1
|
Kertész G, Szkiba Z, Csordás K, Kassa C, Sinkó J, Kriván G, Kállay K. Rotavirus Hepatitis in Pediatric Hematopoietic Stem Cell Transplant Recipients. Pediatr Infect Dis J 2025:00006454-990000000-01317. [PMID: 40359237 DOI: 10.1097/inf.0000000000004858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
BACKGROUND Rotavirus infection is the most common cause of gastroenteritis in children. In our study, we investigated whether changes in hepatic biomarkers measured during rotavirus infection were significantly different between stem cell transplant recipients and the normal population, before, during and after infection. METHODS Rotavirus-infected children who had previously undergone allogeneic stem cell transplantation and rotavirus-infected otherwise healthy children were included. Data were collected retrospectively, including demographic, transplant and laboratory parameters as well as disease outcome measures. We analyzed the aspartate aminotransferase (ASAT), alanine aminotransferase (ALAT), gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), total bilirubin, conjugated bilirubin, albumin and immunoglobulin G serum levels in the TX group before, during and after the rotavirus infection. RESULTS In the TX versus ROTA group, we found a significantly higher value in the following parameters: ASAT, ALAT, total bilirubin and conjugated bilirubin. Significantly lower albumin and globulin levels were observed in the TX group than in the ROTA group. Furthermore, a significantly longer disease duration was observed in the TX group. In the TX group, significant difference was found comparing the ASAT, GGT and ALP levels before, during and after rotavirus infection. CONCLUSIONS Based on our data, more pronounced liver involvement can be observed during rotavirus infection in children undergoing hematopoietic stem cell transplantation; therefore, a possible hepatotropic behavior of the virus may be postulated in this setting.
Collapse
Affiliation(s)
- Gabriella Kertész
- From the Department of Pediatric Hematology and Stem Cell Transplantation
| | - Zsófia Szkiba
- From the Department of Pediatric Hematology and Stem Cell Transplantation
| | - Katalin Csordás
- From the Department of Pediatric Hematology and Stem Cell Transplantation
| | - Csaba Kassa
- From the Department of Pediatric Hematology and Stem Cell Transplantation
| | - János Sinkó
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Gergely Kriván
- From the Department of Pediatric Hematology and Stem Cell Transplantation
| | - Krisztián Kállay
- From the Department of Pediatric Hematology and Stem Cell Transplantation
| |
Collapse
|
2
|
Hou G, Son J, Gomez Castro MF, Kawagishi T, Ren X, Roth AN, Antia A, Zeng Q, DeVeaux AL, Feng N, Kohio HP, Baldridge MT, Dermody TS, Zhu S, Ding S. Innate immune sensing of rotavirus by intestinal epithelial cells leads to diarrhea. Cell Host Microbe 2025; 33:408-419.e8. [PMID: 40037352 PMCID: PMC11932023 DOI: 10.1016/j.chom.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/07/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
Diarrhea is the predominant symptom of acute gastroenteritis resulting from enteric infections and a leading cause of death in infants and young children. However, the role of the host response in diarrhea pathogenesis is unclear. Using rotavirus and neonatal mice as a model, we found that oral inoculation of UV-inactivated replication-defective rotavirus consistently induced watery diarrhea by robust activation of cytosolic double-stranded RNA sensing pathways and type III interferon (IFN-λ) secretion. Diarrhea was significantly diminished in mice lacking the IFN-λ receptor. Mechanistically, IFN-λ signaling downregulates the expression of Dra, a chloride and bicarbonate exchanger, which contributes to reduced water absorption. We confirmed these findings in mice inoculated with reovirus, as well as in donor-derived human intestinal organoids and human biopsy samples. Our data highlight a mechanism of rapid diarrhea induction by host innate immune sensing in the gastrointestinal tract and suggest that diarrhea induction is an active host defense strategy to eliminate the pathogen.
Collapse
Affiliation(s)
- Gaopeng Hou
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Juhee Son
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Maria Florencia Gomez Castro
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Takahiro Kawagishi
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Xingxing Ren
- Institute of Immunology and the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Alexa N Roth
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Qiru Zeng
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Anna L DeVeaux
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Ningguo Feng
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hinissan P Kohio
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Megan T Baldridge
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Terence S Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | - Shu Zhu
- Institute of Immunology and the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
3
|
Wang J, Wang L, Lu W, Farhataziz N, Gonzalez A, Xing J, Zhang Z. TRIM29 controls enteric RNA virus-induced intestinal inflammation by targeting NLRP6 and NLRP9b signaling pathways. Mucosal Immunol 2025; 18:135-150. [PMID: 39396665 DOI: 10.1016/j.mucimm.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/31/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024]
Abstract
Infections by enteric virus and intestinal inflammation are recognized as a leading cause of deadly gastroenteritis, and NLRP6 and NLRP9b signaling control these infection and inflammation. However, the regulatory mechanisms of the NLRP6 and NLRP9b signaling in enteric viral infection remain unexplored. In this study, we found that the E3 ligase TRIM29 suppressed type III interferon (IFN-λ) and interleukin-18 (IL-18) production by intestinal epithelial cells (IECs) when exposed to polyinosinic:polycytidylic acid (poly I:C) and enteric RNA viruses. Knockout of TRIM29 in IECs was efficient to restrict intestinal inflammation triggered by the enteric RNA viruses, rotavirus in suckling mice, and the encephalomyocarditis virus (EMCV) in adults. This attenuation in inflammation was attributed to the increased production of IFN-λ and IL-18 in the IECs and more recruitment of intraepithelial protective Ly6A+CCR9+CD4+ T cells in small intestines from TRIM29-deficient mice. Mechanistically, TRIM29 promoted K48-linked ubiquitination, leading to the degradation of NLRP6 and NLRP9b, resulting in decreased IFN-λ and IL-18 secretion by IECs. Our findings reveal that enteric viruses utilize TRIM29 to inhibit IFN-λ and inflammasome activation in IECs, thereby facilitating viral-induced intestinal inflammation. This indicates that targeting TRIM29 could offer a promising therapeutic strategy for alleviating gut diseases.
Collapse
Affiliation(s)
- Junying Wang
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Academic Institute, Houston Methodist, Houston, TX 77030, USA
| | - Ling Wang
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Academic Institute, Houston Methodist, Houston, TX 77030, USA; Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, 130021, China
| | - Wenting Lu
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Academic Institute, Houston Methodist, Houston, TX 77030, USA
| | - Naser Farhataziz
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Academic Institute, Houston Methodist, Houston, TX 77030, USA
| | - Anastasia Gonzalez
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Academic Institute, Houston Methodist, Houston, TX 77030, USA
| | - Junji Xing
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Academic Institute, Houston Methodist, Houston, TX 77030, USA; Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston Methodist, Houston, TX 77030, USA; Department of Surgery, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| | - Zhiqiang Zhang
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Academic Institute, Houston Methodist, Houston, TX 77030, USA; Department of Surgery, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
4
|
Burke RM, Ramani S, Lynch J, Cooper LV, Cho H, Bandyopadhyay AS, Kirkwood CD, Steele AD, Kang G. Geographic disparities impacting oral vaccine performance: Observations and future directions. Clin Exp Immunol 2025; 219:uxae124. [PMID: 39774633 PMCID: PMC11773816 DOI: 10.1093/cei/uxae124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/01/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Oral vaccines have several advantages compared with parenteral administration: they can be relatively cheap to produce in high quantities, easier to administer, and induce intestinal mucosal immunity that can protect against infection. These characteristics have led to successful use of oral vaccines against rotavirus, polio, and cholera. Unfortunately, oral vaccines for all three diseases have demonstrated lower performance in the highest-burden settings where they are most needed. Rotavirus vaccines are estimated to have >85% effectiveness against hospitalization in children <12 months in countries with low child mortality, but only ~65% effectiveness in countries with high child mortality. Similarly, oral polio vaccines have lower immunogenicity in developing country settings compared with high-resource settings. Data are more limited for oral cholera vaccines, but suggest lower titers among children compared with adults, and, for some vaccines, lower efficacy in endemic settings compared with non-endemic settings. These disparities are likely multifactorial, and available evidence suggests a role for maternal factors (e.g. transplacental antibodies, breastmilk), host factors (e.g. genetic polymorphisms-with the best evidence for rotavirus-or previous infection), and environmental factors (e.g. gut microbiome, co-infections). Overall, these data highlight the rather ambiguous and often contradictory nature of evidence on factors affecting oral vaccine response, cautioning against broad extrapolation of outcomes based on one population or one vaccine type. Meaningful impact on performance of oral vaccines will likely only be possible with a suite of interventions, given the complex and multifactorial nature of the problem, and the degree to which contributing factors are intertwined.
Collapse
Affiliation(s)
- Rachel M Burke
- Global Development Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Julia Lynch
- Office of the Director General, International Vaccine Institute, Seoul, Republic of Korea
| | - Laura V Cooper
- School of Public Health, Imperial College London, London, UK
| | - Haeun Cho
- Department of Data Science and Innovation, International Vaccine Institute, Seoul, Republic of Korea
| | | | - Carl D Kirkwood
- Global Health Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - A Duncan Steele
- Global Health Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Gagandeep Kang
- Global Health Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| |
Collapse
|
5
|
Gebert JT, Scribano FJ, Engevik KA, Huleatt EM, Eledge MR, Dorn LE, Philip AA, Kawagishi T, Greenberg HB, Patton JT, Hyser JM. Viroporin activity is necessary for intercellular calcium signals that contribute to viral pathogenesis. SCIENCE ADVANCES 2025; 11:eadq8115. [PMID: 39823322 PMCID: PMC11740935 DOI: 10.1126/sciadv.adq8115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Viruses engage in a variety of processes to subvert host defenses and create an environment amenable to replication. Here, using rotavirus as a prototype, we show that calcium conductance out of the endoplasmic reticulum by the virus encoded ion channel, NSP4, induces intercellular calcium waves that extend beyond the infected cell and contribute to pathogenesis. Viruses that lack the ability to induce this signaling show diminished viral shedding and attenuated disease in a mouse model of rotavirus diarrhea. This implicates nonstructural protein 4 (NSP4) as a virulence factor and provides mechanistic insight into its mode of action. Critically, this signaling induces a transcriptional signature characteristic of interferon-independent innate immune activation, which is not observed in response to a mutant NSP4 that does not conduct calcium. This implicates calcium dysregulation as a means of pathogen recognition, a theme broadly applicable to calcium-altering pathogens beyond rotavirus.
Collapse
Affiliation(s)
- J. Thomas Gebert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Francesca J. Scribano
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kristen A. Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ethan M. Huleatt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael R. Eledge
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lauren E. Dorn
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| | - Asha A. Philip
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Takahiro Kawagishi
- Departments of Medicine and Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Harry B. Greenberg
- Departments of Medicine and Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John T. Patton
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Joseph M. Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Alkek Center for Metagenomics & Microbiome Research, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
6
|
Cuvry A, Molineaux L, Gozalbo-Rovira R, Neyts J, de Witte P, Rodríguez-Díaz J, Rocha-Pereira J. Human norovirus disturbs intestinal motility and transit time through its capsid proteins. PLoS Pathog 2024; 20:e1012710. [PMID: 39602402 PMCID: PMC11602112 DOI: 10.1371/journal.ppat.1012710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Human norovirus (HuNoV) accounts for over 700 million cases of gastroenteritis annually. Episodes of HuNoV disease are characterized by vomiting and diarrhea as the two most prominent symptoms. Despite its prevalence, our understanding of the pathophysiological mechanisms triggered upon HuNoV infection is limited, mainly due to a lack of suitable animal models. Our aim was to use the recent HuNoV zebrafish larvae model to study the effect of HuNoV infection on intestinal motility and investigate whether one viral protein could act as an enterotoxin, as seen with rotavirus. We studied whether HuNoV infection affects the contraction frequency of the intestinal bulb and the posterior intestine as well as the transit time. Infection of larvae, following injection of a HuNoV GII.4-containing stool sample in the yolk, resulted in an increased contraction frequency in the intestinal bulb. A comparable effect was observed in serotonin-treated larvae, corresponding to the natural function of serotonin. The higher replication efficacy of HuNoV GII.4 likely explains why they have a more marked effect on gut motility, when compared to other genotypes. Additionally, transit time of fluorescent food was prolonged in HuNoV GII.4 infected larvae, suggesting a loss of coordination in bowel movements upon infection. To identify the proteins responsible for the effect, individual HuNoV non-structural proteins and virus-like particles (VLPs) were injected intraperitoneally (ip). VLPs carrying VP1/VP2, but not those with only VP1, induced increased contraction frequencies in the intestinal bulb in a dose-dependent manner. In conclusion, our findings suggest that the viral capsid and potentially the minor capsid protein VP2 play a crucial role in the aetiology of symptoms associated with HuNoV, potentially acting as a viral enterotoxin. This work contributes to the understanding of the pathophysiological mechanisms in HuNoV-induced disease and further attests zebrafish as a valuable HuNoV disease model.
