1
|
Chen Y, Luo G, Song F, Wang X, Zhang S, Ge S, Li T, Zhang J, Xia N. Truncated rotavirus VP4 proteins induce stronger protective immunity compared to P2 - VP8 in animal models. Antiviral Res 2025; 238:106156. [PMID: 40194664 DOI: 10.1016/j.antiviral.2025.106156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/17/2025] [Accepted: 04/05/2025] [Indexed: 04/09/2025]
Abstract
Group A rotavirus (RVA) is the primary causative agent of acute gastroenteritis (AGE) in children under five years of age, resulting in over 120,000 deaths annually. In previous studies, we identified truncated VP4∗ as a potentially more promising vaccine candidate compared to VP8∗ and VP5∗. This study aimed to compare the immunogenicity and protective efficacy of VP4∗ and P2-VP8, the most advanced recombinant rotavirus vaccine undergoing phase 3 clinical trial in various animal models, including mice, guinea pigs, rabbits, and piglets. The results indicated that the binding antibodies and neutralizing antibodies induced by VP4∗ were significantly higher levels compared to P2-VP8. Immunization with VP4∗ provided 100 % protection for mice against challenges with EDIM and LLR strains. Additionally, we were intrigued to discover that the VP4∗ antibody not only inhibited virus adsorption but also prevented the virus from entering cells following pre-adsorption. In summary, VP4∗ demonstrates greater immunogenicity and protective efficacy compared to P2-VP8, making it a more promising candidate antigen for recombinant rotavirus vaccines.
Collapse
MESH Headings
- Animals
- Rotavirus Infections/prevention & control
- Rotavirus Infections/immunology
- Rotavirus Infections/virology
- Capsid Proteins/immunology
- Capsid Proteins/genetics
- Rotavirus Vaccines/immunology
- Rotavirus Vaccines/administration & dosage
- Rotavirus Vaccines/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Rotavirus/immunology
- Rotavirus/genetics
- Mice
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Disease Models, Animal
- Rabbits
- Guinea Pigs
- Swine
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Immunogenicity, Vaccine
- Mice, Inbred BALB C
- Antigens, Viral/immunology
- Antigens, Viral/genetics
- Female
Collapse
Affiliation(s)
- Yaling Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Guoxing Luo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China; Novel Product R&D Department, Xiamen Innovax Biotech Co., Ltd., Xiamen, 361022, Fujian, China
| | - Feibo Song
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xuechun Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Shiyin Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Shengxiang Ge
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China.
| | - Tingdong Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China.
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University. Xiamen, 361102, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
2
|
Alhazmi H, Alzahrani A, Alshaikh S, Azzhary L, Alhaddad F, Alshamrani Z, Alwagdani R. The Significance of Inflammatory Markers in Pediatric Patients with Acute Gastroenteritis Presenting to the Emergency Department. CHILDREN (BASEL, SWITZERLAND) 2025; 12:617. [PMID: 40426796 DOI: 10.3390/children12050617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Background: Considerable mortality and morbidity rates linked to AGE are well documented in the literature. Many inflammatory markers have been studied in the context of research on AGE as tools to predict the clinical course of the disease and determine the need for the use of antimicrobials. This study focuses on CRP, PCT, and WBC counts as inflammatory markers of AGE. Methods: A retrospective chart review study was conducted at King Abdullah Specialized Children's Hospital, Jeddah. Using a non-probability consecutive sampling technique, all patients under the age of 14 diagnosed with gastroenteritis over four years (2020-2024) were included. Results: The sample population consisted of 84 individuals. Pathogen prevalence was identified in only 15%. Salmonella was the most frequently identified bacterial pathogen. While the WBC count and ESR were reassuring in most cases, the CRP and PCT measurements were almost always elevated. Compared to the stronger association observed with WBC counts, the correlation between PCT levels and ED visits were less significant. Higher CRP levels were associated with an increased use of antibiotics. Conclusion: The results of this study highlight that CRP is useful in identifying patients who are likely to have bacterial AGE and require antibiotics. Moreover, the WBC count is a helpful tool in predicting those likely to present to the ED again.
Collapse
Affiliation(s)
- Hazem Alhazmi
- Pediatric Emergency Department, NGHA, Jeddah 11426, Saudi Arabia
| | - Abeer Alzahrani
- King Abdullah's Specialized Children Hospital, Jeddah 11426, Saudi Arabia
| | - Saud Alshaikh
- King Abdullah's Specialized Children Hospital, Jeddah 11426, Saudi Arabia
| | - Lein Azzhary
- King Abdullah's Specialized Children Hospital, Jeddah 11426, Saudi Arabia
| | - Fatimah Alhaddad
- King Abdullah's Specialized Children Hospital, Jeddah 11426, Saudi Arabia
| | - Zeyad Alshamrani
- King Abdullah's Specialized Children Hospital, Jeddah 11426, Saudi Arabia
| | | |
Collapse
|
3
|
Morales-Luna L, Hernández-Ochoa B, González-Valdez A, Vázquez-Bautista M, Arreguin-Espinosa R, Pérez de la Cruz V, Enríquez-Flores S, De la Mora De la Mora I, Hernández-Urzúa E, Castillo-Rodríguez RA, Cárdenas-Rodríguez N, Martínez-Rosas V, Navarrete-Vázquez G, Gómez-Manzo S. Nitazoxanide Analogs: Synthesis, In Vitro Giardicidal Activity, and Effects on Giardia lamblia Metabolic Gene Expression. Int J Mol Sci 2025; 26:4504. [PMID: 40429649 DOI: 10.3390/ijms26104504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Giardiasis is a common intestinal infection caused by Giardia lamblia. The standard treatment for this parasitic infection involves the administration of nitroimidazoles, albendazoles, and nitrothiazoles. However, in recent years, Giardia lamblia strains resistant to these treatments have been reported. Additionally, the current therapies exhibit considerable side effects, highlighting the need for new compounds that specifically target this parasite. The aim of this study was to evaluate nitrothiazole analogs and assess their impact on the metabolic, redox, and structural gene expression of this parasite. First, the compounds CNZ-7, CNZ-8, FLP-2, FLP-6, and FLP-8 were tested at concentrations ranging from 0 to 50 µM to determine their IC50 in G. lamblia cultures. Subsequently, gene expression changes and structural cell damage in trophozoites were analyzed following incubation with the IC50 of each compound. The giardicidal activity of the compounds was also evaluated in a nitazoxanide-resistant strain. The results showed that FLP-2, FLP-6, and FLP-8 exhibited a stronger effect on trophozoite viability compared to nitazoxanide (NTZ) and metronidazole (MTZ). Both compounds induced an increase in the expression of phosphofructokinase (PFK), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), pyruvate kinase (PK), pyruvate phosphate dikinase (PPDK), and pyruvate:ferredoxin oxidoreductase (PFOR). Additionally, FLP-2 caused ultrastructural alterations in trophozoites. Furthermore, FLP-2, FLP-6, and FLP-8 demonstrated efficacy against drug-resistant strains. These findings suggest that FLP-2, FLP-6, and FLP-8 are promising candidates for the treatment of giardiasis, as they effectively reduce parasite viability, modify gene expression, and exhibit activity against drug-resistant G. lamblia strains.
Collapse
Affiliation(s)
- Laura Morales-Luna
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City 06720, Mexico
| | - Abigail González-Valdez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Montserrat Vázquez-Bautista
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
- Programa de Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Roberto Arreguin-Espinosa
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Verónica Pérez de la Cruz
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City 14269, Mexico
| | - Sergio Enríquez-Flores
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Ignacio De la Mora De la Mora
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Elizabeth Hernández-Urzúa
- Laboratorio de Toxicología Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Rosa Angélica Castillo-Rodríguez
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos, Instituto Politécnico Nacional, Boulevard de la Tecnología, 1036 Z-1, P 2/2, Atlacholoaya 62790, Mexico
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Víctor Martínez-Rosas
- Departamento de Ingeniería Química y Bioquímica, Instituto Tecnológico de Milpa Alta, Tecnológico Nacional de México, Milpa Alta, Mexico City 12300, Mexico
| | - Gabriel Navarrete-Vázquez
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Chamilpa, Cuernavaca 62209, Mexico
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| |
Collapse
|
4
|
Lotz CN, Schneeberger PHH, Concu M, Ali SM, Mrimi EC, Keiser J. Implementation of real-time PCR assays for diagnosing intestinal protozoa infections. Parasitol Res 2025; 124:40. [PMID: 40198454 PMCID: PMC11978536 DOI: 10.1007/s00436-025-08483-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/14/2025] [Indexed: 04/10/2025]
Abstract
Intestinal protozoa infections present a major public health challenge, particularly in areas with poor sanitation and limited access to clean water. Effective diagnostic methods are critical, yet traditional microscopy, though widely used for its simplicity, lacks the sensitivity and specificity of modern techniques like real-time Polymerase Chain Reaction (qPCR), making the latter a more effective tool for monitoring and assessing the burden of intestinal protozoa diseases. In this study, we implemented two duplex qPCR assays to detect Entamoeba dispar + Entamoeba histolytica and Cryptosporidium spp. + Chilomastix mesnili, along with singleplex assays for Giardia duodenalis and Blastocystis spp., using a 10 µL reaction volume. This marks the first molecular detection of Chilomastix mesnili by qPCR, enhancing diagnostic precision. Using these, we analyzed stool samples from 70 patients on Pemba Island, Tanzania, before and 54 samples after treatment with 20, 25, or 30 mg of emodepside or placebo, aiming to assess protozoa prevalence for this region and emodepside's potential antiprotozoal effects. Our qPCR reliably detected protozoa in 74.4% of samples, with Entamoeba histolytica and Entamoeba dispar in 31.4% of cases. Notably, one-third of these infections were caused by Entamoeba histolytica. No significant reduction in protozoa was observed after emodepside treatment compared to placebo. The study highlights the utility of qPCR in providing species-level differentiation and improving the speed and cost-effectiveness of testing. The high prevalence of protozoa in this region underscores the need for continued monitoring and control efforts, though emodepside was not effective against protozoa infections.
