Huanchun Chen
- The State Key Laboratory of Agricultural Microbiology, Wuhan 430070, China; College of Veterinary Medicine, Cooperative Innovation Centre of Substantial Pig Production, Hubei Province China, Huazhong Agricultural University, Wuhan 430070, China; National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China
| | - Aizhen Guo
- The State Key Laboratory of Agricultural Microbiology, Wuhan 430070, China; College of Veterinary Medicine, Cooperative Innovation Centre of Substantial Pig Production, Hubei Province China, Huazhong Agricultural University, Wuhan 430070, China; National Animal Tuberculosis Para-Reference Laboratory (Wuhan) of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
50
|
Kazimbaya KM, Chisenga CC, Simuyandi M, Phiri CM, Laban NM, Bosomprah S, Permar SR, Munsaka S, Chilengi R. In-vitro inhibitory effect of maternal breastmilk components on rotavirus vaccine replication and association with infant seroconversion to live oral rotavirus vaccine. PLoS One 2020; 15:e0240714. [PMID: 33170860 PMCID: PMC7654788 DOI: 10.1371/journal.pone.0240714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/30/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Despite contributing to a significant reduction in rotavirus associated diarrhoea in highly burdened low- and middle-income countries, live attenuated, oral rotavirus vaccines have lower immunogenicity and efficacy in these settings in comparison to more developed countries. Breastmilk has been implicated among factors contributing to this lowered oral vaccine efficacy. We conducted in-vitro experiments to investigate the inhibitory effects of maternal antibody and other non-antibody components in breastmilk on rotavirus vaccine strain (Rotarix) multiplication in MA104 cell culture system and assessed associations with in-vivo vaccine seroconversion in vaccinated infants. METHODS Breastmilk samples were collected from mothers before routine rotavirus vaccination of their infant at 6 weeks of age. For each sample, whole breastmilk, purified IgA, purified IgG and IgG and IgA depleted breastmilk samples were prepared as exposure preparations. A 96 well microtitre plate was set up for each sample including a control in which only MA104 cells were grown as well as a virus control with MA104 cells and virus only. The outcome of interest was 50% inhibition dilution of each of the exposure preparations calculated as the titer at which 50% of virus dilution was achieved. Samples from 30 women were tested and correlated to vaccine seroconversion status of the infant. HIV status was also correlated to antiviral breastmilk proteins. RESULTS The mean 50% inhibitory dilution titer when whole breastmilk was added to virus infected MA104 cells was 14.3 (95% CI: 7.1, 22.7). Incubation with purified IgG resulted in a mean 50% inhibitory dilution of 5 (95%CI -1.6, 11.6). Incubating with purified IgA resulted in a mean 50% inhibitory dilution of 6.5 (95% CI -0.7, 13.7) and IgG and IgA depleted breastmilk did not yield any inhibition with a titer of 1.06 (95%CI 0.9, 1.2). Higher milk IgA levels contributed to a failure of infants to seroconvert. HIV was also not associated with any antiviral breastmilk proteins. DISCUSSION AND CONCLUSION Whole breastmilk and breastmilk purified IgG and IgA fractions showed inhibitory activity against the rotavirus vaccine Rotarix™ whilst IgA and IgG depleted breastmilk with non-antibody breastmilk fraction failed to show any inhibition activity in-vitro. These findings suggest that IgA and IgG may have functional inhibitory properties and indicates a possible mechanism of how mothers in rotavirus endemic areas with high titres of IgA and IgG may inhibit viral multiplication in the infant gut and would potentially contribute to the failure of their infants to serocovert. There was not association of HIV with either lactoferrin, lactadherin or tenascin-C concentrations.
Collapse
Affiliation(s)
- Katayi Mwila Kazimbaya
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | | | | | | | | | - Samuel Bosomprah
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
- Department of Biostatistics, School of Public Health, University of Ghana, Legon, Accra, Ghana
| | - Sallie R. Permar
- Department of Pediatrics, Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
| | - Sody Munsaka
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
| | - Roma Chilengi
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| |
Collapse
|