Collapse
Affiliation(s)
- Arno Cuvry
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, Leuven, Belgium
- KU Leuven, Translationeel Onderzoek van Gastro-enterologische Aandoeningen (TARGID), Leuven, Belgium
| | - Lorane Molineaux
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, Leuven, Belgium
| | | | - Johan Neyts
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology, Antiviral Drug & Vaccine Research Group, Leuven, Belgium
| | - Peter de Witte
- KU Leuven, Laboratory for Molecular Biodiscovery, KU Leuven-Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Jesús Rodríguez-Díaz
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain
| | - Joana Rocha-Pereira
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Virus-Host Interactions & Therapeutic Approaches (VITA) Research Group, Leuven, Belgium
| |
Collapse
|
7
|
Yan H, Dong B, Li X, He J, Yu B, Mao X, Yu J, Luo Y, Luo J, Wu A, Pu J, Wang Q, Wang H, Crenshaw J, Shen Y, Chen D. Spray-dried plasma protects against rotavirus-induced gastroenteritis via regulating macrophage and T cells divergence in weanling pigs. Front Vet Sci 2024; 11:1467108. [PMID: 39479205 PMCID: PMC11523297 DOI: 10.3389/fvets.2024.1467108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
Infectious gastroenteritis is the major cause for diarrhea in piglets. The protection of spray-dried plasma (SDP) on viral gastroenteritis during the progression of rotavirus (RV) infection remain unclear. In this study, 64 weanling piglets were randomly assigned to control diets (n = 40) and SDP diets (n = 24) for 14 days, and then pigs were challenged with RV on day 15. Pigs were sacrificed on day 14 (normal condition), day 18 (manifestation stage), and day 21 (convalescent stage) of the trial. Prior to RV infection, SDP increased ADG, M1 macrophages and CD4+ T cells in different organs without increasing proinflammatory cytokines, indicating a more robust immunity with less inflammation. During the manifestation of infection, SDP enhanced mucosal immunity by increasing M1 macrophages, M1/M2 ratio and cytokines in mucosa and increasing intraepithelial CD8+ T cells for RV clearance. During the convalescence, SDP promoted M2 macrophage polarization and reduced pro-inflammatory cytokines to facilitate intestinal repair and prevent prolonged inflammation. Collectively, SDP enhanced mucosal immunity to promote viral clearance and maintained immune homeostasis to prevent long-lasting inflammation as a therapeutically approach for infectious gastroenteritis.
Collapse
Affiliation(s)
- Hui Yan
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Biqiong Dong
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xipeng Li
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jun He
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Bing Yu
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xiangbing Mao
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jie Yu
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yuheng Luo
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Junqiu Luo
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Aimin Wu
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Junning Pu
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Quyuan Wang
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Huifen Wang
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Joe Crenshaw
- R&D Department, APC LLC, Ankeny, IA, United States
| | - Yanbin Shen
- R&D Department, APC LLC, Ankeny, IA, United States
| | - Daiwen Chen
- Key Laboratories for Animal Disease-Resistance Nutrition of China Ministry of Education, China Ministry of Agriculture and Rural Affairs and Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
8
|
Novikov DV, Vasilchikova EA, Vasilchikov PI. Prospects for the use of viral proteins for the construction of chimeric toxins. Arch Virol 2024; 169:208. [PMID: 39327316 DOI: 10.1007/s00705-024-06139-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/09/2024] [Indexed: 09/28/2024]
Abstract
One of the actively developing areas of drug development is the creation of chimeric toxins, recombinant bifunctional molecules designed to affect target cells selectively. The prevalent approach involves fusing bacterial and plant toxins with molecules that facilitate targeted delivery. However, the therapeutic use of such toxins often encounters challenges associated with negative side effects. Concurrently, viruses encode proteins possessing toxin-like properties, exerting multiple effects on the vital activity of cells. In contrast to bacterial and plant toxins, the impact of viral proteins is typically milder, presenting a significant advantage by potentially reducing the likelihood of side effects. This review delineates the characteristics of extensively studied viral proteins with toxic and immunomodulatory properties and explores the prospects of incorporating them into chimeric toxins.
Collapse
Affiliation(s)
- D V Novikov
- Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology, Nizhny Novgorod, Russia
| | - E A Vasilchikova
- National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - P I Vasilchikov
- National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.
| |
Collapse
|
9
|
Herbert J, van Dijk AA. Identification of a cooperative effect between amino acids 169 and 174 in the rotavirus NSP4 double-layered particle-binding domain. J Gen Virol 2024; 105. [PMID: 39320365 DOI: 10.1099/jgv.0.002029] [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] [Indexed: 09/26/2024] Open
Abstract
Segmented RNA viruses are capable of exchanging genome segments via reassortment as a means of immune evasion and to maintain viral fitness. Reassortments of single-genome segments are common among group A rotaviruses. Multiple instances of co-reassortment of two genome segments, GS6(VP6) and GS10(NSP4), have been documented in surveillance. Specifically, a division between NSP4 genotypes has been observed in the NSP4 double-layered particle (DLP)-binding domain. A previously hypothesized mechanism for this co-reassortment has been suggested to be the interaction between VP6 and NSP4 during DLP transport from viroplasms for particle maturation. In this study, we used sequence analysis, RNA secondary structure prediction, molecular dynamics and reverse genetics to form a hypothesis regarding the role of the NSP4 DLP-binding domain. Sequence analysis showed that the polarity of NSP4 DLP-binding domain amino acids 169 and 174 is clearly divided between E1 and E2 NSP4 genotypes. Viruses with E1 NSP4s had 169A/I or 169S/T with 174S. E2 NSP4s had 169R/K and 174A. RNA secondary structure prediction showed that mutation in both 545 (aa169) and 561 (aa174) causes global structure remodelling. Molecular dynamics showed that the NSP4/VP6 interaction stability is increased by mutating both aa positions 169 and 174. Using reverse genetics, we showed that an R169I mutation alone does not prevent rescue. Conversely, 174A to 174S prevented rescue, and rescue could be returned by combining 174S with 169I. When compared to rSA11 NSP4-wt, both rSA11 NSP4-R169I and rSA11 NSP4-R169I/A174S had a negligible but significant reduction in titre at specific time points. This study suggests that amino acid 174 of NSP4 may be essential in maintaining the VP6/NSP4 interaction required for DLP transport. Our results suggest that maintenance of specific polarities of amino acids at positions 169 and 174 may be required for the fitness of rotavirus field strains.
Collapse
Affiliation(s)
- Jayme Herbert
- University of the Free State, Bloemfontein, South Africa
- Deltamune PTY (LTD), Pretoria, South Africa
| | | |
Collapse
|
10
|
Li S, Tang X, Zhou J, Bian X, Wang J, Gu L, Zhu X, Tao R, Sun M, Zhang X, Li B. The synergy of recombinant NSP4 and VP4 from porcine rotavirus elicited a strong mucosal response. Virology 2024; 597:110130. [PMID: 38850894 DOI: 10.1016/j.virol.2024.110130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/29/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Porcine rotavirus (PoRV) is one of the main pathogens causing diarrhea in piglets, and multiple genotypes coexist. However, an effective vaccine is currently lacking. Here, the potential adjuvant of nonstructural protein 4 (NSP4) and highly immunogenic structural protein VP4 prompted us to construct recombinant NSP486-175aa (NSP4*) and VP426-476aa (VP4*) proteins, combine them as immunogens to evaluate their efficacy. Results indicated that NSP4* enhanced systemic and local mucosal responses induced by VP4*. The VP4*-IgG, VP4*-IgA in feces and IgA-secreting cells in intestines induced by the co-immunization were significantly higher than those induced by VP4* alone. Co-immunization of NSP4* and VP4* also induced strong cellular immunity with significantly increased IFN-λ than the single VP4*. Summarily, the NSP4* as a synergistical antigen exerted limited effects on the PoRV NAbs elevation, but conferred strong VP4*-specific mucosal and cellular efficacy, which lays the foundation for the development of a more effective porcine rotavirus subunit vaccine.
Collapse
Affiliation(s)
- Sufen Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xuechao Tang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China
| | - Jinzhu Zhou
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225000, China; GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, 210014, China
| | - Xianyu Bian
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China
| | - Jianxin Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China
| | - Laqiang Gu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China
| | - Xuejiao Zhu
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225000, China; GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, 210014, China
| | - Ran Tao
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China
| | - Min Sun
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225000, China; GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, 210014, China
| | - Xuehan Zhang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225000, China; GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, 210014, China.
| | - Bin Li
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agricultural and Rural Affairs, Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Nanjing, 210014, China; College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225000, China; GuoTai (Taizhou) Center of Technology Innovation for Veterinary Biologicals, Taizhou, 210014, China.
| |
Collapse
|
11
|
Kotaki T, Kanai Y, Onishi M, Minami S, Chen Z, Nouda R, Nurdin JA, Yamasaki M, Kobayashi T. Generation of single-round infectious rotavirus with a mutation in the intermediate capsid protein VP6. J Virol 2024; 98:e0076224. [PMID: 38837379 PMCID: PMC11265344 DOI: 10.1128/jvi.00762-24] [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: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 06/07/2024] Open
Abstract
Rotavirus causes severe diarrhea in infants. Although live attenuated rotavirus vaccines are available, vaccine-derived infections have been reported, which warrants development of next-generation rotavirus vaccines. A single-round infectious virus is a promising vaccine platform; however, this platform has not been studied extensively in the context of rotavirus. Here, we aimed to develop a single-round infectious rotavirus by impairing the function of the viral intermediate capsid protein VP6. Recombinant rotaviruses harboring mutations in VP6 were rescued using a reverse genetics system. Mutations were targeted at VP6 residues involved in virion assembly. Although the VP6-mutated rotavirus expressed viral proteins, it did not produce progeny virions in wild-type cells; however, the virus did produce progeny virions in VP6-expressing cells. This indicates that the VP6-mutated rotavirus is a single-round infectious rotavirus. Insertion of a foreign gene, and replacement of the VP7 gene segment with that of human rotavirus clinical isolates, was successful. No infectious virions were detected in mice infected with the single-round infectious rotavirus. Immunizing mice with the single-round infectious rotavirus induced neutralizing antibody titers as high as those induced by wild-type rotavirus. Taken together, the data suggest that this single-round infectious rotavirus has potential as a safe and effective rotavirus vaccine. This system is also applicable for generation of safe and orally administrable viral vectors.IMPORTANCERotavirus, a leading cause of acute gastroenteritis in infants, causes an annual estimated 128,500 infant deaths worldwide. Although live attenuated rotavirus vaccines are available, they are replicable and may cause vaccine-derived infections. Thus, development of safe and effective rotavirus vaccine is important. In this study, we report the development of a single-round infectious rotavirus that can replicate only in cells expressing viral VP6 protein. We demonstrated that (1) the single-round infectious rotavirus did not replicate in wild-type cells or in mice; (2) insertion of foreign genes and replacement of the outer capsid gene were possible; and (3) it was as immunogenic as the wild-type virus. Thus, the mutated virus shows promise as a next-generation rotavirus vaccine. The system is also applicable to orally administrable viral vectors, facilitating development of vaccines against other enteric pathogens.