Collapse
Affiliation(s)
- Christian N Lotz
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Pierre H H Schneeberger
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Maura Concu
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Said M Ali
- Public Health Laboratory-Ivo de Carneri, Chake, Pemba, Tanzania
| | - Emmanuel C Mrimi
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
- Ifakara Health Institute, Ifakara, Tanzania
| | - Jennifer Keiser
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
5
|
Colston JM, Flynn TG, Denton AH, Schiaffino F, Majowicz SE, Devleesschauwer B, Di Bari C, Minato Y, Kosek MN. Updating global estimates of pathogen-attributable diarrhoeal disease burden: a methodology and integrated protocol for a broad-scope systematic review of a syndrome with diverse infectious aetiologies. BMJ Open 2025; 15:e093018. [PMID: 40180367 PMCID: PMC11969593 DOI: 10.1136/bmjopen-2024-093018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
INTRODUCTION Sustaining declines in global infectious disease burden will increasingly require efforts targeted to specific aetiological agents and common transmission pathways, particularly in this era of global change and human interconnectivity accelerating transmission and emergence of infectious pathogens. Systematic reviews and meta-analyses can be an effective and resource-efficient method for synthesising evidence regarding disease epidemiology for a wide range of pathogens and are the evidence source used by initiatives like the Planetary Child Health and Enterics Observatory (Plan-EO) and the WHO to determine the aetiology-specific epidemiology of diarrhoeal disease. Therefore, we developed this integrated systematic review methodology and protocol that aims to compile a database of published prevalence estimates for 17 diarrhoea-causing pathogens as inputs for disease burden estimation. METHODS AND ANALYSIS We will seek estimates of the prevalence of each endemic enteric pathogen estimated from published population-based studies that diagnosed their presence in stool samples from both asymptomatic subjects and those experiencing diarrhoea. The pathogens include the enteric viruses adenovirus, astrovirus, norovirus, rotavirus and sapovirus, the bacteria Campylobacter, Shigella, Salmonella enterica, Vibrio cholerae and the Escherichia coli (E. coli) pathotypes enteroaggregative E. coli, enteropathogenic E. coli, enterotoxigenic E. coli and Shiga-toxin-producing E. coli and the intestinal protozoa Cryptosporidium, Cyclospora, Entamoeba histolytica and Giardia. Meta-analytical methods for analyses of the resulting database (including risk of bias analysis) will be published alongside their findings. ETHICS AND DISSEMINATION This systematic review is exempt from ethics approval because the work is carried out on published documents. The database that results from this review will be made available as a supplementary file of the resulting published manuscript. It will also be made available for download from the Plan-EO website, where updated versions will be posted on a quarterly basis. PROSPERO REGISTRATION NUMBER CRD42023427998.
Collapse
Affiliation(s)
- Josh M Colston
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Thomas G Flynn
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Andrea H Denton
- Claude Moore Health Sciences Library, University of Virginia, Charlottesville, Virginia, USA
| | - Francesca Schiaffino
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Faculty of Veterinary Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Shannon E Majowicz
- School of Public Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Brecht Devleesschauwer
- Department of Epidemiology and Public Health, Sciensano, Brussels, Belgium
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Carlotta Di Bari
- Department of Epidemiology and Public Health, Sciensano, Brussels, Belgium
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Yuki Minato
- Department of Nutrition and Food Safety, World Health Organization, Geneva, Switzerland
| | - Margaret N Kosek
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
6
|
Liu X, Zhang R, Wang M, Tang C, Yang F, Yang Q, Huang C, Zhang Y, Ren Z, Liu L, Zhou G, Li J. Prevalence of pathogens associated with neonatal gastrointestinal infections: a systematic review and meta-analysis. Gut Pathog 2025; 17:16. [PMID: 40148930 PMCID: PMC11948943 DOI: 10.1186/s13099-025-00693-5] [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] [Received: 11/05/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
Gastrointestinal infections represent a significant global health burden, ranking as the second leading cause of mortality among infants and children. Identifying of pathogens causing neonatal gastrointestinal infections has presented tough challenges. This study aimed to summarize the prevalence of common pathogens associated with neonatal gastrointestinal infections through a comprehensive systematic review and meta-analysis of published literature. The last search was performed on January 08, 2025, from databases including EMBASE, PubMed, Cochrane Libary, and Web of Science. The outcome variable was infection rate, and the detection methods used were blood culture, tissue culture, or molecular biology methods. Two researchers independently extracted the research data and evaluated its quality using the JBI Critical Appraisal Tools. Twenty-three studies met the inclusion criteria. The pooled prevalence rates of common pathogens were as follows: Bacteria, including Escherichia (22.2%; 95% CI 8.3-40.4%, I2 = 98%), Clostridium (21.8%; 95% CI 2.2-53.8%, I2 = 96%), Klebsiella (19.2%; 95% CI 8.3-33.4%, I2 = 97%), Staphylococcus (13.6%; 95% CI 6.0-23.7%, I2 = 91%), Enterococcus (12.4%; 95% CI 1.8-30.3%, I2 = 96%), and Streptococcus (6.8%; 95% CI 2.5-12.9%, I2 = 43%). Fungi, including Candida (3.8%; 95% CI 0.6-9.6%, I2 = 84%). Viruses, including Rotavirus (11.6%; 95% CI 1.0-31.5%, I2 = 94%) and Adenovirus (4.1%; 95% CI 0.5-11.0%, I2 = 58%). Peritoneal culture methods demonstrated significantly higher positivity rates compared to other detection methods. Escherichia coli exhibited consistently high positivity rates across the three main detection methods. Klebsiella showed the highest positivity rates among bacterial isolates in both blood and peritoneal cultures. Pathogen detection and prevalence in necrotizing enterocolitis (NEC) cases were markedly higher compared to other conditions. This meta-analysis identifies key pathogens in gastrointestinal infections, including Klebsiella pneumoniae, Escherichia coli, Candida, Rotavirus, Adenovirus, and others that are suspected before clinical sample results are available. It also highlights that intestinal pathogen infections are linked to an increased risk of neonatal necrotizing enterocolitis (NEC) and emphasizes the advantages of peritoneal culture in detecting these infections.
Collapse
Affiliation(s)
- Xinyu Liu
- Faculty of Life Science and Technology and the Affiliated Anning First People'S Hospital, Kunming University of Science and Technology, Kunming, 650302, China
| | - Rui Zhang
- Faculty of Life Science and Technology and the Affiliated Anning First People'S Hospital, Kunming University of Science and Technology, Kunming, 650302, China
| | - Mengdie Wang
- Faculty of Life Science and Technology and the Affiliated Anning First People'S Hospital, Kunming University of Science and Technology, Kunming, 650302, China
| | - Chuncai Tang
- Department of Neonatology, The Affiliated Anning First People'S Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan, China
| | - Feifei Yang
- Faculty of Life Science and Technology and the Affiliated Anning First People'S Hospital, Kunming University of Science and Technology, Kunming, 650302, China
| | - Qingjuan Yang
- Department of Neonatology, The Affiliated Anning First People'S Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan, China
| | - Changyong Huang
- Department of Neonatology, The Affiliated Anning First People'S Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan, China
| | - Ying Zhang
- Department of Neonatology, The Affiliated Anning First People'S Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan, China
| | - Zhengmin Ren
- Department of Neonatology, The Affiliated Anning First People'S Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan, China
| | - Liqiao Liu
- Department of Neonatology, The Affiliated Anning First People'S Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan, China
| | - Guozhong Zhou
- Department of Pain Medicine, The Affiliated Anning First People'S Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan, China.
| | - Jia Li
- Department of Neonatology, The Affiliated Anning First People'S Hospital of Kunming University of Science and Technology, Kunming, 650302, Yunnan, China.
| |
Collapse
|
7
|
Mohammadi M. Exploring evidence from cells to clinics: is human bocavirus a gastrointestinal pathogen or just a risk factor? Arch Virol 2025; 170:87. [PMID: 40126644 DOI: 10.1007/s00705-025-06265-x] [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: 08/24/2024] [Accepted: 01/08/2025] [Indexed: 03/26/2025]
Abstract
Human bocaviruses (HBoVs), first identified in 2005 and composed of genotypes 1-4, have been increasingly detected worldwide in pediatric patients with acute gastroenteritis. HBoV-1 has been primarily associated with respiratory symptoms, while HBoV2-4 are mostly found in gastrointestinal (GI) samples. Results from case-control studies are still controversial; however, epidemiological evidence has shown a significant association between HBoV-2 and gastroenteritis. This review will primarily focus on this association, with a brief discussion of evidence related to other HBoV genotypes. Pathological and molecular studies on the pathogenesis of HBoV, particularly in GI cells, are very scarce, possibly due to the difficulties of in vitro HBoV culture. Nonetheless, some relevant findings from colorectal cancer samples have yielded valuable insights regarding the behavior of HBoV in the GI system. In the present review, we provide an updated overview of the epidemiological evidence for an association of HBoV infection with acute gastroenteritis and focus on the cellular and molecular perspectives of HBoV pathogenicity. Finally, we look at the knowledge gaps about how HBoV affects the GI system and explore future directions.
Collapse
Affiliation(s)
- Mehrdad Mohammadi
- Department of Laboratory Technical Sciences Development, Gene Fanavaran Teb Azma Company, Isfahan, Iran.
| |
Collapse
|
8
|
Karataş M, Bloemen M, Cuypers L, Wollants E, Van Ranst M, Matthijnssens J. 14 years of rotavirus A surveillance: unusual dominance of equine-like G3P[8] genotype with DS-1-like genotype constellation after the pandemic, Belgium, 2009 to 2023. Euro Surveill 2025; 30:2400442. [PMID: 40156349 PMCID: PMC11951416 DOI: 10.2807/1560-7917.es.2025.30.12.2400442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/10/2025] [Indexed: 04/01/2025] Open
Abstract
IntroductionDespite vaccine availability, rotavirus persists as a leading cause of gastroenteritis in children younger than 5 years.AimWe aimed to evaluate temporal changes in rotavirus epidemiology in Belgium between 2009 and 2023, including the period of the COVID-19 pandemic.MethodsWe collected 8,024 rotavirus-positive stool samples throughout Belgium. For 6,352 samples, we determined the G and/or P genotypes through sequencing of the genes encoding the outer capsid proteins VP7 and VP4.ResultsBefore the COVID-19pandemic, we received on average 622 samples per rotavirus epidemiological year, which decreased to 114 and 111 samples during the two pandemic rotavirus epidemiological years, followed by a peak of 1,048 samples in the first post-pandemic year. Notably, the proportion of cases in the age group 2-5-years increased from 20.3% before to 33% after the pandemic (p < 0.001). Over the 14-year study period, the most common genotypes were G2P[4], G3P[8] and G9P[8]. Post-pandemic data show an unusually strong dominance of the equine-like G3P[8] genotype which carried a DS-1-like genotype constellation in the period 2021 to 2023. Additionally, vaccinated individuals were significantly overrepresented among patients infected with the equine-like VP7 carrying G3P[8] rotavirus compared with other genotypes, including typical human VP7 G3P[8].ConclusionDespite the presence of typical yearly genotype fluctuations, several epidemiological changes were associated with the COVID-19 pandemic, including the unusual dominance of an emerging rotavirus strain against which current vaccines may be less effective. It is essential to closely monitor this strain to determine if the phenomenon is temporary.
Collapse
Affiliation(s)
- Mustafa Karataş
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Mandy Bloemen
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Lize Cuypers
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical Microbiology, Leuven, Belgium
- University Hospitals of Leuven, Department of Laboratory Medicine, National Reference Centre for Rotavirus, Leuven, Belgium
| | - Elke Wollants
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Marc Van Ranst
- University Hospitals of Leuven, Department of Laboratory Medicine, National Reference Centre for Rotavirus, Leuven, Belgium
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| | - Jelle Matthijnssens
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Clinical and Epidemiological Virology, Leuven, Belgium
| |
Collapse
|
9
|
Das R, Hossain MN, Levine MM, Kotloff KL, Nasrin D, Hossain MJ, Omore R, Sur D, Ahmed T, Breiman RF, Faruque ASG, Freeman MC. Impact of water, sanitation, and hygiene indicators on enteric viral pathogens among under-5 children in low resource settings. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 961:178401. [PMID: 39787644 PMCID: PMC11999324 DOI: 10.1016/j.scitotenv.2025.178401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 12/01/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Poor water, sanitation, and hygiene (WASH) are the primary risks of exposure to enteric viral infection. Our study aimed to describe the role of WASH conditions and practices as risk factors for enteric viral infections in children under 5. Literature on the risk factors associated with all-cause diarrhea masks the taxa-specific drivers of diarrhea from specific pathogens, limiting the application of relevant control strategies. We analyzed data from children enrolled in the Global Enteric Multicenter Study (GEMS) across seven study sites between December 2007 and March 2011 as cases (moderate-to-severe diarrhea: MSD) and asymptomatic controls. MSD was defined as new and acute diarrhea, with at least one of the following criteria for MSD: dehydration based on the study clinician's assessment, dysentery, or hospitalization with diarrhea or dysentery. Multiple logistic regression was used to examine the role of water quality, sanitation access, and hygiene facilities on the enteric viral pathogens adjusted for potential covariates. Among MSD symptomatic children (cases), longer water retrieval time (≥15 vs <15 min) was associated with increased Norovirus (aOR 1.33, 95 % CI 1.08-1.64) and Astrovirus (aOR 1.43, 95 % CI 1.01-2.02); scooping as drinking water retrieval method was associated with lower Rotavirus (aOR 0.77, 95 % CI 0.62-0.96), but higher Adenovirus (aOR 2.3, 95 % CI 1.32-4.11) infection compared to non-users. Among asymptomatic children (controls), consumption of non-tube well drinking water was associated with higher Norovirus infection (aOR 1.38, 95 % CI 1.01-1.89). Longer drinking water retrieval time (≥15 vs <15 min) increased Norovirus (aOR 1.47, 95 % CI 1.21-1.78) and Rotavirus (aOR 1.51, 95 % CI 1.20-1.89) infections. Pouring (aOR 0.51, 95 % CI 0.32-0.83) or scooping drinking water with a cup (aOR: 0.52; 95 % CI: 0.32, 0.86) lower Astrovirus infection; restricted water access (aOR 1.57, 95 % CI 1.21-2.02) higher Rotavirus infection. Handwashing before cooking was associated with lower Astrovirus (aOR 0.64, 95 % CI 0.47-0.88) infection in asymptomatic children. Our analysis did not find a significant effect of poor sanitation on different enteric viral pathogens examined. Norovirus and Astrovirus were detected more commonly in sub-Saharan Africa while Rotavirus was less prevalent than South Asia. Though we found statistically significant associations, we did not observe any overall pattern between WASH and enteric viral pathogens. Our findings provide insights to guide further research on targeted interventions for enteric viral pathogens, responsible for a major burden of pediatric diarrhea globally.
Collapse
Affiliation(s)
- Rina Das
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; Nutrition Research Division, icddr,b, Dhaka 1212, Bangladesh.
| | - Md Nasif Hossain
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Myron M Levine
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Karen L Kotloff
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dilruba Nasrin
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - M Jahangir Hossain
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, the Gambia
| | - Richard Omore
- Kenya Medical Research Institute (KEMRI), Nairobi, Kenya
| | - Dipika Sur
- National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Tahmeed Ahmed
- Nutrition Research Division, icddr,b, Dhaka 1212, Bangladesh; James P. Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh; Department of Global Health, University of Washington, Seattle, WA 98104, USA
| | - Robert F Breiman
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - A S G Faruque
- Nutrition Research Division, icddr,b, Dhaka 1212, Bangladesh
| | - Matthew C Freeman
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Ali HA, Nuh AM, Abdi HA, Muse AH. Multilevel analysis of prevalence and determinants of diarrhea among under-five children in Somalia: insights from the Somalia demographic and health survey 2020. BMC Public Health 2025; 25:202. [PMID: 39833845 PMCID: PMC11744809 DOI: 10.1186/s12889-025-21435-x] [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: 07/12/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Diarrheal diseases remain a critical public health challenge, particularly for children under five in low- and middle-income countries such as Somalia. This study aimed to assess the prevalence and determinants of diarrhea in this vulnerable population, utilizing data from the 2020 Somalia Demographic and Health Survey. METHODS This investigation employed secondary data from the 2020 Somalia Demographic and Health Survey. The analysis involved the extraction and cleaning of variables using STATA version 17. The primary outcome variable was childhood diarrhea, analyzed alongside 19 explanatory variables encompassing sociodemographic and environmental factors. Descriptive statistics, chi-square tests, and multilevel logistic regression models were employed to elucidate associations. RESULTS The study identified a diarrhea prevalence of 5.24%. Notably, sociodemographic factors significantly influenced this prevalence. Families within the second wealth category exhibited reduced odds of diarrhea (AOR = 0.699; 95% CI: 0.522-0.936), with those in the middle wealth category experiencing even lower odds (AOR = 0.524; CI: 0.391-0.702). Vitamin A supplementation was found to be critical; children lacking recent supplementation had increased odds of diarrhea (AOR = 1.790; CI: 1.440-2.225). Furthermore, feeding practices indicated that children not using bottle nipples faced higher odds of diarrhea (AOR = 1.377; CI: 1.172-1.617). Maternal age also played a significant role, with mothers aged 45-49 presenting higher odds of their children experiencing diarrhea (AOR = 7.650; CI: 1.762-33.184). Institutional births were associated with lower odds of diarrhea (AOR = 0.693; CI: 0.568-0.847), while children of educated fathers exhibited increased odds (AOR = 1.414; CI: 1.090-1.833). Additionally, lack of deworming medication significantly heightened the odds of diarrhea (AOR = 4.450; CI: 3.619-5.482). CONCLUSION The prevalence of diarrhea among children under five relatively high in Somalia. Key determinants include vitamin A supplementation, feeding with bottle, household wealth, age of mothers, place of delivery, paternal education, and deworming medication. Public health interventions should focus on enhancing household economic status, and ensuring regular deworming. Increasing the coverage and frequency of vitamin A supplementation could enhance children's immune systems and reduce diarrhea incidence. Moreover, targeted economic support, health care programs for older mothers are essential to mitigate the adverse effects of socioeconomic factors on child health, ultimately leading to a substantial reduction in diarrheal diseases among young children.
Collapse
Affiliation(s)
- Hamse Arab Ali
- Faculty of Science and Humanities, School of Postgraduate Studies and Research (SPGSR), Amoud University , Borama, Somaliland.
| | - Abdulkadir Mohamed Nuh
- Department of Nutrition and Dietetics, Addis-ababa Medical University College, Hargeisa, Somaliland
| | - Hamse Adam Abdi
- College of Applied and Natural Science, University of Hargeisa, Hargeisa, Somaliland
| | - Abdisalam Hassan Muse
- Faculty of Science and Humanities, School of Postgraduate Studies and Research (SPGSR), Amoud University, Borama, Somaliland
| |
Collapse
|
11
|
Song F, Zeng Y, Sheng R, Lin Y, Wang X, Hong C, Luo G, Wang Y, Fang M, He S, Zhang S, Zheng Q, Li T, Ge S, Zhang J, Xia N. VP8 Mosaic Nanoparticles Elicit Cross-Neutralizing Immune Responses and Provide Protection Against Heterotypic Rotavirus Challenge in Mice. ACS NANO 2024; 18:31809-31822. [PMID: 39497609 DOI: 10.1021/acsnano.4c07061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Group A rotaviruses (RVA) remain one of the dominant pathogens causing diarrhea in children under 5 years of age worldwide, despite a sharp decrease of RVA-associated diarrhea and mortality since the introduction of rotavirus vaccines. The decreased effectiveness of live attenuated rotavirus vaccines, coupled with the emergence of new rotavirus genotypes and the risk of cross-species virus transmission, underscores the necessity to develop more effective and broad-spectrum rotavirus vaccines. In this study, we utilized nanoparticles coupled with the SpyCatcher-SpyTag system to effectively display the truncated VP8-1 protein. The modular display of the monovalent VP8-1 proteins markedly increased the immunogenicity of VP8-1. Furthermore, the bivalent display of VP8-1 proteins from simian rotavirus SA11 and lamb rotavirus LLR on the same particle not only increased immunogenicity against homotypic antigens but also elicited robust heterotypic immune responses and conferred effective protection against a distant heterotypic rotavirus with sequence identities of only 62%-66% in an adult mouse model. Therefore, mosaic VP8 nanoparticles could be considered as a viable strategy for the development of the next-generation broad-spectrum rotavirus vaccine.