Collapse
Affiliation(s)
- Tomohiro Kotaki
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yuta Kanai
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Megumi Onishi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shohei Minami
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Zelin Chen
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ryotaro Nouda
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Jeffery A. Nurdin
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Moeko Yamasaki
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| |
Collapse
|
12
|
Dellis C, Tatsi EB, Koukou DM, Filippatos F, Vetouli EE, Zoumakis E, Michos A, Syriopoulou V. Genotyping and Molecular Characterization of VP6 and NSP4 Genes of Unusual Rotavirus Group A Isolated from Children with Acute Gastroenteritis. Adv Virol 2024; 2024:3263228. [PMID: 38993264 PMCID: PMC11239230 DOI: 10.1155/2024/3263228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/29/2024] [Accepted: 05/27/2024] [Indexed: 07/13/2024] Open
Abstract
Group A rotavirus (RVA), which causes acute gastroenteritis (AGE) in children worldwide, is categorized mainly based on VP7 (genotype G) and VP4 (genotype P) genes. Genotypes that circulate at <1% are considered unusual. Important genes also include VP6 (genotype I) and NSP4 (genotype E). VP6 establishes the group and affects immunogenicity, while NSP4, as an enterotoxin, is responsible for the clinical symptoms. The aim of this study was to genotype the VP6 and NSP4 genes and molecularly characterize the NSP4 and VP6 genes of unusual RVA. Unusual RVA strains extracted from fecal samples of children ≤16 years with AGE were genotyped in VP6 and NSP4 genes with Sanger sequencing. In a 15-year period (2007-2021), 54.8% (34/62) of unusual RVA were successfully I and E genotyped. Three different I and E genotypes were identified; I2 (73.5%, 25/34) and E2 (35.3%, 12/34) were the most common. E3 genotype was detected from 2017 onwards. The uncommon combination of I2-E3 was found in 26.5% (9/34) of the strains and G3-P[9]-I2-E3 remained the most frequent G-P-I-E combination (20.6%, 7/34). Children infected with RVA E2 strains had a statistically higher frequency of dehydration (50%) than those infected with RVA E3 strains (p = 0.019). Multiple substitutions were detected in NSP4, but their functional effect remains unknown. The result indicates the genetic diversity of RVA strains. Continuous surveillance of the RVA based on the whole genome will provide better knowledge of its evolution.
Collapse
Affiliation(s)
- Charilaos Dellis
- First Department of Pediatrics Infectious Diseases and Chemotherapy Research Laboratory Medical School National and Kapodistrian University of Athens "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Elizabeth-Barbara Tatsi
- First Department of Pediatrics Infectious Diseases and Chemotherapy Research Laboratory Medical School National and Kapodistrian University of Athens "Aghia Sophia" Children's Hospital, Athens, Greece
- University Research Institute of Maternal and Child Health and Precision Medicine, Athens, Greece
| | - Dimitra-Maria Koukou
- First Department of Pediatrics Infectious Diseases and Chemotherapy Research Laboratory Medical School National and Kapodistrian University of Athens "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Filippos Filippatos
- First Department of Pediatrics Infectious Diseases and Chemotherapy Research Laboratory Medical School National and Kapodistrian University of Athens "Aghia Sophia" Children's Hospital, Athens, Greece
| | | | - Emmanouil Zoumakis
- First Department of Pediatrics Infectious Diseases and Chemotherapy Research Laboratory Medical School National and Kapodistrian University of Athens "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Athanasios Michos
- First Department of Pediatrics Infectious Diseases and Chemotherapy Research Laboratory Medical School National and Kapodistrian University of Athens "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Vasiliki Syriopoulou
- First Department of Pediatrics Infectious Diseases and Chemotherapy Research Laboratory Medical School National and Kapodistrian University of Athens "Aghia Sophia" Children's Hospital, Athens, Greece
| |
Collapse
|
13
|
Fukuda Y, Kondo K, Nakata S, Morita Y, Adachi N, Kogawa K, Ukae S, Kudou Y, Adachi S, Yamamoto M, Fukumura S, Tsugawa T. Whole-genome analysis of human group A rotaviruses in 1980s Japan and evolutionary assessment of global Wa-like strains across half a century. J Gen Virol 2024; 105. [PMID: 38836747 DOI: 10.1099/jgv.0.001998] [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] [Indexed: 06/06/2024] Open
Abstract
Historically, the Wa-like strains of human group A rotavirus (RVA) have been major causes of gastroenteritis. However, since the 2010s, the circulation of non-Wa-like strains has been increasingly reported, indicating a shift in the molecular epidemiology of RVA. Although understanding RVA evolution requires the analysis of both current and historical strains, comprehensive pre-1980's sequencing data are scarce globally. We determined the whole-genome sequences of representative strains from six RVA gastroenteritis outbreaks observed at an infant home in Sapporo, Japan, between 1981 and 1989. These outbreaks were mainly caused by G1 or G3 Wa-like strains, resembling strains from the United States in the 1970s-1980s and from Malawi in the 1990s. Phylogenetic analysis of these infant home strains, together with Wa-like strains collected worldwide from the 1970s to 2020, revealed a notable trend: pre-2010 strains diverged into multiple lineages in many genomic segments, whereas post-2010 strains tended to converge into a single lineage. However, Bayesian skyline plot indicated near-constant effective population sizes from the 1970s to 2020, and selection pressure analysis identified positive selection only at amino acid 75 of NSP2. These results suggest that evidence supporting the influence of rotavirus vaccines, introduced globally since 2006, on Wa-like RVA molecular evolution is lacking at present, and phylogenetic analysis may simply reflect natural fluctuations in RVA molecular evolution. Evaluating the long-term impact of RV vaccines on the molecular evolution of RVA requires sustained surveillance.
Collapse
Affiliation(s)
- Yuya Fukuda
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenji Kondo
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shuji Nakata
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuyuki Morita
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Noriaki Adachi
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keiko Kogawa
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Susumu Ukae
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoshimasa Kudou
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shuhei Adachi
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaki Yamamoto
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinobu Fukumura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takeshi Tsugawa
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
14
|
Engevik KA, Scribano FJ, Gebert JT, Hyser JM. Purinergic Signaling Drives Multiple Aspects of Rotavirus Pathophysiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.592953. [PMID: 38765995 PMCID: PMC11100750 DOI: 10.1101/2024.05.07.592953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Rotavirus causes life-threatening diarrhea in children, resulting in ∼200,000 deaths/year. The current treatment during infection is Oral Rehydration Solution which successfully replenishes fluids but does not alleviate diarrhea volume or severity. As a result, there is an urgent need to better understand rotavirus pathophysiology and develop more effective pediatric therapeutics. Rotavirus primarily infects the tips of small intestinal villi, yet has far-reaching effects on cell types distant from infected cells. We recently identified that rotavirus infected cells release the purinergic signaling molecule ADP, which activates P2Y1 receptors on nearby uninfected cells in vitro . To elucidate the role of purinergic signaling via P2Y1 receptors during rotavirus infection in vivo , we used the mouse-like rotavirus strain D6/2 which generates a severe infection in mice. C57BL/6J mouse pups were given an oral gavage of D6/2 rotavirus and assessed over the course of 5-7 days. Beginning at day 1 post infection, infected pups were treated daily by oral gavage with saline or 4 mg/kg MRS2500, a selective P2Y1 antagonist. Mice were monitored for diarrhea severity, diarrhea incidence, and viral shedding. Neonatal mice were euthanized at days 3 and 5 post-infection and small intestine was collected to observe infection. MRS2500 treatment decreased the severity, prevalence, and incidence of rotavirus diarrhea. Viral stool shedding, assessed by qPCR for rotavirus gene levels, revealed that MRS2500 treated pups had significantly lower viral shedding starting at day 4 post infection compared to saline treated pups, which suggests P2Y1 signaling may enhance rotavirus replication. Finally, we found that inhibition of P2Y1 with MRS2500 limited transmitted rotavirus diarrhea to uninfected pups within a litter. Together, these results suggest that P2Y1 signaling is involved in the pathogenesis of a homologous murine rotavirus strain, making P2Y1 receptors a promising anti-diarrheal, anti-viral therapeutic target to reduce rotavirus disease burden.
Collapse
|
15
|
Gebert JT, Scribano FJ, Engevik KA, Philip AA, Kawagishi T, Greenberg HB, Patton JT, Hyser JM. Viroporin activity from rotavirus nonstructural protein 4 induces intercellular calcium waves that contribute to pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.592929. [PMID: 38765992 PMCID: PMC11100692 DOI: 10.1101/2024.05.07.592929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Acute gastroenteritis remains the second leading cause of death among children under the age of 5 worldwide. While enteric viruses are the most common etiology, the drivers of their virulence remain incompletely understood. We recently found that cells infected with rotavirus, the most prevalent enteric virus in infants and young children, initiate hundreds of intercellular calcium waves that enhance both fluid secretion and viral spread. Understanding how rotavirus triggers intercellular calcium waves may allow us to design safer, more effective vaccines and therapeutics, but we still lack a mechanistic understanding of this process. In this study, we used existing virulent and attenuated rotavirus strains, as well as reverse engineered recombinants, to investigate the role of rotavirus nonstructural protein 4 (NSP4) in intercellular calcium wave induction using in vitro , organoid, and in vivo model systems. We found that the capacity to induce purinergic intercellular calcium waves (ICWs) segregated with NSP4 in both simian and murine-like rotavirus backgrounds, and NSP4 expression alone was sufficient to induce ICWs. NSP4's ability to function as a viroporin, which conducts calcium out of the endoplasmic reticulum, was necessary for ICW induction. Furthermore, viroporin activity and the resulting ICWs drove transcriptional changes indicative of innate immune activation, which were lost upon attenuation of viroporin function. Multiple aspects of RV disease severity in vivo correlated with the generation of ICWs, identifying a critical link between viroporin function, intercellular calcium waves, and enteric viral virulence.
Collapse
|
16
|
Yang H, Fan X, Mao X, Yu B, He J, Yan H, Wang J. The protective role of prebiotics and probiotics on diarrhea and gut damage in the rotavirus-infected piglets. J Anim Sci Biotechnol 2024; 15:61. [PMID: 38698473 PMCID: PMC11067158 DOI: 10.1186/s40104-024-01018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/29/2024] [Indexed: 05/05/2024] Open
Abstract
Rotavirus is one of the pathogenic causes that induce diarrhea in young animals, especially piglets, worldwide. However, nowadays, there is no specific drug available to treat the disease, and the related vaccines have no obvious efficiency in some countries. Via analyzing the pathogenesis of rotavirus, it inducing diarrhea is mainly due to disturb enteric nervous system, destroy gut mucosal integrity, induce intracellular electrolyte imbalance, and impair gut microbiota and immunity. Many studies have already proved that prebiotics and probiotics can mitigate the damage and diarrhea induced by rotavirus infection in hosts. Based on these, the current review summarizes and discusses the effects and mechanisms of prebiotics and probiotics on rotavirus-induced diarrhea in piglets. This information will highlight the basis for the swine production utilization of prebiotics and probiotics in the prevention or treatment of rotavirus infection in the future.
Collapse
Affiliation(s)
- Heng Yang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Xiangqi Fan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China.
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, No. 211, Gongpinghuimin Road, Wenjiang District, Chengdu, Sichuan Province, 611130, People's Republic of China
| |
Collapse
|
17
|
Carossino M, Vissani MA, Barrandeguy ME, Balasuriya UBR, Parreño V. Equine Rotavirus A under the One Health Lens: Potential Impacts on Public Health. Viruses 2024; 16:130. [PMID: 38257830 PMCID: PMC10819593 DOI: 10.3390/v16010130] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/29/2023] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Group A rotaviruses are a well-known cause of viral gastroenteritis in infants and children, as well as in many mammalian species and birds, affecting them at a young age. This group of viruses has a double-stranded, segmented RNA genome with high genetic diversity linked to point mutations, recombination, and, importantly, reassortment. While initial molecular investigations undertaken in the 1900s suggested host range restriction among group A rotaviruses based on the fact that different gene segments were distributed among different animal species, recent molecular surveillance and genome constellation genotyping studies conducted by the Rotavirus Classification Working Group (RCWG) have shown that animal rotaviruses serve as a source of diversification of human rotavirus A, highlighting their zoonotic potential. Rotaviruses occurring in various animal species have been linked with contributing genetic material to human rotaviruses, including horses, with the most recent identification of equine-like G3 rotavirus A infecting children. The goal of this article is to review relevant information related to rotavirus structure/genomic organization, epidemiology (with a focus on human and equine rotavirus A), evolution, inter-species transmission, and the potential zoonotic role of equine and other animal rotaviruses. Diagnostics, surveillance and the current status of human and livestock vaccines against RVA are also reviewed.
Collapse
Affiliation(s)
- Mariano Carossino
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Maria Aldana Vissani
- Escuela de Veterinaria, Facultad de Ciencias Agrarias y Veterinarias, Universidad del Salvador, Pilar, Buenos Aires B1630AHU, Argentina; (M.A.V.); (M.E.B.)