Collapse
Affiliation(s)
- Feibo Song
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yuanjun Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Roufang Sheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yunyun Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xuechun Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Congming Hong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Guoxing Luo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yingbin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shuizhen He
- Haicang Hospital of Xiamen, Xiamen 361026, China
| | - Shiyin Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qingbing Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Tingdong Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shengxiang Ge
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Life Sciences, Xiamen University, Xiamen 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Collaborative Innovation Center of Biologic Products, National Innovation Platform for Industry-Education Integration in Vaccine Research, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
12
|
Ford-Siltz LA, Tohma K, Kihn K, Kendra JA, Deredge D, Wintrode P, Gao Y, Parra GI. Characterization of cross-reactive, non-neutralizing monoclonal antibodies against a pandemic GII.4 norovirus variant. Microbiol Spectr 2024; 12:e0114324. [PMID: 39470287 PMCID: PMC11619288 DOI: 10.1128/spectrum.01143-24] [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: 05/07/2024] [Accepted: 09/10/2024] [Indexed: 10/30/2024] Open
Abstract
Antibodies are thought to play a major role in protection against human norovirus infection. Mouse humoral responses closely mimic those of humans; thus, mouse models are used to characterize norovirus epitopes on the major viral capsid protein, VP1. We have developed a panel of mouse monoclonal antibodies (mAbs) produced against the last pandemic variant to emerge, Sydney 2012. While most mAbs (25/44) were mapped to variable antigenic sites on VP1, 19 of the mAbs were cross-reactive against multiple genotypes or GII.4 variants. Most (12/19) of the cross-reactive mAbs bound to the Shell domain and were cross-reactive with different GII noroviruses. Interestingly, mAb 30A11 exhibited cross-reactivity against all tested norovirus genotypes (GI, GII, GIV, and GIX). This mAb was mapped to a highly conserved region of the Shell domain (51PIDPWII57) using peptide ELISA and immunofluorescence. Of those mapping to the Protruding (P) domain, two (19C10 and 14B11) showed cross-reactivity with GII noroviruses. Using hydrogen-deuterium exchange mass spectrometry, we mapped 19C10 to a conserved region of the P domain near the P/Shell interface, which explains its cross-reactivity with different GII noroviruses and lack of histo-blood group antigen-blocking activity. Binding and mutational analyses showed that residues 518, 519, and 525 are important for 19C10 and 14B11 epitope recognition. While the antibodies described here are mostly non-neutralizing, they can be useful tools for research and diagnostics of noroviruses. The role of non-neutralizing, cross-reactive antibodies targeting different areas of the viral capsid merits further research to facilitate our understanding of immunity to norovirus infection and disease. IMPORTANCE To gain insights into the overall immune responses to human norovirus, we characterized non-neutralizing, cross-reactive monoclonal antibodies (mAbs) developed against a pandemic GII.4 norovirus. We determined the binding epitope of an antibody that exhibited cross-reactivity against all tested noroviruses, which makes it a useful tool for research and diagnostics. The epitope of two additional non-neutralizing mAbs was mapped to a less conserved region on the viral capsid protein, explaining their cross-reactivity patterns. Often overlooked, the role of non-neutralizing, cross-reactive mAbs merits further research to facilitate our understanding of immunity to norovirus infection and disease.
Collapse
Affiliation(s)
| | - Kentaro Tohma
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| | - Kyle Kihn
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Joseph A. Kendra
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| | - Daniel Deredge
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Patrick Wintrode
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Yamei Gao
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| | - Gabriel I. Parra
- Division of Viral Products, CBER, FDA, Silver Spring, Maryland, USA
| |
Collapse
|
13
|
Bocchio F, Mancabelli L, Milani C, Lugli GA, Tarracchini C, Longhi G, Conto FD, Turroni F, Ventura M. Compendium of Bifidobacterium-based probiotics: characteristics and therapeutic impact on human diseases. MICROBIOME RESEARCH REPORTS 2024; 4:2. [PMID: 40207278 PMCID: PMC11977362 DOI: 10.20517/mrr.2024.52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 04/11/2025]
Abstract
The human microbiota, a complex community of microorganisms residing in and on the human body, plays a crucial role in maintaining health and preventing disease. Bifidobacterium species have shown remarkable therapeutic potential across a range of health conditions, thus being considered optimal probiotic bacteria. This review provides insights into the concept of probiotics and explores the impact of bifidobacteria on human health, focusing on the gastrointestinal, respiratory, skeletal, muscular, and nervous systems. It also integrates information on the available genetic bases underlying the beneficial effects of each bifidobacterial probiotic species on different aspects of human physiology. Notably, Bifidobacterium-based probiotics have proven effective in managing gastrointestinal conditions such as constipation, antibiotic-associated diarrhea, irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and Helicobacter pylori infections. These benefits are achieved by modulating the intestinal microbiota, boosting immune responses, and strengthening the gut barrier. Moreover, Bifidobacterium species have been reported to reduce respiratory infections and asthma severity. Additionally, these probiotic bacteria offer benefits for skeletal and muscular health, as evidenced by Bifidobacterium adolescentis and Bifidobacterium breve, which have shown anti-inflammatory effects and symptom relief in arthritis models, suggesting potential in treating conditions like rheumatoid arthritis. Furthermore, probiotic therapies based on bifidobacterial species have shown promising effects in alleviating anxiety and depression, reducing stress, and enhancing cognitive function. Overall, this review integrates the extensive scientific literature now available that supports the health-promoting applications of probiotic Bifidobacterium species and underscores the need for further research to confirm their clinical efficacy across different body systems.
Collapse
Affiliation(s)
- Fabiana Bocchio
- Department of Medicine and Surgery, University of Parma, Parma 43124, Italy
- Authors contributed equally
| | - Leonardo Mancabelli
- Department of Medicine and Surgery, University of Parma, Parma 43124, Italy
- Interdepartmental Research Centre “Microbiome Research Hub”, University of Parma, Parma 43124, Italy
- Authors contributed equally
| | - Christian Milani
- Interdepartmental Research Centre “Microbiome Research Hub”, University of Parma, Parma 43124, Italy
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma 43124, Italy
| | - Gabriele Andrea Lugli
- Interdepartmental Research Centre “Microbiome Research Hub”, University of Parma, Parma 43124, Italy
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma 43124, Italy
| | - Chiara Tarracchini
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma 43124, Italy
| | - Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma 43124, Italy
| | - Flora De Conto
- Department of Medicine and Surgery, University of Parma, Parma 43124, Italy
| | - Francesca Turroni
- Interdepartmental Research Centre “Microbiome Research Hub”, University of Parma, Parma 43124, Italy
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma 43124, Italy
| | - Marco Ventura
- Interdepartmental Research Centre “Microbiome Research Hub”, University of Parma, Parma 43124, Italy
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma 43124, Italy
| |
Collapse
|
14
|
Anueyiagu KN, Agu CG, Umar U, Lopes BS. Antimicrobial Resistance in Diverse Escherichia coli Pathotypes from Nigeria. Antibiotics (Basel) 2024; 13:922. [PMID: 39452189 PMCID: PMC11504273 DOI: 10.3390/antibiotics13100922] [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: 08/17/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/26/2024] Open
Abstract
Escherichia coli is a gram-negative commensal bacterium living in human and animal intestines. Its pathogenic strains lead to high morbidity and mortality, which can adversely affect people by causing urinary tract infections, food poisoning, septic shock, or meningitis. Humans can contract E. coli by eating contaminated food-such as raw or undercooked raw milk, meat products, and fresh produce sold in open markets-as well as by coming into contact with contaminated settings like wastewater, municipal water, soil, and faeces. Some pathogenic strains identified in Nigeria, include Enterohemorrhagic (Verotoxigenic), Enterotoxigenic, Enteropathogenic, Enteroinvasive, and Enteroaggregative E. coli. This causes acute watery or bloody diarrhoea, stomach cramps, and vomiting. Apart from the virulence profile of E. coli, antibiotic resistance mechanisms such as the presence of blaCTX-M found in humans, animals, and environmental isolates are of great importance and require surveillance and monitoring for emerging threats in resource-limited countries. This review is aimed at understanding the underlying mechanisms of evolution and antibiotic resistance in E. coli in Nigeria and highlights the use of improving One Health approaches to combat the problem of emerging infectious diseases.
Collapse
Affiliation(s)
- Kenneth Nnamdi Anueyiagu
- Department of Public Health Technology, Federal College of Animal Health and Production Technology, Vom 200273, Nigeria;
| | | | - Uzal Umar
- Department of Medical Microbiology and Parasitology, University of Jos, Jos 930105, Nigeria;
| | - Bruno Silvester Lopes
- School of Health and Life Sciences, Teesside University, Middlesbrough TS1 3BX, UK
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| |
Collapse
|
15
|
Ballard AM, Corozo Angulo B, Laramee N, Pace Gallagher J, Haardörfer R, Freeman MC, Trostle J, Eisenberg JNS, Lee GO, Levy K, Caruso BA. Multilevel factors drive child exposure to enteric pathogens in animal feces: A qualitative study in northwestern coastal Ecuador. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003604. [PMID: 39292655 PMCID: PMC11410186 DOI: 10.1371/journal.pgph.0003604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/26/2024] [Indexed: 09/20/2024]
Abstract
Exposure to animal feces and associated enteric pathogens poses significant risks to child health. However, public health strategies to mitigate enteric infections among children largely aim to reduce exposure to human feces, overlooking transmission pathways related to animal feces. In this study we examine if and how children are exposed to enteric pathogens in animal feces in northwestern coastal Ecuador. We conducted qualitative interviews with mothers of children aged 10-18 months that owned (n = 32) and did not own (n = 26) animals in urban and rural communities. Using thematic analysis, we identified community, household, and child behavioral factors that influence exposure. We also compared child exposure by household animal ownership. Our findings revealed myriad opportunities for young children to be exposed to enteric pathogens in many locations and from multiple animal sources, regardless of household animal ownership. Animal feces management practices (AFM) used by mothers, such as rinsing feces into ditches and throwing feces into surrounding areas, may increase environmental contamination outside their homes and in their communities. Unsafe AFM practices were similar to unsafe child feces management practices reported in other studies, including practices related to defecation location, feces removal and disposal, environmental contamination cleaning, and handwashing. Findings suggest that animal feces may contaminate the environment along similar pathways as human feces. Identification and incorporation of safe AFM practices, similar to those developed for child feces management, would 1) mitigate child exposure to enteric pathogens by reducing animal feces contamination in domestic and public spaces; and 2) enable an integrated approach to address enteric pathogen exposure pathways related to animal and child feces.
Collapse
Affiliation(s)
- April M Ballard
- Department of Population Health Sciences, Georgia State University School of Public Health, Atlanta, Georgia, United States of America
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, Georgia, United States of America
| | | | - Nicholas Laramee
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, Georgia, United States of America
| | - Jayden Pace Gallagher
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, Georgia, United States of America
| | - Regine Haardörfer
- Department of Behavioral, Social, and Health Education Sciences, Emory University Rollins School of Public Health, Atlanta, Georgia, United States of America
| | - Matthew C Freeman
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, Georgia, United States of America
| | - James Trostle
- Department of Anthropology, Trinity College, Hartford, Connecticut, United States of America
| | - Joseph N S Eisenberg
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Gwenyth O Lee
- Rutgers Global Health Institute and Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, New Jersey, United States of America
| | - Karen Levy
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, Washington, United States of America
| | - Bethany A Caruso
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, Georgia, United States of America
- Hubert Department of Global Health, Emory University Rollins School of Public Health, Atlanta, Georgia, United States of America
- Department of Behavioral, Social, and Health Education Sciences, Emory University Rollins School of Public Health, Atlanta, Georgia, United States of America
| |
Collapse
|
16
|
Zizza A, Guido M, Sedile R, Benelli M, Nuzzo M, Paladini P, Romano A, Grima P. A Multi-Pathogen Retrospective Study in Patients Hospitalized for Acute Gastroenteritis. Diseases 2024; 12:213. [PMID: 39329882 PMCID: PMC11431555 DOI: 10.3390/diseases12090213] [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: 06/27/2024] [Revised: 07/25/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Acute gastroenteritis (AGE) is a gastrointestinal tract disease often caused by consuming food or water contaminated by bacteria, viruses, or parasites, that can lead to severe symptoms requiring hospitalization. A retrospective study on patients admitted for AGE between 2021 and 2023 at the Pediatrics and Infectious Diseases Departments of Lecce Hospital was conducted. Demographic characteristics, year and month of admission, length of hospital stay, etiological agents, co-infections, and blood chemistry data of patients were collected. The study included 103 patients ranging in age from 0 to 15 years, with 58.25% being male. A total of 78 bacterial, 35 viral, and 7 parasitic infections were identified. The most commonly detected pathogens were Escherichia coli (38.83%), Norovirus (28.16%), Campylobacter jejuni (22.33%), and Salmonella typhi/paratyphi (10.68%). Only a few cases of Cryptosporidium (5.83%) were identified. Additionally, 17 co-infections (16.50%) were detected. Viral infections are the primary cause of hospitalization for AGE in children <5 years, while bacterial infections are more common among older patients. The significantly higher number of children <5 years old with elevated creatinine compared to children ≥5 years suggested that young children are more susceptible to dehydration than older children. Few cases of AGE were attributed to pathogens for which a vaccine has already been licensed. AGE is a serious health concern that could be effectively prevented by implementing food-based and community-level sanitation systems, as well as by increasing vaccination coverage of available vaccines and developing new effective and safe vaccines.
Collapse
Affiliation(s)
- Antonella Zizza
- Institute of Clinical Physiology, National Research Council, 73100 Lecce, Italy;
| | - Marcello Guido
- Laboratory of Hygiene, Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| | - Raffaella Sedile
- Institute of Clinical Physiology, National Research Council, 73100 Lecce, Italy;
| | - Marzia Benelli
- Pediatric Unit, Vito Fazzi Hospital, 73100 Lecce, Italy; (M.B.); (P.P.)
| | - Milva Nuzzo
- Infectious Diseases Unit, Vito Fazzi Hospital, 73100 Lecce, Italy; (M.N.); (A.R.); (P.G.)
| | - Pasquale Paladini
- Pediatric Unit, Vito Fazzi Hospital, 73100 Lecce, Italy; (M.B.); (P.P.)
| | - Anacleto Romano
- Infectious Diseases Unit, Vito Fazzi Hospital, 73100 Lecce, Italy; (M.N.); (A.R.); (P.G.)
| | - Pierfrancesco Grima
- Infectious Diseases Unit, Vito Fazzi Hospital, 73100 Lecce, Italy; (M.N.); (A.R.); (P.G.)
| |
Collapse
|
17
|
Kachooei A, Mirhoseinian M, Jalilvand S, Latifi T, Feizi M, Shahosseini Z, Arashkia A, Marashi SM, Shoja Z. Molecular characterization of human astrovirus infection in children under 5 years of age with acute gastroenteritis in Tehran, Iran, 2021-2022: co-infection with rotavirus. Virus Genes 2024; 60:357-369. [PMID: 38744749 DOI: 10.1007/s11262-024-02075-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
Human astroviruses (HAstVs) are considered important causative pathogens of acute gastroenteritis (AGE) in children under 5 years of age worldwide, along with group A rotavirus (RVA), norovirus (NoV), and enteric adenovirus (EAdV). The present study was aimed to both detect HAstV and its co-infections and investigate genetic analysis of circulating HAstV and co-infected virus in hospitalized children under 5 years of age with AGE in Iran. Accordingly, a sum of 200 stool specimens were screened by PCR for HAstV during 2021-2022. The HAstV was found in 0.5% of 200 specimens (n = 1) while was co-infected with RVA. The genetic and phylogenetic analysis indicated HAstV1 genotype, which clustered with viruses from lineage 1b, which has not been previously reported in Iran. The detected RVA strain belonged to G1 lineage II/P[8]-lineage III, which has been reported previously in Iran as the most common strain. The further genetic analysis of RVA VP6 and NSP4 demonstrated an atypical genotype pattern G1P[8]-I1-E2, as a mono-reassortant of a Wa-like genogroup, which appeared to be reassorted with the NSP4 gene of E2 genotype of the G2P[4] DS-1 genogroup. Although the clinical outcomes of the AGE-causing viruses co-infection is not yet entirely clear, it seems that future studies will be helpful to merge clinical and epidemiological data of co-infecting viruses for a more accurate medical and clinical relevance in symptomatic children.
Collapse
Affiliation(s)
- Atefeh Kachooei
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahtab Mirhoseinian
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Tayebeh Latifi
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Mahsa Feizi
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Arash Arashkia
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
- Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Sayed Mahdi Marashi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
- Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
18
|
Traore KA, Akapovi MM, Ouedraogo N, Ouoba JB, Roques P, Barro N. Geographical distribution of enteric pathogenic viruses in Burkina Faso: a systematic review and meta-analysis. BMC Infect Dis 2024; 24:756. [PMID: 39080551 PMCID: PMC11290313 DOI: 10.1186/s12879-024-09668-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Viruses, which are transmitted mainly via the digestive tract, are responsible for the high morbidity and mortality of diseases, particularly in low-income countries. Although several studies have established the prevalence and characterization of various enteric viruses in Burkina Faso, to date, no aggregate data have been released. OBJECTIVE Our objective was to describe the available data on the prevalence and circulating genotypes of enteric pathogen viruses responsible for human infections in Burkina Faso by carrying out a systematic review and meta-analysis. METHODS Potentially relevant studies were identified by a search of PubMed, ScienceDirect, Google Scholar, university libraries and by a manual search of the reference lists of identified studies. The search with no restrictions on language or age was limited to studies conducted only in Burkina. Study selection, data extraction, and methodological quality of the included studies were performed independently by two investigators. Heterogeneity between studies was assessed using the Cochrane Q test and I2 test statistics based on the random effects model. Comprehensive meta-analysis (CMA 3.7) was employed to compute the pooled prevalence of pathogens identified in the studies. RESULTS Forty-three (43) studies reporting 4,214 diagnosed cases in all aged human populations were selected. Overall, 72.6% of the pathogens diagnosed were gastroenteritis, and 27.2% were entero-transmissible hepatitis viruses. Rotavirus was the most common cause of human viral gastroenteritis, accounting for 27.7% (95% CI: 20.9 - 35.8) of the cases, followed by norovirus (16% (95% CI: 12.25 - 20.6)) and sapovirus (11.2% (95% CI: 6.2 - 19.4)). In terms of human entero-transmissible infections, hepatitis A virus (HAV) was the most prevalent (52% [95% CI: 14.2-87.7] of total antibodies), followed by hepatitis E virus (HEV) (28.3% [95% CI: 17.7-42]). CONCLUSIONS This study highlights the substantial burden of viral enteric infections and highlights the need for more molecular epidemiological studies to improve preventive measures against these viruses.