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| | - Maria E. Barrandeguy
- Escuela de Veterinaria, Facultad de Ciencias Agrarias y Veterinarias, Universidad del Salvador, Pilar, Buenos Aires B1630AHU, Argentina; (M.A.V.); (M.E.B.)
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
| | - Udeni B. R. Balasuriya
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
- Louisiana Animal Disease Diagnostic Laboratory, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Viviana Parreño
- Instituto de Virología, CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires B1686LQF, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| |
Collapse
|
18
|
Greigert V, Saraav I, Son J, Zhu Y, Dayao D, Antia A, Tzipori S, Witola WH, Stappenbeck TS, Ding S, Sibley LD. Cryptosporidium infection of human small intestinal epithelial cells induces type III interferon and impairs infectivity of Rotavirus. Gut Microbes 2024; 16:2297897. [PMID: 38189373 PMCID: PMC10793699 DOI: 10.1080/19490976.2023.2297897] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 01/09/2024] Open
Abstract
Cryptosporidiosis is a major cause of severe diarrheal disease in infants from resource poor settings. The majority of infections are caused by the human-specific pathogen C. hominis and absence of in vitro growth platforms has limited our understanding of host-pathogen interactions and development of effective treatments. To address this problem, we developed a stem cell-derived culture system for C. hominis using human enterocytes differentiated under air-liquid interface (ALI) conditions. Human ALI cultures supported robust growth and complete development of C. hominis in vitro including all life cycle stages. Cryptosporidium infection induced a strong interferon response from enterocytes, possibly driven, in part, by an endogenous dsRNA virus in the parasite. Prior infection with Cryptosporidium induced type III IFN secretion and consequently blunted infection with Rotavirus, including live attenuated vaccine strains. The development of hALI provides a platform for further studies on human-specific pathogens, including clinically important coinfections that may alter vaccine efficacy.
Collapse
Affiliation(s)
- Valentin Greigert
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Iti Saraav
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Juhee Son
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yinxing Zhu
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Denise Dayao
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - William H. Witola
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Thaddeus S. Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
19
|
The Potential Role of Microorganisms on Enteric Nervous System Development and Disease. Biomolecules 2023; 13:biom13030447. [PMID: 36979382 PMCID: PMC10046024 DOI: 10.3390/biom13030447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/14/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The enteric nervous system (ENS), the inherent nervous system of the gastrointestinal (GI) tract is a vast nervous system that controls key GI functions, including motility. It functions at a critical interface between the gut luminal contents, including the diverse population of microorganisms deemed the microbiota, as well as the autonomic and central nervous systems. Critical development of this axis of interaction, a key determinant of human health and disease, appears to occur most significantly during early life and childhood, from the pre-natal through to the post-natal period. These factors that enable the ENS to function as a master regulator also make it vulnerable to damage and, in turn, a number of GI motility disorders. Increasing attention is now being paid to the potential of disruption of the microbiota and pathogenic microorganisms in the potential aetiopathogeneis of GI motility disorders in children. This article explores the evidence regarding the relationship between the development and integrity of the ENS and the potential for such factors, notably dysbiosis and pathogenic bacteria, viruses and parasites, to impact upon them in early life.
Collapse
|
20
|
Kachooei A, Tava Koli A, Minaeian S, Hosseini M, Jalilvand S, Latifi T, Arashkia A, Ataei-Pirkooh A, Shoja Z. Molecular characterization of rotavirus infections in children less than 5 years of age with acute gastroenteritis in Tehran, Iran, 2021-2022: Emergence of uncommon G9P[4] and G9P[8] rotavirus strains. J Med Virol 2023; 95:e28529. [PMID: 36698258 DOI: 10.1002/jmv.28529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023]
Abstract
The present study was conducted to monitor the genotypes of circulating species A rotavirus (RVA) in Iran and investigate genetic linkages between specific RVA VP7, VP4, VP6, and NSP4 segments. For this purpose, 48 RVA strains were detected during the 2021-2022 seasons. The two combinations of G9P[4] and G9P[8] RVA strains were predominant. However, several other combinations of RVA also were detected. Based on the distribution of I and E genotypes (46 strains) with respect to G and P, the most common strains were G9P[4]-I2-E2 (19.5%), G9P[4]-I2-E1 (6.5%), G9P[4]-I1-E1 (4.3%), G9P[8]-I1-E1 (19.5%), and G9P[8]-I2-E2 (10.9%), which were followed by several other combinations of G and P RVA strains with different pattern of I-E genotypes and also emerging, rare and uncommon strains. The present study described the continued circulation of G9 strains with the emergence of uncommon G9P[4] and G9P[8] reassortants with three and two different I-E genotypes, respectively, which have not been reported previously in Iran. Our findings indicated that these uncommon strains exhibited a unique genotype pattern comprising a mixture of genogroup 1 and 2 genes and suggest the need for further analysis of rare, uncommon, and emerging strains of RVA at all 11 gene segments to determine intergenogroup and intragenotype reassortments.
Collapse
Affiliation(s)
- Atefeh Kachooei
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Ahmad Tava Koli
- Rsearch Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Minaeian
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdieh Hosseini
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran.,Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Angila Ataei-Pirkooh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zabihollah Shoja
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran.,Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
21
|
Tate JE, Cortese MM, Offit PA, Parashar UD. Rotavirus Vaccines. PLOTKIN'S VACCINES 2023:1005-1024.e11. [DOI: 10.1016/b978-0-323-79058-1.00053-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
22
|
Rotavirus Downregulates Tyrosine Hydroxylase in the Noradrenergic Sympathetic Nervous System in Ileum, Early in Infection and Simultaneously with Increased Intestinal Transit and Altered Brain Activities. mBio 2022; 13:e0138722. [PMID: 36094089 PMCID: PMC9600178 DOI: 10.1128/mbio.01387-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
While rotavirus diarrhea has been considered to occur only due to intrinsic intestinal effects within the enteric nervous system, we provide evidence for central nervous system control underlying the clinical symptomology. Our data visualize infection by large-scale three-dimensional (3D) volumetric tissue imaging of a mouse model and demonstrate that rotavirus infection disrupts the homeostasis of the autonomous system by downregulating tyrosine hydroxylase in the noradrenergic sympathetic nervous system in ileum, concomitant with increased intestinal transit. Interestingly, the nervous response was found to occur before the onset of clinical symptoms. In adult infected animals, we found increased pS6 immunoreactivity in the area postrema of the brain stem and decreased phosphorylated STAT5-immunoreactive neurons in the bed nucleus of the stria terminalis, which has been associated with autonomic control, including stress response. Our observations contribute to knowledge of how rotavirus infection induces gut-nerve-brain interaction early in the disease.
Collapse
|
23
|
Cao H, Wu J, Luan N, Wang Y, Lin K, Liu C. Evaluation of a bivalent recombinant vaccine candidate targeting norovirus and rotavirus: Antibodies to rotavirus NSP4 exert antidiarrheal effects without virus neutralization. J Med Virol 2022; 94:3847-3856. [PMID: 35474320 DOI: 10.1002/jmv.27809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/19/2022] [Accepted: 04/23/2022] [Indexed: 11/10/2022]
Abstract
We previously found that when tandemly expressed with SR69A -VP8*, nonstructural protein 4 (NSP4) of the rotavirus Wa strain exerts a minor effect on elevating the antibody responses targeting the rotavirus antigen VP8* of the 60-valent nanoparticle SR69A -VP8* but could fully protect mice from diarrhea induced by the rotavirus strain Wa. In this study, we chose comparably less immunogenic norovirus 24-valent P particles with homogenous (i.e., VP8* from rotavirus) and heterogeneous (i.e., protruding domain of norovirus) antigens and in more challenging rotavirus SA11 strain-induced diarrhea mouse models to evaluate its main role in recombinant gastroenteritis virus-specific vaccines. The results showed that although as an adjuvant NSP4 exerted limited effects on the elevation of norovirus-specific or VP8*-specific neutralizing antibody production, as an antigen it could confer potent protection, particularly when synergized with VP8*, in rotavirus SA11 strain-induced diarrhea mouse models, possibly blocking the invasion of the intestinal wall by enterotoxin. NSP4 may be unnecessary for other recombinant vaccines as adjuvants, and its display mode should be evaluated specifically to avoid blocking coexpressed antigens in the norovirus P particles. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Han Cao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| | - Jinyuan Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| | - Ning Luan
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| | - Yunfei Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| | - Kangyang Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| | - Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan, China
| |
Collapse
|
24
|
Motayo BO, Faneye AO, Adeniji JA. VP7, VP4, and NSP4 genes of species a rotaviruses isolated from sewage in Nigeria, 2014/2015: partial sequence characterization and biophysical analysis of NSP4 (enterotoxin). Virus Genes 2022; 58:180-187. [PMID: 35303217 DOI: 10.1007/s11262-022-01895-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/08/2022] [Indexed: 10/18/2022]
Abstract
Species A rotavirus are an important cause of childhood gastroenteritis, and the main contributor to its pathogenicity is the enterotoxin (NSP4) protein. Some biophysical properties of partial NSP4 genes of RVAs isolated from sewage in Nigeria during 2014/2015 were investigated. Samples were typed by RT-PCR and Sanger sequencing of partial VP4, VP7 and NSP4 genes. Phylogeny identified lineages within genotypes, predicted glycosylation sites; hydrophobicity profiles and amino acid alignments were employed to determine some biophysical properties of the NSP4 protein. The VP7 sequences of our isolates were the most diversified, the majority of the isolates carried NSP4 genes of the E1 genotype. Genotype specific variations both in hydrophobicity and potential glycosylation were identified, mutations were highest within the H3 hydrophobic domain and VP4 binding domain. The study of RVA NSP4 genes from non-clinical samples revealed that there were structural consistencies with those of reference genes.
Collapse
Affiliation(s)
- Babatunde O Motayo
- Department of Medical Microbiology, Federal Medical Center, Abeokuta, Nigeria.
| | | | | |
Collapse
|
25
|
Melnik LI, Garry RF. Enterotoxigenic Escherichia coli Heat-Stable Toxin and Ebola Virus Delta Peptide: Similarities and Differences. Pathogens 2022; 11:pathogens11020170. [PMID: 35215114 PMCID: PMC8878840 DOI: 10.3390/pathogens11020170] [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: 12/09/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) STb toxin exhibits striking structural similarity to Ebola virus (EBOV) delta peptide. Both ETEC and EBOV delta peptide are enterotoxins. Comparison of the structural and functional similarities and differences of these two toxins illuminates features that are important in induction of pathogenesis by a bacterial and viral pathogen.
Collapse
Affiliation(s)
- Lilia I. Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Viral Hemorrhagic Fever Consortium, New Orleans, LA 70112, USA
- Correspondence: ; Tel.: +1-(504)988-3818
| | - Robert F. Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Viral Hemorrhagic Fever Consortium, New Orleans, LA 70112, USA
| |
Collapse
|
26
|
Afchangi A, Jalilvand S, Arashkia A, Latifi T, Farahmand M, Abolghasem Shirazi MM, Mousavi Nasab SD, Marashi SM, Roohvand F, Shoja Z. Co-administration of rotavirus nanospheres VP6 and NSP4 proteins enhanced the anti-NSP4 humoral responses in immunized mice. Microb Pathog 2022; 163:105405. [PMID: 35045328 DOI: 10.1016/j.micpath.2022.105405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/28/2021] [Accepted: 01/12/2022] [Indexed: 11/29/2022]
Abstract
Inconveniences associated with the efficacy and safety of the World Health Organization (WHO) approved/prequalified live attenuated rotavirus (RV) vaccines, sounded for finding alternative non-replicating modals and proper RV antigens (Ags). Herein, we report the development of a RV candidate vaccine based on the combination of RV VP6 nanospheres (S) and NSP4112-175 proteins (VP6S + NSP4). Self-assembled VP6S protein was produced in insect cells. Analyses by western blotting and transmission electron microscopy (TEM) indicated expression of VP6 trimer structures with sizes of ≥140 kDa and presence of VP6S. Four group of mice were immunized (2-dose formulation) intra-peritoneally (IP) by either¨VP6S + NSP4¨ or each protein alone (VP6S or NSP4112-175) emulsified in aluminium hydroxide or control. Results indicated that VP6S + NSP4 formulation induced significant anti-VP6 IgG (P < 0.001) and IgA (P < 0.05) as well as anti-NSP4 IgG (P < 0.001) and enhancement of protective immunity. Analyses of anti-VP6S and anti-NSP4 IgG subclass (IgG1 and IgG2a) showed IgG1/IgG2a ≥6 and IgG1/IgG2a ≥3 ratios, respectively indicating Th2 polarization of immune responses. The combination of VP6S + NSP4 proteins emulsified in aluminum hydroxide adjuvant might present a dual universal, efficient and cost-effective candidate vaccine against RV infection.
Collapse
Affiliation(s)
- Atefeh Afchangi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Arash Arashkia
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Farahmand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed Dawood Mousavi Nasab
- Department of Research and Development, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Sayed Mahdi Marashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
27
|
Melnik LI, Guha S, Ghimire J, Smither AR, Beddingfield BJ, Hoffmann AR, Sun L, Ungerleider NA, Baddoo MC, Flemington EK, Gallaher WR, Wimley WC, Garry RF. Ebola virus delta peptide is an enterotoxin. Cell Rep 2022; 38:110172. [PMID: 34986351 DOI: 10.1016/j.celrep.2021.110172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/27/2021] [Accepted: 12/03/2021] [Indexed: 12/21/2022] Open
Abstract
During the 2013-2016 West African (WA) Ebola virus (EBOV) outbreak, severe gastrointestinal symptoms were common in patients and associated with poor outcome. Delta peptide is a conserved product of post-translational processing of the abundant EBOV soluble glycoprotein (sGP). The murine ligated ileal loop model was used to demonstrate that delta peptide is a potent enterotoxin. Dramatic intestinal fluid accumulation follows injection of biologically relevant amounts of delta peptide into ileal loops, along with gross alteration of villous architecture and loss of goblet cells. Transcriptomic analyses show that delta peptide triggers damage response and cell survival pathways and downregulates expression of transporters and exchangers. Induction of diarrhea by delta peptide occurs via cellular damage and regulation of genes that encode proteins involved in fluid secretion. While distinct differences exist between the ileal loop murine model and EBOV infection in humans, these results suggest that delta peptide may contribute to EBOV-induced gastrointestinal pathology.
Collapse
Affiliation(s)
- Lilia I Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Shantanu Guha
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jenisha Ghimire
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Allison R Smither
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Brandon J Beddingfield
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Andrew R Hoffmann
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Leisheng Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | - Melody C Baddoo
- Tulane Cancer Center, Tulane University, New Orleans, LA 70112, USA
| | | | - William R Gallaher
- Department of Microbiology, Immunology and Parasitology, LSU Health Sciences Center, New Orleans, LA 70112, USA; Mockingbird Nature Research Group, Pearl River, LA 70452, USA
| | - William C Wimley
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Robert F Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Zalgen Labs, Germantown, MD 20876, USA.
| |
Collapse
|
28
|
Mandolo JJ, Henrion MYR, Mhango C, Chinyama E, Wachepa R, Kanjerwa O, Malamba-Banda C, Shawa IT, Hungerford D, Kamng’ona AW, Iturriza-Gomara M, Cunliffe NA, Jere KC. Reduction in Severity of All-Cause Gastroenteritis Requiring Hospitalisation in Children Vaccinated against Rotavirus in Malawi. Viruses 2021; 13:2491. [PMID: 34960760 PMCID: PMC8707889 DOI: 10.3390/v13122491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 01/21/2023] Open
Abstract
Rotavirus is the major cause of severe gastroenteritis in children aged <5 years. Introduction of the G1P[8] Rotarix® rotavirus vaccine in Malawi in 2012 has reduced rotavirus-associated hospitalisations and diarrhoeal mortality. However, the impact of rotavirus vaccine on the severity of gastroenteritis presented in children requiring hospitalisation remains unknown. We conducted a hospital-based surveillance study to assess the impact of Rotarix® vaccination on the severity of gastroenteritis presented by Malawian children. Stool samples were collected from children aged <5 years who required hospitalisation with acute gastroenteritis from December 2011 to October 2019. Gastroenteritis severity was determined using Ruuska and Vesikari scores. Rotavirus was detected using enzyme immunoassay. Rotavirus genotypes were determined using nested RT-PCR. Associations between Rotarix® vaccination and gastroenteritis severity were investigated using adjusted linear regression. In total, 3159 children were enrolled. After adjusting for mid-upper arm circumference (MUAC), age, gender and receipt of other vaccines, all-cause gastroenteritis severity scores were 2.21 units lower (p < 0.001) among Rotarix®-vaccinated (n = 2224) compared to Rotarix®-unvaccinated children (n = 935). The reduction in severity score was observed against every rotavirus genotype, although the magnitude was smaller among those infected with G12P[6] compared to the remaining genotypes (p = 0.011). Each one-year increment in age was associated with a decrease of 0.43 severity score (p < 0.001). Our findings provide additional evidence on the impact of Rotarix® in Malawi, lending further support to Malawi's Rotarix® programme.
Collapse
Affiliation(s)
- Jonathan J. Mandolo
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Biomedical Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Marc Y. R. Henrion
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Chimwemwe Mhango
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Biomedical Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - End Chinyama
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
| | - Richard Wachepa
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
| | - Oscar Kanjerwa
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
| | - Chikondi Malamba-Banda
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- Department of Medical Laboratory Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Isaac T. Shawa
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Medical Laboratory Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Daniel Hungerford
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| | - Arox W. Kamng’ona
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Department of Biomedical Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
| | - Miren Iturriza-Gomara
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
- Centre for Vaccine Innovation and Access, Program for Appropriate Technology in Health (PATH), 1218 Geneva, Switzerland
| | - Nigel A. Cunliffe
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| | - Khuzwayo C. Jere
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (J.J.M.); (M.Y.R.H.); (C.M.); (E.C.); (R.W.); (O.K.); (C.M.-B.); (I.T.S.); (A.W.K.)
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- Department of Medical Laboratory Sciences, School of Life Sciences and Health Professions, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| |
Collapse
|
29
|
Patra U, Mukhopadhyay U, Mukherjee A, Dutta S, Chawla-Sarkar M. Treading a HOSTile path: Mapping the dynamic landscape of host cell-rotavirus interactions to explore novel host-directed curative dimensions. Virulence 2021; 12:1022-1062. [PMID: 33818275 PMCID: PMC8023246 DOI: 10.1080/21505594.2021.1903198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/20/2021] [Accepted: 03/10/2021] [Indexed: 12/27/2022] Open
Abstract
Viruses are intracellular pathogens and are dependent on host cellular resources to carry out their cycles of perpetuation. Obtaining an integrative view of host-virus interaction is of utmost importance to understand the complex and dynamic interplay between viral components and host machineries. Besides its obvious scholarly significance, a comprehensive host-virus interaction profile also provides a platform where from host determinants of pro-viral and antiviral importance can be identified and further be subjected to therapeutic intervention. Therefore, adjunct to conventional methods of prophylactic vaccination and virus-directed antivirals, this host-targeted antiviral approach holds promising therapeutic potential. In this review, we present a comprehensive landscape of host cellular reprogramming in response to infection with rotavirus (RV) which causes profuse watery diarrhea in neonates and infants. In addition, an emphasis is given on how host determinants are either usurped or subverted by RV in course of infection and how therapeutic manipulation of specific host factors can effectively modulate the RV life cycle.
Collapse
Affiliation(s)
- Upayan Patra
- Division of Virology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | - Urbi Mukhopadhyay
- Division of Virology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | - Arpita Mukherjee
- Division of Virology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | - Shanta Dutta
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| | - Mamta Chawla-Sarkar
- Division of Virology, National Institute of Cholera and Enteric Diseases, Beliaghata, Kolkata, India
| |
Collapse
|
30
|
Abstract
Group A rotavirus (RVA), one of the leading pathogens causing severe acute gastroenteritis in children and a wide variety of young animals worldwide, induces apoptosis upon infecting cells. Though RVA-induced apoptosis mediated via the dual modulation of its NSP4 and NSP1 proteins is relatively well studied, the nature and signaling pathway(s) involved in RVA-induced necroptosis are yet to be fully elucidated. Here, we demonstrate the nature of RVA-induced necroptosis, the signaling cascade involved, and correlation with RVA-induced apoptosis. Infection with the bovine NCDV and human DS-1 RV strains was shown to activate receptor-interacting protein kinase 1 (RIPK1)/RIPK3/mixed lineage kinase domain-like protein (MLKL), the key necroptosis molecules in virus-infected cells. Using immunoprecipitation assay, RIPK1 was found to bind phosphorylated RIPK3 (pRIPK3) and pMLKL. pMLKL, the major executioner molecule in the necroptotic pathway, was translocated to the plasma membrane of RVA-infected cells to puncture the cell membrane. Interestingly, transfection of RVA NSP4 also induced necroptosis through the RIPK1/RIPK3/MLKL necroptosis pathway. Blockage of each key necroptosis molecule in the RVA-infected or NSP4-transfected cells resulted in decreased necroptosis but increased cell viability and apoptosis, thereby resulting in decreased viral yields in the RVA-infected cells. In contrast, suppression of RVA-induced apoptosis increased necroptosis and virus yields. Our findings suggest that RVA NSP4 also induces necroptosis via the RIPK1/RIPK3/MLKL necroptosis pathway. Moreover, necroptosis and apoptosis-which have proviral and antiviral effects, respectively-exhibited a crosstalk in RVA-infected cells. These findings significantly increase our understanding of the nature of RVA-induced necroptosis and the crosstalk between RVA-induced necroptosis and apoptosis. IMPORTANCE Viral infection usually culminates in cell death through apoptosis, necroptosis, and rarely, pyroptosis. Necroptosis is a form of programmed necrosis that is mediated by signaling complexes of the receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like protein (MLKL). Although apoptosis induction by rotavirus and its NSP4 protein is well known, rotavirus-induced necroptosis is not fully understood. Here, we demonstrate that rotavirus and also its NSP4 protein can induce necroptosis in cultured cells through the activation of the RIPK1/RIPK3/MLKL necroptosis pathway. Moreover, rotavirus-induced necroptosis and apoptosis have opposite effects on viral yield, i.e., they function as proviral and antiviral processes, respectively, and counterbalance each other in rotavirus-infected cells. Our findings provide important insights for understanding the nature of rotavirus-induced necroptosis and the development of novel therapeutic strategies against infection with rotavirus and other RNA viruses.
Collapse
|
31
|
Harada Y, Sekine H, Kubota K, Sadatomi D, Iizuka S, Fujitsuka N. Calcium-activated chloride channel is involved in the onset of diarrhea triggered by EGFR tyrosine kinase inhibitor treatment in rats. Biomed Pharmacother 2021; 141:111860. [PMID: 34246954 DOI: 10.1016/j.biopha.2021.111860] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
EGFR tyrosine kinase inhibitors (TKIs) are mainly used to treat non-small cell lung cancer; however, adverse effects such as severe diarrhea represent a major obstacle towards the continuation of EGFR-TKIs therapy. Chloride channels, which control the fluid flow in the intestinal lumen, are proposed as an important target to remediate EGFR-TKIs-induced diarrhea, but the mechanism remains unclear. The aim of this study was to clarify the mechanism underlying EGFR-TKIs-induced diarrhea with a particular focus on the role of intestinal chloride channels. Here, we show that osimertinib-treated rats exhibit diarrhea and an increase in fecal water content without showing any severe histopathological changes. This diarrhea was attenuated by intraperitoneal treatment with the calcium-activated chloride channel (CaCC) inhibitor CaCCinh-A01. These findings were confirmed in afatinib-treated rats with diarrhea. Moreover, treatment with the Japanese traditional herbal medicine, hangeshashinto (HST), decreased fecal water content and improved fecal appearance in rats treated with EGFR-TKIs. HST inhibited the ionomycin-induced CaCC activation in HEK293 cells in patch-clamp current experiments and its active ingredients were identified. In conclusion, secretory diarrhea induced by treatment with EGFR-TKIs might be partially mediated by the activation of CaCC. Therefore, blocking the CaCC could be a potential new treatment for EGFR-TKI-induced diarrhea.