Collapse
Affiliation(s)
- Kuan Abdoulaye Traore
- Laboratoire Sciences de la Vie et de la Terre (LaSVT), Université Norbert ZONGO (UNZ), Koudougou, Burkina Faso.
- Laboratoire de Biologie Moléculaire, d'Epidémiologie et de Surveillance des Bactéries et Virus Transmissibles par les Aliments (LaBESTA), Université Joseph KI-ZERBO, Ouagadougou, Burkina Faso.
| | - Messanh Marius Akapovi
- Laboratoire de Biologie Moléculaire, d'Epidémiologie et de Surveillance des Bactéries et Virus Transmissibles par les Aliments (LaBESTA), Université Joseph KI-ZERBO, Ouagadougou, Burkina Faso
| | - Nafissatou Ouedraogo
- Laboratoire de Biologie Moléculaire, d'Epidémiologie et de Surveillance des Bactéries et Virus Transmissibles par les Aliments (LaBESTA), Université Joseph KI-ZERBO, Ouagadougou, Burkina Faso
- Université de Dédougou (UDD), Dédougou, Burkina Faso
| | - Jean Bienvenue Ouoba
- Laboratoire de Biologie Moléculaire, d'Epidémiologie et de Surveillance des Bactéries et Virus Transmissibles par les Aliments (LaBESTA), Université Joseph KI-ZERBO, Ouagadougou, Burkina Faso
- Centre universitaire de Manga (CUM), Manga, Burkina Faso
| | - Pierre Roques
- Virology Unit, Institut Pasteur de Guinée (IPGui), Conakry, Guinea
| | - Nicolas Barro
- Laboratoire de Biologie Moléculaire, d'Epidémiologie et de Surveillance des Bactéries et Virus Transmissibles par les Aliments (LaBESTA), Université Joseph KI-ZERBO, Ouagadougou, Burkina Faso
| |
Collapse
|
19
|
Thystrup C, Majowicz SE, Kitila DB, Desta BN, Fayemi OE, Ayolabi CI, Hugho E, Buys EM, Akanni GB, Machava NE, Monjane C, Hald T, Pires SM. Etiology-specific incidence and mortality of diarrheal diseases in the African region: a systematic review and meta-analysis. BMC Public Health 2024; 24:1864. [PMID: 38997671 PMCID: PMC11241906 DOI: 10.1186/s12889-024-19334-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Diarrheal diseases substantially affect public health impact in low- and middle-income countries (LMIC), particularly in Africa, where previous studies have indicated a lack of comprehensive data. With a growing number of primary studies on enteric infections in Africa, this study aimed to estimate the incidence and mortality of diarrheal pathogens across all ages in Africa in the year 2020. We also explored different methodological assumptions to allow comparison with other approaches. METHODS Through a systematic review and meta-analysis of data from African LMICs, we estimated the etiology proportions for diarrheal diseases and deaths. We combined the etiology proportions with incidence data collected from a population survey in Africa from 2020 and mortality data from the Global Health Observatory of WHO. RESULTS We estimated 1,008 billion diarrhea cases (95% UI 447 million-1,4 billion) and 515,031 diarrhea deaths (95% UI 248,983-1,007,641) in the African region in 2020. In children under five, enteroaggregative E. coli (EAEC) (44,073 cases per 100,000 people, 95% UI 18,818 - 60,922) and G. lamblia (36,116 cases per 100,000 people, 95% UI 15,245 - 49,961) were the leading causes of illness. Enteroinvasive E. coli (EIEC) (155 deaths per 100,000 people, 95% UI 106.5-252.9) and rotavirus (61.5 deaths per 100,000 people, 95% UI 42.3-100.3) were the primary causes of deaths. For children over five and adults, Salmonella spp. caused the largest number of diarrheal cases in the population of children ≥ 5 and adults (122,090 cases per 100,000 people, 95% UI 51,833 - 168,822), while rotavirus (16.4 deaths per 100,000 people, 95% UI 4.2-36.7) and enteroaggregative E. coli (EAEC) (14.6 deaths per 100,000 people, 95% UI 3.9-32.9) causing the most deaths. Geographically, the highest incidence of diarrhea was in Eastern Africa for children under five (114,389 cases per 100,000 people, 95% UI 34,771 - 172,884) and Central Africa for children over five and adults (117,820 cases per 100,000 people, 95% UI 75,111-157,584). Diarrheal mortality was highest in Western Africa for both children below five and above (children < 5: 194.5 deaths per 100,000 people, 95% UI 120-325.4; children ≥ 5 and above: 33.5 deaths per 100,000 people, 95% UI 12.9-75.1). CONCLUSION These findings provide new information on the incidence and mortality of sixteen pathogens and highlight the need for surveillance and control of diarrheal infectious diseases in Africa. The cause-specific estimates are crucial for prioritizing diarrheal disease prevention in the region.
Collapse
Affiliation(s)
- Cecilie Thystrup
- Kgs. Lyngby, National Food Institute, Technical University of Denmark, Lyngby, Denmark.
| | - Shannon E Majowicz
- School of Public Health Sciences, University of Waterloo, Waterloo, Canada
| | - Dinaol B Kitila
- School of Biological Sciences and Biotechnology, Haramaya University, Dire Dawa, Ethiopia
- College of Veterinary Medicine, Haramaya University, Dire Dawa, Ethiopia
| | - Binyam N Desta
- School of Public Health Sciences, University of Waterloo, Waterloo, Canada
| | - Olanrewaju E Fayemi
- Centre for Research, Innovation, and Collaboration, Department of Biological Sciences, Mountain Top University, Prayer City, Nigeria
| | - Christianah I Ayolabi
- Centre for Research, Innovation, and Collaboration, Department of Biological Sciences, Mountain Top University, Prayer City, Nigeria
- Department of Microbiology, University of Lagos, Lagos, Nigeria
| | - Ephrasia Hugho
- Kilimanjaro Clinical Research Institute, Moshi, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, 2021 CRD42021251511, Tanzania
| | - Elna M Buys
- Department of Consumer and Food Sciences, University of Pretoria, Pretoria, South Africa
| | - Gabriel B Akanni
- Centre for Research, Innovation, and Collaboration, Department of Biological Sciences, Mountain Top University, Prayer City, Nigeria
| | - Norgia E Machava
- Instituto Superior de Ciencias de Saúde (ISCISA), Maputo, Mozambique
| | - Celso Monjane
- Medical Faculty, Eduardo Mondlane University, Maputo, Mozambique
| | - Tine Hald
- Kgs. Lyngby, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Sara M Pires
- Kgs. Lyngby, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
20
|
Cohen DG, Wingert RA. Forever young by Alpha(diversity)ville: restricting intestinal microbiome maturation stunts immune system development and increases susceptibility to infection. Tissue Barriers 2024; 12:2281209. [PMID: 37978888 PMCID: PMC11262204 DOI: 10.1080/21688370.2023.2281209] [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: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
The microbiome is a keystone of adult gastrointestinal (GI) tract health, where it facilitates digestion, wards off pathogen colonization, and exerts a powerful influence on the physiological health of organs ranging from the brain to the kidneys. From its establishment at birth and through the initial years of childhood, the human microbiome is particularly dynamic, shifting in its composition and alpha (species) diversity to an adult profile as dietary sustenance transitions from milk-based sources to others such as solid food. An innovative study has now demonstrated how microbiome maturation is requisite both for the progression of immune system development and for long-term gut barrier function. These insights have significant ramifications for designing pediatric approaches to cultivate immune cell ontogeny in the formative stages of human infancy.
Collapse
Affiliation(s)
- Dorrian G. Cohen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, USA
| |
Collapse
|
21
|
Allingham C, Taniguchi M, Kinchla AJ, Moore MD. The Influence of Simulated Organic Matter on the Inactivation of Viruses: A Review. Viruses 2024; 16:1026. [PMID: 39066189 PMCID: PMC11281590 DOI: 10.3390/v16071026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Viruses impose a significant public health burden globally, and one of the key elements in controlling their transmission is the ability to inactivate them using disinfectants. However, numerous challenges to inactivating foodborne viruses exist due to inherent viral characteristics (such as recalcitrance to commonly used inactivation agents) and external factors (such as improper cleaning before application of inactivation agent, improper contact time, etc.). Given the potential for improper application of disinfectants (such as shorter than recommended contact time, improper disinfectant concentration, etc.), understanding the performance of a disinfectant in the presence of an organic load is important. To accomplish this, the introduction of simulated organic loads is often used when studying the efficacy of a disinfectant against different viruses. However, the different types of simulated organic loads used in foodborne virus inactivation studies or their relative effects on inactivation have not been reviewed. The purpose of this review is to survey different simulated organic load formulations used in studying foodborne virus inactivation, as well as present and compare the influence of these different formulations on viral inactivation. The findings included in this review suggest that many simulated organic load formulations can reduce disinfectants' efficacy against viruses. Based on the findings in this review, blood, particularly serum or feces, are among the most commonly used and efficacious forms of simulated organic load in many tests.
Collapse
Affiliation(s)
- Christina Allingham
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (M.T.); (A.J.K.)
| | - Miyu Taniguchi
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (M.T.); (A.J.K.)