Collapse
Affiliation(s)
- Yumi Harada
- Tsumura Kampo Research Laboratories, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki 300-1192, Japan
| | - Hitomi Sekine
- Tsumura Kampo Research Laboratories, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki 300-1192, Japan
| | - Kunitsugu Kubota
- Tsumura Kampo Research Laboratories, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki 300-1192, Japan
| | - Daichi Sadatomi
- Tsumura Kampo Research Laboratories, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki 300-1192, Japan
| | - Seiichi Iizuka
- Tsumura Kampo Research Laboratories, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki 300-1192, Japan
| | - Naoki Fujitsuka
- Tsumura Kampo Research Laboratories, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki 300-1192, Japan.
| |
Collapse
|
32
|
Xing J, Zhou X, Fang M, Zhang E, Minze LJ, Zhang Z. DHX15 is required to control RNA virus-induced intestinal inflammation. Cell Rep 2021; 35:109205. [PMID: 34161762 PMCID: PMC8276442 DOI: 10.1016/j.celrep.2021.109205] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/10/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
RNA helicases play critical roles in various biological processes, including serving as viral RNA sensors in innate immunity. Here, we find that RNA helicase DEAH-box helicase 15 (DHX15) is essential for type I interferon (IFN-I, IFN-β), type III IFN (IFN-λ3), and inflammasome-derived cytokine IL-18 production by intestinal epithelial cells (IECs) in response to poly I:C and RNA viruses with preference of enteric RNA viruses, but not DNA virus. Importantly, we generate IEC-specific Dhx15-knockout mice and demonstrate that DHX15 is required for controlling intestinal inflammation induced by enteric RNA virus rotavirus in suckling mice and reovirus in adult mice in vivo, which owes to impaired IFN-β, IFN-λ3, and IL-18 production in IECs from Dhx15-deficient mice. Mechanistically, DHX15 interacts with NLRP6 to trigger NLRP6 inflammasome assembly and activation for inducing IL-18 secretion in IECs. Collectively, our report reveals critical roles for DHX15 in sensing enteric RNA viruses in IECs and controlling intestinal inflammation.
Collapse
Affiliation(s)
- Junji Xing
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Xiaojing Zhou
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Biochemistry, Clinical Medical College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Mingli Fang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Evan Zhang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Laurie J Minze
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Zhiqiang Zhang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
33
|
Crawford SE, Ramani S, Blutt SE, Estes MK. Organoids to Dissect Gastrointestinal Virus-Host Interactions: What Have We Learned? Viruses 2021; 13:999. [PMID: 34071878 PMCID: PMC8230193 DOI: 10.3390/v13060999] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/29/2022] Open
Abstract
Historically, knowledge of human host-enteric pathogen interactions has been elucidated from studies using cancer cells, animal models, clinical data, and occasionally, controlled human infection models. Although much has been learned from these studies, an understanding of the complex interactions between human viruses and the human intestinal epithelium was initially limited by the lack of nontransformed culture systems, which recapitulate the relevant heterogenous cell types that comprise the intestinal villus epithelium. New investigations using multicellular, physiologically active, organotypic cultures produced from intestinal stem cells isolated from biopsies or surgical specimens provide an exciting new avenue for understanding human specific pathogens and revealing previously unknown host-microbe interactions that affect replication and outcomes of human infections. Here, we summarize recent biologic discoveries using human intestinal organoids and human enteric viral pathogens.
Collapse
Affiliation(s)
- Sue E. Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (S.E.C.); (S.R.); (S.E.B.)
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (S.E.C.); (S.R.); (S.E.B.)
| | - Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (S.E.C.); (S.R.); (S.E.B.)
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (S.E.C.); (S.R.); (S.E.B.)
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
34
|
Khalkhali P, Khavandegar A, Mozhgani SH, Teimoori A, Moradi A, Ajorloo M, Lorestani N, Kaveh K, Akbar S, Razavi Nikoo H. Genotyping and sequence characterization of the NSP4 gene of human group A rotavirus strains in Northern Iran. J Med Virol 2021; 93:4824-4830. [PMID: 33818782 DOI: 10.1002/jmv.26998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 11/12/2022]
Abstract
Rotavirus is known to be responsible for remarkable numbers of severe diarrheal episodes and even death in infants and young children. In this study, we aimed to survey genetic diversity and variation analysis of viroporin, which is encoded by the rotavirus NSP4 segment. Thirty-five rotavirus-positive specimens were obtained, and RNA extraction and polymerase chain reaction amplification were performed. After the sequencing process, four specimens were excluded, and the final 31 samples remained for genetic diversity and variation analysis. The predominant single G/P combination was G1P[8] (~78%), followed by G2P[8] (~13%), and equal percentages (3%) of G2P[4], G3P[8], and G-non-typeable-P[8]. Further analyses revealed that variations could be found in the three regions of NSP4, including VP4 binding site (aa 112-146), double-layered particle binding site (aa 161-175), and finally, in the predicted amphipathic alpha-helix. Phylogenic tree analysis demonstrated that the mentioned samples clustered with genotype E1 and E2 reference sequences. As previously reported in the literature, in this study, it was revealed that no apparent correlation exists in the deduced amino acid sequences corresponding to this region between the rotaviruses collected from patients with and without diarrhea.
Collapse
Affiliation(s)
- Parinaz Khalkhali
- International Branch, Golestan University of Medical Sciences, Gorgan, Iran
| | - Armin Khavandegar
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Sayed-Hamidreza Mozhgani
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Ali Teimoori
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolvahhab Moradi
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehdi Ajorloo
- Department of Clinical Laboratory Sciences, School of Allied Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.,Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Nazanin Lorestani
- Department of Microbiology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Kimia Kaveh
- International Branch, Golestan University of Medical Sciences, Gorgan, Iran
| | - Soroush Akbar
- Department of Biochemistry and Biophysics, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hadi Razavi Nikoo
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.,Infectious Disease Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
35
|
Rotavirus in Calves and Its Zoonotic Importance. Vet Med Int 2021; 2021:6639701. [PMID: 33968359 PMCID: PMC8081619 DOI: 10.1155/2021/6639701] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 12/24/2022] Open
Abstract
Rotavirus is a major pathogen responsible for diarrheal disease in calves, resulting in loss of productivity and economy of farmers. However, various facets of diarrheal disease caused by rotavirus in calves in the world are inadequately understood, considering that diarrheal disease caused by rotavirus is a vital health problem in calves that interrupts production benefits with reduced weight gain and increased mortality, and its potential for zoonotic spread. The pathological changes made by rotavirus are almost exclusively limited to the small intestine that leads to diarrhea. It is environmentally distributed worldwide and was extensively studied. Reassortment is one of the important mechanisms for generating genetic diversity of rotaviruses and eventually for viral evolution. So, the primary strategy is to reduce the burden of rotavirus infections by practicing early colostrum's feeding in newborn calves, using vaccine, and improving livestock management. Rotaviruses have a wide host range, infecting many animal species as well as humans. As it was found that certain animal rotavirus strains had antigenic similarities to some human strains, this may be an indication for an animal to play a role as a source of rotavirus infection in humans. Groups A to C have been shown to infect both humans and animals. The most commonly detected strains in both human and animals are G2, G3, G4, and G9, P [6]. Therefore, this review was made to get overview epidemiology status and zoonotic importance of bovine rotavirus.
Collapse
|
36
|
Liu C, Huang P, Zhao D, Xia M, Zhong W, Jiang X, Tan M. Effects of rotavirus NSP4 protein on the immune response and protection of the S R69A-VP8* nanoparticle rotavirus vaccine. Vaccine 2021; 39:263-271. [PMID: 33309483 PMCID: PMC7822095 DOI: 10.1016/j.vaccine.2020.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/05/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022]
Abstract
Rotavirus causes severe diarrhea and dehydration in young children. Even with the implementation of the current live vaccines, rotavirus infections still cause significant mortality and morbidity, indicating a need for new rotavirus vaccines with improved efficacy. To this end, we have developed an SR69A-VP8*/S60-VP8* nanoparticle rotavirus vaccine candidate that will be delivered parenterally with Alum adjuvant. In this study, as parts of our further development of this nanoparticle vaccine, we evaluated 1) roles of rotavirus nonstructural protein 4 (NSP4) that is the rotavirus enterotoxin, a possible vaccine target, and an immune stimulator, and 2) effects of CpG adjuvant that is a toll-like receptor 9 (TLR9) ligand and agonist on the immune response and protection of our SR69A-VP8*/S60-VP8* nanoparticle vaccine. The resulted vaccine candidates were examined for their IgG responses in mice. In addition, the resulted mouse sera were assessed for i) blocking titers against interactions of rotavirus VP8* proteins with their glycan ligands, ii) neutralization titers against rotavirus replication in cell culture, and iii) passive protection against rotavirus challenge with diarrhea in suckling mice. Our data showed that the Alum adjuvant appeared to work better with the SR69A-VP8*/S60-VP8* nanoparticles than the CpG adjuvant, while an addition of the NSP4 antigen to the SR69A-VP8*/S60-VP8* vaccine may not help to further increase the immune response and protection of the resulted vaccine.
Collapse
Affiliation(s)
- Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, Yunnan Province, China
| | - Pengwei Huang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Dandan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Ming Xia
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Weiming Zhong
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xi Jiang
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ming Tan
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
37
|
Li Y, Xia S, Jiang X, Feng C, Gong S, Ma J, Fang Z, Yin J, Yin Y. Gut Microbiota and Diarrhea: An Updated Review. Front Cell Infect Microbiol 2021; 11:625210. [PMID: 33937093 PMCID: PMC8082445 DOI: 10.3389/fcimb.2021.625210] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/26/2021] [Indexed: 12/15/2022] Open
Abstract
Diarrhea is a common problem to the whole world and the occurrence of diarrhea is highly associated with gut microbiota, such as bacteria, fungi, and viruses. Generally, diarrheal patients or animals are characterized by gut microbiota dysbiosis and pathogen infections may lead to diarrheal phenotypes. Of relevance, reprograming gut microbiota communities by dietary probiotics or fecal bacteria transplantation are widely introduced to treat or prevent diarrhea. In this review, we discussed the influence of the gut microbiota in the infection of diarrhea pathogens, and updated the research of reshaping the gut microbiota to prevent or treat diarrhea for the past few years. Together, gut microbiota manipulation is of great significance to the prevention and treatment of diarrhea, and further insight into the function of the gut microbiota will help to discover more anti-diarrhea probiotics.
Collapse
Affiliation(s)
- Yunxia Li
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Siting Xia
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xiaohan Jiang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Can Feng
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Saiming Gong
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jie Ma
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Jie Yin, ; Zhengfeng Fang,
| | - Jie Yin
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- *Correspondence: Jie Yin, ; Zhengfeng Fang,
| | - Yulong Yin
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
38
|
Non-structural Enterotoxin (NSP4) Gene based Molecular Characterization of Caprine and Ovine Rotavirus A, India. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2020. [DOI: 10.22207/jpam.14.4.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rotavirus A (RVA) causes viral gastroenteritis in humans and animals, including calves, piglets, and foals. The current study reports the genetic characterization of the full-length enterotoxin gene, NSP4, from caprine and ovine species. Upon characterizing eight full-length NSP4 genes by sequencing, it was found that the four caprine and three ovine RVAs NSP4 genes are of E2 genotype and the sole ovine RVA isolate was found to be of E1 genotype. In the sequence and phyloanalysis of the NSP4 gene the seven E2 genotypes clustered with bovine, human, and caprine isolates from India and Bangladesh, respectively. The E1 genotype of ovine RVA was closer to human RVA isolate from India. The nucleotide per cent identity analysis revealed that all E2 genotype strains of caprine and ovine species ranged from 88.4% to 90.4% and it was found common to both the reference human RVA isolates DS-1 and AU-1. Whereas, the E1 genotype ovine strain clustered with human RVA isolates with 93.1% nucleotide per cent identity. The RVA strains circulating in caprine and ovine populations may share a common origin which is usually found in artiodactyl species because humans share a common dwelling with animals. Future studies are needed to confirm these findings of their relationship with humans and large animals.