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, Minato City, Tokyo 108-8477, Japan
| | - Amanda J. Kinchla
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (M.T.); (A.J.K.)
| | - Matthew D. Moore
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (M.T.); (A.J.K.)
| |
Collapse
|
22
|
Gaona-López C, Méndez-Álvarez D, Moreno-Rodríguez A, Bautista-Martínez JL, De Fuentes-Vicente JA, Nogueda-Torres B, García-Torres I, López-Velázquez G, Rivera G. TATA-Binding Protein-Based Virtual Screening of FDA Drugs Identified New Anti-Giardiasis Agents. Int J Mol Sci 2024; 25:6238. [PMID: 38892424 PMCID: PMC11172525 DOI: 10.3390/ijms25116238] [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: 05/01/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Parasitic diseases, predominantly prevalent in developing countries, are increasingly spreading to high-income nations due to shifting migration patterns. The World Health Organization (WHO) estimates approximately 300 million annual cases of giardiasis. The emergence of drug resistance and associated side effects necessitates urgent research to address this growing health concern. In this study, we evaluated over eleven thousand pharmacological compounds sourced from the FDA database to assess their impact on the TATA-binding protein (TBP) of the early diverging protist Giardia lamblia, which holds medical significance. We identified a selection of potential pharmacological compounds for combating this parasitic disease through in silico analysis, employing molecular modeling techniques such as homology modeling, molecular docking, and molecular dynamics simulations. Notably, our findings highlight compounds DB07352 and DB08399 as promising candidates for inhibiting the TBP of Giardia lamblia. Also, these compounds and DB15584 demonstrated high efficacy against trophozoites in vitro. In summary, this study identifies compounds with the potential to combat giardiasis, offering the prospect of specific therapies and providing a robust foundation for future research.
Collapse
Affiliation(s)
- Carlos Gaona-López
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico;
| | - Domingo Méndez-Álvarez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico;
| | - Adriana Moreno-Rodríguez
- Laboratorio de Estudios Epidemiológicos, Clínicos, Diseños Experimentales e Investigación, Facultad de Ciencias Químicas, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (A.M.-R.); (J.L.B.-M.)
| | - Juan Luis Bautista-Martínez
- Laboratorio de Estudios Epidemiológicos, Clínicos, Diseños Experimentales e Investigación, Facultad de Ciencias Químicas, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (A.M.-R.); (J.L.B.-M.)
| | | | - Benjamín Nogueda-Torres
- Departamento de Parasitología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico;
| | - Itzhel García-Torres
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico; (I.G.-T.); (G.L.-V.)
| | - Gabriel López-Velázquez
- Laboratorio de Biomoléculas y Salud Infantil, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico; (I.G.-T.); (G.L.-V.)
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico;
| |
Collapse
|
23
|
Black RE, Perin J, Yeung D, Rajeev T, Miller J, Elwood SE, Platts-Mills JA. Estimated global and regional causes of deaths from diarrhoea in children younger than 5 years during 2000-21: a systematic review and Bayesian multinomial analysis. Lancet Glob Health 2024; 12:e919-e928. [PMID: 38648812 PMCID: PMC11099298 DOI: 10.1016/s2214-109x(24)00078-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Information on the causes of deaths from diarrhoea in children younger than 5 years is needed to design improved preventive and therapeutic approaches. We aimed to conduct a systematic analysis of studies to report estimates of the causes of deaths from diarrhoea in children younger than 5 years at global and regional levels during 2000-21. METHODS For this systematic review and Bayesian multinomial analysis, we included 12 pathogens with the highest attributable incidence in the Global Enteric Multicenter Study. We searched PubMed, Scopus, Embase, Web of Science, Global Health Index Medicus, Global Health OVID, IndMed, Health Information Platform for the Americas (PLISA), Africa-Wide Information, and Cochrane Collaboration for articles published between Jan 1, 2000, and Dec 31, 2020, using the search terms "child", "hospital", "diarrhea", "diarrhoea", "dysentery", "rotavirus", "Escherichia coli", "salmonella", "shigella", "campylobacter", "Vibrio cholerae", "cryptosporidium", "norovirus", "astrovirus", "sapovirus", and "adenovirus". To be included, studies had to have a patient population of children younger than 5 years who were hospitalised for diarrhoea (at least 90% of study participants), at least a 12-month duration, reported prevalence in diarrhoeal stools of at least two of the 12 pathogens, all patients with diarrhoea being included at the study site or a systematic sample, at least 100 patients with diarrhoea, laboratory tests done on rectal swabs or stool samples, and standard laboratory methods (ie, quantitative PCR [qPCR] or non-qPCR). Studies published in any language were included. Studies were excluded if they were limited to nosocomial, chronic, antibiotic-associated, or outbreak diarrhoea or to a specific population (eg, only children with HIV or AIDS). Each article was independently reviewed by two researchers; a third arbitrated in case of disagreement. If both reviewers identified an exclusion criterion, the study was excluded. Data sought were summary estimates. Data on causes from published studies were adjusted when necessary to account for the poor sensitivity of non-qPCR methods and for attributable fraction based on quantification of pathogens in children who are ill or non-ill. The causes of deaths from diarrhoea were modelled on the causes of hospitalisations for diarrhoea. We separately modelled studies reporting causes of diarrhoea in children who were hospitalised in low-income and middle-income countries (LMICs) and in high-income countries (HICs). FINDINGS Of 74 282 papers identified in the initial database search, we included 138 studies (91 included data from LMICs and 47 included data from HICs) from 73 countries. We modelled estimates for 194 WHO member states (hereafter referred to as countries), including 42 HICs and 152 LMICs. We could attribute a cause to 1 003 448 (83·8%) of the estimated 1 197 044 global deaths from diarrhoea in children younger than 5 years in 2000 and 360 730 (81·3%) of the estimated 443 833 global deaths from diarrhoea in children younger than 5 years in 2021. The cause with the largest estimated global attribution was rotavirus; in LMICs, the proportion of deaths from diarrhoea due to rotavirus in children younger than 5 years appeared lower in 2021 (108 322 [24·4%] of 443 342, 95% uncertainty interval 21·6-29·5) than in 2000 (316 382 [26·5%] of 1 196 134, 25·7-28·5), but the 95% CIs overlapped. In 2000, the second largest estimated attribution was norovirus GII (95 817 [8·0%] of 1 196 134 in LMICs and 225 [24·7%] of 910 in HICs); in 2021, Shigella sp had the second largest estimated attribution in LMICs (36 082 [8·1%] of 443 342), but norovirus remained with the second largest estimated attribution in HICs (84 [17·1%] of 490). INTERPRETATION Our results indicate progress in the reduction of deaths from diarrhoea caused by 12 pathogens in children younger than 5 years in the past two decades. There is a need to increase efforts for prevention, including with rotavirus vaccine, and treatment to eliminate further deaths. FUNDING Bill & Melinda Gates Foundation via Johns Hopkins University and the University of Virginia.
Collapse
Affiliation(s)
- Robert E Black
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| | - Jamie Perin
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Diana Yeung
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Tushara Rajeev
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Jacob Miller
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sarah E Elwood
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
24
|
Sharma PC, McCandless M, Sontakke SP, Varshney N, Brodell RT, Kyle PB, Daley W. Navigating Viral Gastroenteritis: Epidemiological Trends, Pathogen Analysis, and Histopathological Findings. Cureus 2024; 16:e61197. [PMID: 38939260 PMCID: PMC11210331 DOI: 10.7759/cureus.61197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Gastroenteritis is a common cause of morbidity and mortality globally. Its cause encompasses a spectrum of agents, including viruses, bacteria, parasites, toxins, and drugs. Viruses account for a considerable portion of gastroenteritis cases across all age groups, typically presenting with symptoms like nausea, vomiting, diarrhea, dehydration, anorexia, and weight loss. While sporadic cases occur, viral gastroenteritis is more frequently observed in outbreaks within closely knit communities such as daycare facilities, nursing homes, and cruise ships. Therefore, it becomes necessary to determine when healthcare providers should consider this condition in their differential diagnosis and to develop the most effective strategy to confirm the diagnosis. METHODS De-identified data of patients with gastroenteritis were collected over a five-year period utilizing the Patient Cohort Explorer, an electronic health record at the University of Mississippi Medical Center. Confirmatory laboratory tests employed the BioFire® FilmArray® multiplex polymerase chain reaction for gastrointestinal pathogens. Out of the 22 most common agents associated with gastroenteritis, only viral pathogens, specifically adenovirus, astrovirus, norovirus, rotavirus, and sapovirus, were included in the analysis. When available, histopathology was reviewed. RESULTS Among the various causes of gastroenteritis, both infectious and non-infectious, our findings revealed that 25.46% of the cases were linked to viral pathogens. This included a significantly higher percentage of pediatric patients (72.73%) when compared to adults (27.07%), with a p-value of 0.015. Norovirus genogroups I and II emerged as the most frequently detected viruses across all age groups, with a significant prevalence among adults. No discernible gender-based differences were observed. The histopathological findings included inflammation, ulceration, erosion, architectural distortion, and the pathognomonic viral inclusion bodies associated with adenovirus. CONCLUSION Our comprehensive analysis of viral gastroenteritis cases highlights the substantial burden of this condition, particularly among pediatric patients. Norovirus emerges as a prevalent culprit which emphasizes the importance of vigilant surveillance and timely diagnosis, especially in settings where outbreaks are common.
Collapse
Affiliation(s)
- Poonam C Sharma
- Pathology, University of Mississippi Medical Center, Jackson, USA
| | | | - Sumit P Sontakke
- Medical Foundations, Ross University School of Medicine, Bridgetown, BRB
| | - Neha Varshney
- Pathology, University of Mississippi Medical Center, Jackson, USA
| | - Robert T Brodell
- Pathology and Dermatology, University of Mississippi Medical Center, Jackson, USA
| | - Patrick B Kyle
- Pathology, University of Mississippi Medical Center, Jackson, USA
| | - William Daley
- Pathology, University of Mississippi Medical Center, Jackson, USA
| |
Collapse
|
25
|
Park K, Shin BM. Comparative evaluation of two molecular multiplex syndromic panels with acute gastroenteritis. Diagn Microbiol Infect Dis 2024; 109:116211. [PMID: 38447492 DOI: 10.1016/j.diagmicrobio.2024.116211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 03/08/2024]
Abstract
We compared the Allplex Gastrointestinal V/B1/B2 Assays and Seeplex Diarrhea V/B1/B2 ACE Detection Assays in patients with acute gastroenteritis (AGE). Of the total 432 specimens, 48.8% and 54.9% samples were positive for any bacterial or viral target using Seeplex and Allplex, respectively (P = 0.002). The overall percent agreement (OPA) between the two panels was >95% and the lowest OPA was 95.4% for CdB. Allplex identified 40 samples positive for Salmonella spp., while Seeplex and OBC identified only 27 (67.5%) and 8 (20%), respectively. Shigella spp. were detected by assays in six samples, but none were identified using culture. Clostridium perfringens with Seeplex was detected in 70 (16.2%). It remained an informative species in identifying AGE although cpe gene showed only 9.8% positivity. Pathogenic Escherichia coli with Allplex could be detected in 40 (9.3%) samples, which could provide valuable information for the diagnosis of AGE.