Collapse
|
39
|
Chang-Graham AL, Perry JL, Engevik MA, Engevik KA, Scribano FJ, Gebert JT, Danhof HA, Nelson JC, Kellen JS, Strtak AC, Sastri NP, Estes MK, Britton RA, Versalovic J, Hyser JM. Rotavirus induces intercellular calcium waves through ADP signaling. Science 2020; 370:370/6519/eabc3621. [PMID: 33214249 PMCID: PMC7957961 DOI: 10.1126/science.abc3621] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/16/2020] [Indexed: 01/14/2023]
Abstract
Rotavirus causes severe diarrheal disease in children by broadly dysregulating intestinal homeostasis. However, the underlying mechanism(s) of rotavirus-induced dysregulation remains unclear. We found that rotavirus-infected cells produce paracrine signals that manifested as intercellular calcium waves (ICWs), observed in cell lines and human intestinal enteroids. Rotavirus ICWs were caused by the release of extracellular adenosine 5'-diphosphate (ADP) that activated P2Y1 purinergic receptors on neighboring cells. ICWs were blocked by P2Y1 antagonists or CRISPR-Cas9 knockout of the P2Y1 receptor. Blocking the ADP signal reduced rotavirus replication, inhibited rotavirus-induced serotonin release and fluid secretion, and reduced diarrhea severity in neonatal mice. Thus, rotavirus exploited paracrine purinergic signaling to generate ICWs that amplified the dysregulation of host cells and altered gastrointestinal physiology to cause diarrhea.
Collapse
Affiliation(s)
- Alexandra L. Chang-Graham
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Jacob L. Perry
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Melinda A. Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, USA,Department of Pathology, Texas Children’s Hospital, USA
| | - Kristen A. Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Francesca J. Scribano
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - J. Thomas Gebert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Heather A. Danhof
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Joel C. Nelson
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA
| | - Joseph S. Kellen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Alicia C. Strtak
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Narayan P. Sastri
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA,Department of Medicine, Gastroenterology and Hepatology, Baylor College of Medicine, USA
| | - Robert A. Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, USA,Department of Pathology, Texas Children’s Hospital, USA
| | - Joseph M. Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA,Corresponding author. Correspondence and requests for materials should be addressed to J.H.
| |
Collapse
|
40
|
Sun S, Lin X, Yang Y, Cen J, Luo F, Chen X. Acupoint application for rotavirus diarrhea in infants and children: A protocol for systematic review and meta analysis. Medicine (Baltimore) 2020; 99:e22227. [PMID: 32957362 PMCID: PMC7505364 DOI: 10.1097/md.0000000000022227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Diarrheal disease currently claims the lives of approximately 500,000 children each year. Rotaviruses are the pathogens primarily responsible for more severe cases and more than one-third of diarrhea-associated deaths in children under 5 years old globally. At present, commonly used drug therapies for rotavirus diarrhea in Western medicine, such as oral rehydration salts, montmorillonite, probiotics, and nitazoxanide, often cannot achieve satisfactory curative effects. Moreover, infants' and children's compliance with drugs and injections is often lower than their compliance with acupoint application therapy. A large number of studies have shown that acupoint application can increase the clinical cure rate and shorten the duration of diarrhea. However, there is a lack of systematic reviews on the safety and efficacy of acupoint application in the treatment of rotavirus diarrhea. Therefore, we will conduct a study to evaluate the safety and efficacy of acupoint application for rotavirus diarrhea in infants and children. METHODS We will search the relevant medical literature using PubMed, EMBASE, Web of Science, Cochrane CENTRAL, China National Knowledge Infrastructure, the Wanfang Database, the Chinese Biomedical Literature Database, and the Chinese Scientific Journal Database from inception to August 2020. Both MeSH and free text terms will be utilized to obtain the maximum numbers of papers. No language restrictions will be applied, and the publication type will be limited to randomized controlled trials. Two teams will independently review and assess the studies for inclusion in the review. RevMan V 5.0 software will be applied for data extraction. The methodological quality of the included studies will be evaluated according to the Cochrane Handbook. RESULTS The results of this study will be published in a peer-reviewed journal. CONCLUSION The conclusion of this systematic review will provide evidence regarding whether acupoint application is an effective intervention for infants and children with rotavirus diarrhea. INPLASY REGISTRATION NUMBER INPLASY202070123.
Collapse
Affiliation(s)
- Shaodan Sun
- Department of Pediatrics, The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine
| | - Xiaojie Lin
- Department of Medical Education Office, Guangdong, Provincial Hospital of Traditional Chinese Medicine, Guangzhou
| | - Yang Yang
- Department of Rehabilitation, Affiliated Jiangmen Traditional Chinese Medicine Hospital of Ji’nan University, Ji’nan University, Jiangmen
| | - Jingtu Cen
- Department of Otolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou
| | - Fei Luo
- Department of Pediatrics, The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Xiaogang Chen
- Department of Pediatrics, The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine
- Department of Pediatrics, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
41
|
Engevik MA, Banks LD, Engevik KA, Chang-Graham AL, Perry JL, Hutchinson DS, Ajami NJ, Petrosino JF, Hyser JM. Rotavirus infection induces glycan availability to promote ileum-specific changes in the microbiome aiding rotavirus virulence. Gut Microbes 2020; 11:1324-1347. [PMID: 32404017 PMCID: PMC7524290 DOI: 10.1080/19490976.2020.1754714] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Multiple studies have identified changes within the gut microbiome in response to diarrheal-inducing bacterial pathogens. However, examination of the microbiome in response to viral pathogens remains understudied. Compounding this, many studies use fecal samples to assess microbiome composition; which may not accurately mirror changes within the small intestine, the primary site for most enteric virus infections. As a result, the functional significance of small intestinal microbiome shifts during infection is not well defined. To address these gaps, rotavirus-infected neonatal mice were examined for changes in bacterial community dynamics, host gene expression, and tissue recovery during infection. Profiling bacterial communities using 16S rRNA sequencing suggested significant and distinct changes in ileal communities in response to rotavirus infection, with no significant changes for other gastrointestinal (GI) compartments. At 1-d post-infection, we observed a loss in Lactobacillus species from the ileum, but an increase in Bacteroides and Akkermansia, both of which exhibit mucin-digesting capabilities. Concomitant with the bacterial community shifts, we observed a loss of mucin-filled goblet cells in the small intestine at d 1, with recovery occurring by d 3. Rotavirus infection of mucin-producing cell lines and human intestinal enteroids (HIEs) stimulated release of stored mucin granules, similar to in vivo findings. In vitro, incubation of mucins with Bacteroides or Akkermansia members resulted in significant glycan degradation, which altered the binding capacity of rotavirus in silico and in vitro. Taken together, these data suggest that the response to and recovery from rotavirus-diarrhea is unique between sub-compartments of the GI tract and may be influenced by mucin-degrading microbes.
Collapse
Affiliation(s)
- Melinda A. Engevik
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA,Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Lori D. Banks
- Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kristen A. Engevik
- Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Alexandra L. Chang-Graham
- Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Jacob L. Perry
- Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Diane S. Hutchinson
- Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Nadim J. Ajami
- Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph F. Petrosino
- Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph M. Hyser
- Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, TX, USA,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA,CONTACT Joseph M. Hyser 1 Baylor Plaza, HoustonTX77030, USA
| |
Collapse
|
42
|
Roth AN, Helm EW, Mirabelli C, Kirsche E, Smith JC, Eurell LB, Ghosh S, Altan-Bonnet N, Wobus CE, Karst SM. Norovirus infection causes acute self-resolving diarrhea in wild-type neonatal mice. Nat Commun 2020; 11:2968. [PMID: 32528015 PMCID: PMC7289885 DOI: 10.1038/s41467-020-16798-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 05/25/2020] [Indexed: 01/08/2023] Open
Abstract
Human noroviruses are the leading cause of severe childhood diarrhea worldwide, yet we know little about their pathogenic mechanisms. Murine noroviruses cause diarrhea in interferon-deficient adult mice but these hosts also develop systemic pathology and lethality, reducing confidence in the translatability of findings to human norovirus disease. Herein we report that a murine norovirus causes self-resolving diarrhea in the absence of systemic disease in wild-type neonatal mice, thus mirroring the key features of human norovirus disease and representing a norovirus small animal disease model in wild-type mice. Intriguingly, lymphocytes are critical for controlling acute norovirus replication while simultaneously contributing to disease severity, likely reflecting their dual role as targets of viral infection and key components of the host response.
Collapse
Affiliation(s)
- Alexa N Roth
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Emily W Helm
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Carmen Mirabelli
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Erin Kirsche
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jonathan C Smith
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Laura B Eurell
- Office of Research, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Sourish Ghosh
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie M Karst
- Department of Molecular Genetics & Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
43
|
Kumar D, Singh A, Kumar P, Uversky VN, Rao CD, Giri R. Understanding the penetrance of intrinsic protein disorder in rotavirus proteome. Int J Biol Macromol 2020; 144:892-908. [PMID: 31739058 PMCID: PMC7112477 DOI: 10.1016/j.ijbiomac.2019.09.166] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 01/03/2023]
Abstract
Rotavirus is a major cause of severe acute gastroenteritis in the infants and young children. The past decade has evidenced the role of intrinsically disordered proteins/regions (IDPs)/(IDPRs) in viral and other diseases. In general, (IDPs)/(IDPRs) are considered as dynamic conformational ensembles that devoid of a specific 3D structure, being associated with various important biological phenomena. Viruses utilize IDPs/IDPRs to survive in harsh environments, to evade the host immune system, and to highjack and manipulate host cellular proteins. The role of IDPs/IDPRs in Rotavirus biology and pathogenicity are not assessed so far, therefore, we have designed this study to deeply look at the penetrance of intrinsic disorder in rotavirus proteome consisting 12 proteins encoded by 11 segments of viral genome. Also, for all human rotaviral proteins, we have deciphered molecular recognition features (MoRFs), which are disorder based binding sites in proteins. Our study shows the wide spread of intrinsic disorder in several rotavirus proteins, primarily the nonstructural proteins NSP3, NSP4, and NSP5 that are involved in viral replication, translation, viroplasm formation and/or maturation. This study may serve as a primer for understanding the role of IDPs/MoRFs in rotavirus biology, design of alternative therapeutic strategies, and development of disorder-based drugs.
Collapse
Affiliation(s)
- Deepak Kumar
- Indian Institute of Technology Mandi, VPO Kamand, Himachal Pradesh 175005, India
| | - Ankur Singh
- Indian Institute of Technology Mandi, VPO Kamand, Himachal Pradesh 175005, India
| | - Prateek Kumar
- Indian Institute of Technology Mandi, VPO Kamand, Himachal Pradesh 175005, India
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - C Durga Rao
- SRM University, AP - Amaravati, Neerukonda, Mangalagiri Mandal Guntur District, Mangalagiri, Andhra Pradesh 522502, India.
| | - Rajanish Giri
- Indian Institute of Technology Mandi, VPO Kamand, Himachal Pradesh 175005, India; BioX Center, Indian Institute of Technology Mandi, Himachal Pradesh, India.
| |
Collapse
|
44
|
Yu B, Zhu X, Yang X, Jin L, Xu J, Ma T, Yang H. Plumbagin Prevents Secretory Diarrhea by Inhibiting CaCC and CFTR Channel Activities. Front Pharmacol 2019; 10:1181. [PMID: 31649543 PMCID: PMC6795057 DOI: 10.3389/fphar.2019.01181] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022] Open
Abstract
Secretory diarrhea, which primarily originates through intestinal pathogens and viruses, is a health burden in many regions worldwide. Enterocyte Cl− channels, as the final step in enterotoxin-induced fluid secretion, constitute an attractive class of targets for diarrhea therapy. Chloride channel inhibitors have become a new class of candidates for antisecretion and anti-intestinal motility agents. In the present study, we identified plumbagin as a transmembrane protein 16A (TMEM16A) inhibitor in a cell-based fluorescence-quenching assay, and the IC50 value was ∼12.46 µM. Short-circuit current measurements showed that plumbagin reversibly inhibited the Eact-induced Cl− current on the apical side of TMEM16A-transfected Fischer rat thyroid (FRT) cells with no significant effect on cytoplasmic Ca2+ signaling. Notably, plumbagin also inhibited the activity of intestinal epithelial calcium-activated chloride channel (CaCC) and cystic fibrosis transmembrane conductance regulator (CFTR) in both HT-29 cells and mouse colons, but had no effects on the activity of the Na+-K+ ATPase or K+ channels. In in vivo experiments, the administration of plumbagin reduced both Escherichia coli heat-stable enterotoxin (STa)- and cholera toxin (CT)-induced intestinal fluid secretion. In neonatal mouse models of CT- and rotavirus infection-induced diarrhea, 0.4 µg plumbagin inhibited secretory diarrhea by >40% and 50%, respectively, without affecting intestinal epithelial integrity or the rotaviral infection. In addition, plumbagin exerted inhibitory effects on the vasoactive intestinal peptide (VIP)-, prostaglandin E2 (PGE2)-, and 5-hydroxytryptamine (5-HT)-stimulated Cl− currents. In the evaluations of intestinal motility, plumbagin significantly delayed intestinal motility and inhibited intestinal smooth muscle contractility without an evident impact on contractive frequency. Collectively, our results indicate that plumbagin inhibits both Ca2+- and cAMP-activated Cl− channels, accounting for the mechanisms of plumbagin inhibition of chloride secretion and intestinal motility. Thus, plumbagin can be a lead compound in the treatment of CT-induced, Traveler’s, and rotaviral diarrhea, as well as other types of secretory diarrhea that result from excessive intestinal fluid secretion and increased intestinal peristalsis.
Collapse
Affiliation(s)
- Bo Yu
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Xiaojuan Zhu
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Xinyu Yang
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Lingling Jin
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jia Xu
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Tonghui Ma
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Hong Yang
- School of Life Sciences, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| |
Collapse
|
45
|
Kunzelmann K, Centeio R, Wanitchakool P, Cabrita I, Benedetto R, Saha T, Hoque KM, Schreiber R. Control of Ion Transport by Tmem16a Expressed in Murine Intestine. Front Physiol 2019; 10:1262. [PMID: 31680994 PMCID: PMC6797858 DOI: 10.3389/fphys.2019.01262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022] Open
Abstract
Cl– secretion by the human and murine intestinal epithelium occurs through the cystic fibrosis transmembrane conductance regulator (cftr). However, the Ca2+ activated Cl– channel Tmem16a was shown to contribute to Cl– secretion, mainly, but not exclusively, as a basolaterally located Cl– channel that controls basolateral Ca2+ signaling, and thus activation of basolateral Ca2+ dependent Sk4 K+ channels. In intestinal goblet cells, Tmem16a was shown to regulated Ca2+ signals required for exocytosis of mucus. Because a recent report denied the existence and functional role of Tmem16a in murine intestine, we reexamined in detail expression of mRNA and protein for Tmem16a in mouse colon. In experiments using short-circuited Ussing chamber and whole cell patch-clamp techniques, we further compared ion transport in wild type (WT) colon with that in mice with intestinal epithelial specific knockout of Tmem16a. As reported earlier we fully confirm expression of Tmem16a in colonic epithelial cells and the role of Tmem16a for both Ca2+-dependent and cAMP-regulated ion secretion.
Collapse
Affiliation(s)
- Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Raquel Centeio
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | | | - Inês Cabrita
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Roberta Benedetto
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| | - Tultul Saha
- Division of Pathophysiology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Kazi Mirajul Hoque
- Division of Pathophysiology, National Institute of Cholera and Enteric Diseases, Kolkata, India.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Regensburg, Germany
| |
Collapse
|
46
|
Rotavirus Calcium Dysregulation Manifests as Dynamic Calcium Signaling in the Cytoplasm and Endoplasmic Reticulum. Sci Rep 2019; 9:10822. [PMID: 31346185 PMCID: PMC6658527 DOI: 10.1038/s41598-019-46856-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/04/2019] [Indexed: 02/08/2023] Open
Abstract
Like many viruses, rotavirus (RV) dysregulates calcium homeostasis by elevating cytosolic calcium ([Ca2+]cyt) and decreasing endoplasmic reticulum (ER) stores. While an overall, monophasic increase in [Ca2+]cyt during RV infection has been shown, the nature of the RV-induced aberrant calcium signals and how they manifest over time at the single-cell level have not been characterized. Thus, we generated cell lines and human intestinal enteroids (HIEs) stably expressing cytosolic and/or ER-targeted genetically-encoded calcium indicators to characterize calcium signaling throughout RV infection by time-lapse imaging. We found that RV induces highly dynamic [Ca2+]cyt signaling that manifest as hundreds of discrete [Ca2+]cyt spikes, which increase during peak infection. Knockdown of nonstructural protein 4 (NSP4) attenuates the [Ca2+]cyt spikes, consistent with its role in dysregulating calcium homeostasis. RV-induced [Ca2+]cyt spikes were primarily from ER calcium release and were attenuated by inhibiting the store-operated calcium entry (SOCE) channel Orai1. RV-infected HIEs also exhibited prominent [Ca2+]cyt spikes that were attenuated by inhibiting SOCE, underlining the relevance of these [Ca2+]cyt spikes to gastrointestinal physiology and role of SOCE in RV pathophysiology. Thus, our discovery that RV increases [Ca2+]cyt by dynamic calcium signaling, establishes a new, paradigm-shifting understanding of the spatial and temporal complexity of virus-induced calcium signaling.
Collapse
|
47
|
Lewnard JA, Lopman BA, Parashar UD, Bennett A, Bar-Zeev N, Cunliffe NA, Samuel P, Guerrero ML, Ruiz-Palacios G, Kang G, Pitzer VE. Heterogeneous susceptibility to rotavirus infection and gastroenteritis in two birth cohort studies: Parameter estimation and epidemiological implications. PLoS Comput Biol 2019; 15:e1007014. [PMID: 31348775 PMCID: PMC6690553 DOI: 10.1371/journal.pcbi.1007014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 08/12/2019] [Accepted: 04/09/2019] [Indexed: 11/19/2022] Open
Abstract
Cohort studies, randomized trials, and post-licensure studies have reported reduced natural and vaccine-derived protection against rotavirus gastroenteritis (RVGE) in low- and middle-income countries. While susceptibility of children to rotavirus is known to vary within and between settings, implications for estimation of immune protection are not well understood. We sought to re-estimate naturally-acquired protection against rotavirus infection and RVGE, and to understand how differences in susceptibility among children impacted estimates. We re-analyzed data from studies conducted in Mexico City, Mexico and Vellore, India. Cumulatively, 573 rotavirus-unvaccinated children experienced 1418 rotavirus infections and 371 episodes of RVGE over 17,636 child-months. We developed a model that characterized susceptibility to rotavirus infection and RVGE among children, accounting for aspects of the natural history of rotavirus and differences in transmission rates between settings. We tested whether model-generated susceptibility measurements were associated with demographic and anthropometric factors, and with the severity of RVGE symptoms. We identified greater variation in susceptibility to rotavirus infection and RVGE in Vellore than in Mexico City. In both cohorts, susceptibility to rotavirus infection and RVGE were associated with male sex, lower birth weight, lower maternal education, and having fewer siblings; within Vellore, susceptibility was also associated with lower socioeconomic status. Children who were more susceptible to rotavirus also experienced higher rates of rotavirus-negative diarrhea, and higher risk of moderate-to-severe symptoms when experiencing RVGE. Simulations suggested that discrepant estimates of naturally-acquired immunity against RVGE can be attributed, in part, to between-setting differences in susceptibility of children, but result primarily from the interaction of transmission rates with age-dependent risk for infections to cause RVGE. We found that more children in Vellore than in Mexico City belong to a high-risk group for rotavirus infection and RVGE, and demonstrate that unmeasured individual- and age-dependent susceptibility may influence estimates of naturally-acquired immune protection against RVGE.
Collapse
Affiliation(s)
- Joseph A. Lewnard
- Division of Epidemiology and Biostatistics, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Benjamin A. Lopman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States of America
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Umesh D. Parashar
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Aisleen Bennett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Center for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, University of Liverpool, Liverpool, United Kingdom
| | - Naor Bar-Zeev
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Center for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, University of Liverpool, Liverpool, United Kingdom
- International Vaccine Access Center, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Nigel A. Cunliffe
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, College of Medicine, University of Malawi, Blantyre, Malawi
- Center for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, University of Liverpool, Liverpool, United Kingdom
| | - Prasanna Samuel
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - M. Lourdes Guerrero
- Instituto Nacional de Ciences Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Gagandeep Kang
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Virginia E. Pitzer
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| |
Collapse
|
48
|
Astrovirus and the microbiome. Curr Opin Virol 2019; 37:10-15. [PMID: 31163291 DOI: 10.1016/j.coviro.2019.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/26/2019] [Accepted: 05/02/2019] [Indexed: 11/22/2022]
Abstract
Although astroviruses are most commonly associated with acute gastrointestinal illness in humans, their ability to infect a broad range of hosts and cause a spectrum of disease makes them widespread and complex pathogens. The precise mechanisms that dictate the course of astrovirus disease have not been studied extensively but are likely driven by multifactorial host-microbe interactions. Recent insights from studies of animal astrovirus infections have revealed both beneficial and detrimental effects for the host. However, further in-depth studies are needed to fully explore the consequences of astrovirus-induced changes in the gut microenvironment as well as the role of the microbiota in astrovirus infection.
Collapse
|
49
|
Tamim S, Matthijnssens J, Heylen E, Zeller M, Van Ranst M, Salman M, Hasan F. Evidence of zoonotic transmission of VP6 and NSP4 genes into human species A rotaviruses isolated in Pakistan in 2010. Arch Virol 2019; 164:1781-1791. [PMID: 31079214 DOI: 10.1007/s00705-019-04271-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/30/2019] [Indexed: 10/26/2022]
Abstract
Introduction of animal group A rotavirus (RVA) gene segments into the human RVA population is a major factor shaping the genetic landscape of human RVA strains. The VP6 and NSP4 genes of 74 G/P-genotyped RVA isolates collected in Rawalpindi during 2010 were analyzed, revealing the presence of VP6 genotypes I1 (60.8%) and I2 (39.2%) and NSP4 genotypes E1 (60.8%), E2 (28.3%) and E-untypable (10.8%) among the circulating human RVA strains. The typical human RVA combinations I1E1 and I2E2 were found in 59.4% and 24.3% of the cases, respectively, whereas 5.4% of the RVA strains were reassortants, i.e., either I1E2 or I2E1. The phylogeny of the NSP4 gene showed that one G2P[4] and two G1P[6] RVA strains clustered with porcine E1 RVA strains or RVA strains that were considered to be (partially) of porcine origin. In addition, the NSP4 gene segment of the unusual human G6P[1] RVA strains clustered closely with bovine E2 RVA strains, further strengthening the hypothesis of an interspecies transmission event. The study further demonstrates the role of genomic re-assortment and the involvement of interspecies transmission in the evolution of human RVA strains. The VP6 and NSP4 nucleotide sequences analyzed in the study received the GenBank accession numbers KC846908- KC846971 and KC846972-KC847037, respectively.
Collapse
Affiliation(s)
- Sana Tamim
- Public Health Laboratories Division, Department of Virology/Immunology, National Institute of Health, Islamabad, Pakistan.
| | - Jelle Matthijnssens
- Laboratory of Viral Metagenomics, Rega Institute, Herestraat 49 box 1040, 3000, Leuven, Belgium
| | - Elisabeth Heylen
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Mark Zeller
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Marc Van Ranst
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Leuven, Belgium
| | - Muhammad Salman
- Public Health Laboratories Division, Department of Virology/Immunology, National Institute of Health, Islamabad, Pakistan
| | - Fariha Hasan
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
50
|
Rotavirus-Induced Early Activation of the RhoA/ROCK/MLC Signaling Pathway Mediates the Disruption of Tight Junctions in Polarized MDCK Cells. Sci Rep 2018; 8:13931. [PMID: 30224682 PMCID: PMC6141481 DOI: 10.1038/s41598-018-32352-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/06/2018] [Indexed: 02/02/2023] Open
Abstract
Intestinal epithelial tight junctions (TJ) are a major barrier restricting the entry of various harmful factors including pathogens; however, they also represent an important entry portal for pathogens. Although the rotavirus-induced early disruption of TJ integrity and targeting of TJ proteins as coreceptors are well-defined, the precise molecular mechanisms involved remain unknown. In the present study, infection of polarized MDCK cells with the species A rotavirus (RVA) strains human DS-1 and bovine NCDV induced a redistribution of TJ proteins into the cytoplasm, a reversible decrease in transepithelial resistance, and an increase in paracellular permeability. RhoA/ROCK/MLC signaling was identified as activated at an early stage of infection, while inhibition of this pathway prevented the rotavirus-induced early disruption of TJ integrity and alteration of TJ protein distribution. Activation of pMYPT, PKC, or MLCK, which are known to participate in TJ dissociation, was not observed in MDCK cells infected with either rotavirus strain. Our data demonstrated that binding of RVA virions or cogent VP8* proteins to cellular receptors activates RhoA/ROCK/MLC signaling, which alters TJ protein distribution and disrupts TJ integrity via contraction of the perijunctional actomyosin ring, facilitating virion access to coreceptors and entry into cells.
Collapse
|