Collapse
Affiliation(s)
- Kuenyoul Park
- Department of Laboratory Medicine, Sanggye Paik Hospital, School of Medicine, Inje University, 1342, Dongil-ro, Nowon-gu, Seoul 01757, Korea
| | - Bo-Moon Shin
- Department of Laboratory Medicine, Sanggye Paik Hospital, School of Medicine, Inje University, 1342, Dongil-ro, Nowon-gu, Seoul 01757, Korea.
| |
Collapse
|
26
|
Latifi T, Kachooei A, Jalilvand S, Zafarian S, Roohvand F, Shoja Z. Correlates of immune protection against human rotaviruses: natural infection and vaccination. Arch Virol 2024; 169:72. [PMID: 38459213 DOI: 10.1007/s00705-024-05975-y] [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: 08/12/2023] [Accepted: 12/12/2023] [Indexed: 03/10/2024]
Abstract
Species A rotaviruses are the leading viral cause of acute gastroenteritis in children under 5 years of age worldwide. Despite progress in the characterization of the pathogenesis and immunology of rotavirus-induced gastroenteritis, correlates of protection (CoPs) in the course of either natural infection or vaccine-induced immunity are not fully understood. There are numerous factors such as serological responses (IgA and IgG), the presence of maternal antibodies (Abs) in breast milk, changes in the intestinal microbiome, and rotavirus structural and non-structural proteins that contribute to the outcome of the CoP. Indeed, while an intestinal IgA response and its surrogate, the serum IgA level, are suggested as the principal CoPs for oral rotavirus vaccines, the IgG level is more likely to be a CoP for parenteral non-replicating rotavirus vaccines. Integrating clinical and immunological data will be instrumental in improving rotavirus vaccine efficacy, especially in low- and middle-income countries, where vaccine efficacy is significantly lower than in high-income countries. Further knowledge on CoPs against rotavirus disease will be helpful for next-generation vaccine development. Herein, available data and literature on interacting components and proposed CoPs against human rotavirus disease are reviewed, and limitations and gaps in our knowledge in this area are discussed.
Collapse
Affiliation(s)
- Tayebeh Latifi
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, USA
| | - Atefeh Kachooei
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
- Department of Virology, Faculty 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
| | - Saman Zafarian
- Department of Microbial Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
- Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
27
|
Vicente B, Freitas AD, Freitas M, Midlej V. Systematic Review of Diagnostic Approaches for Human Giardiasis: Unveiling Optimal Strategies. Diagnostics (Basel) 2024; 14:364. [PMID: 38396402 PMCID: PMC10887752 DOI: 10.3390/diagnostics14040364] [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: 01/08/2024] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Giardiasis, caused by the protozoan Giardia intestinalis, affects around 400 million people worldwide, emphasizing the critical need for accurate diagnosis to enhance human health, especially in children. Prolonged giardiasis in childhood can lead to intellectual deficits and other complications. A variety of diagnostic tools, including microscopic, immunological, and molecular methods, are available for detecting G. intestinalis infection. Choosing the most suitable method can be challenging due to the abundance of options. This systematic review assesses the reliability and applicability of these diagnostic modalities. Utilizing the Dimensions and Wordart platforms for data analysis, we focus on relevant literature addressing diagnostic methods for human giardiasis. Microscopic techniques, particularly Ritchie's method, emerge as the primary choice, followed by enzyme-linked immunosorbent assay (ELISA) and polymerase chain reaction (PCR). PCR's limited use is attributed to its high cost and infrastructure challenges in developing nations. In conclusion, our analysis supports microscopic methods as the gold standard for giardiasis diagnosis. However, in cases where symptoms persist despite a negative diagnosis, employing more sensitive diagnostic approaches is advisable.
Collapse
Affiliation(s)
- Bruno Vicente
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-900, Brazil; (B.V.); (A.D.F.); (M.F.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-900, Brazil
| | - Anna De Freitas
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-900, Brazil; (B.V.); (A.D.F.); (M.F.)
- Programa de Pós-Graduação em Biologia Parasitária, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-900, Brazil
| | - Marcus Freitas
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-900, Brazil; (B.V.); (A.D.F.); (M.F.)
| | - Victor Midlej
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro 21040-900, Brazil; (B.V.); (A.D.F.); (M.F.)
| |
Collapse
|
28
|
Lee TA, Chen CY, Chang YJ, Guo BC, Lin WY, Wu CH, Wu HP. Presentations of Children with Suspected Sepsis Caused by Acute Infectious Diarrhea in the Pediatric Emergency Department. CHILDREN (BASEL, SWITZERLAND) 2024; 11:171. [PMID: 38397283 PMCID: PMC10887777 DOI: 10.3390/children11020171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Acute infectious diarrhea is a common cause of hospitalization in children. Hence, early identification of acute bacterial gastroenteritis with suspected sepsis in pediatric emergency departments (EDs) is important. This study aimed to describe the clinical spectrum and initial characteristics of children who were presented to a pediatric ED with acute infectious diarrhea and suspected sepsis. METHODS Between April 2020 to March 2021, children with clinical diagnoses of acute bacterial colitis and suspected sepsis who were admitted to the pediatric ED were prospectively enrolled. The following data were obtained and compared between different age groups of children: including demographics, presentation, laboratory tests, culture results, treatment modalities, complications, and short-term outcomes. RESULTS A total of 105 patients (70 males and 35 females; mean age: 3.75 ± 3.52 years) were enrolled in this study. Of them, 89 (84.8%) patients were <6 years of age, and 80 (76.2%) patients required hospitalization for a duration of 4.7 ± 2.08 days. C-reactive protein (CRP) and procalcitonin (PCT) levels were significantly higher in the admission (both p < 0.001) and anti-biotic treatment groups (both p < 0.001). Salmonella enteritidis was the most common organism cultured from the stool and blood samples (39 of 91 (38.5%) and 2 of 105 (1.9%), respectively). CONCLUSIONS The primary causative organism of acute infectious diarrhea identified in this study was S. enteritidis. Age and elevated serum CRP or PCT levels could be important factors in the decisions of emergency physicians regarding hospitalization and antibiotic therapies for pediatric acute infectious diarrhea.
Collapse
Affiliation(s)
- Tai-An Lee
- Department of Emergency Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua 50544, Taiwan;
| | - Chun-Yu Chen
- Department of Emergency Medicine, Tungs’ Taichung MetroHarbor Hospital, Taichung 43503, Taiwan;
- Department of Nursing, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 35664, Taiwan
| | - Yu-Jun Chang
- Laboratory of Epidemiology and Biostastics, Changhua Christian Hospital, Changhua 50006, Taiwan;
| | - Bei-Cyuan Guo
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan;
| | - Wen-Ya Lin
- Department of Pediatric Emergency Medicine, Taichung Veteran General Hospital, Taichung 43503, Taiwan;
- Department of Pediatrics, Taichung Veteran General Hospital, Taichung 43503, Taiwan
| | - Chao-Hsin Wu
- Department of Emergency Medicine, Tungs’ Taichung MetroHarbor Hospital, Taichung 43503, Taiwan;
- Department of Post-Baccalaureate Medicine, National Chung Hsing University, Taichung 40202, Taiwan
| | - Han-Ping Wu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| |
Collapse
|
29
|
Kombat MY, Kushitor SB, Sutherland EK, Boateng MO, Manortey S. Prevalence and predictors of diarrhea among children under five in Ghana. BMC Public Health 2024; 24:154. [PMID: 38212722 PMCID: PMC10782682 DOI: 10.1186/s12889-023-17575-7] [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: 07/17/2023] [Accepted: 12/24/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Globally, childhood diarrhea is a major public health concern. Despite numerous interventions that have been put in place to reduce its incidence over the years, childhood diarrhea remains a problem and is the fourth leading cause of child mortality in Ghana. This study examined the predictors of diarrhea among children under the age of five in Ghana. METHODS Data from the 2014 Ghana Demographic and Health survey, a cross-sectional survey, was used for the purpose of this study. A total of 2,547 children under the age of five were included in this study. Logistic regression analysis was performed to establish the factors associated with childhood diarrhea and ascertain explanatory variables. RESULTS The prevalence of diarrhea was 11.7%. Male children (13.4%) and those living in rural areas (12%), particularly in the Brong Ahafo region (17%) recorded the highest prevalence of diarrhea. Children aged 6 to 35 months of age, maternal age and education, sex of children and region of residence were the predictors of diarrhea among children under the age of five years in this study. CONCLUSION To lessen the prevalence of diarrhea among children under five in Ghana, existing interventions must be evaluated in the context of the predictors identified. Based on observations deduced from this study, the Ministry of Health, Ghana Health Service and other health regulatory agencies should intensify monitoring and awareness in the various regions, particularly in the transition and savannah zones on the causes, risk factors, and methods of preventing diarrhea in children under five. Various stakeholders including government and non-governmental organizations should take into account the predictors of diarrhea identified in the design of interventions to effectively reduce morbidity and mortality associated with childhood diarrhea.
Collapse
Affiliation(s)
| | - Sandra Boatemaa Kushitor
- Department of Community Health, Ensign Global College, Kpong, Ghana
- Department of Food Science and Centre for Sustainability Transitions, Stellenbosch University, Stellenbosch, South Africa
| | - Edward Kofi Sutherland
- Department of Community Health, Ensign Global College, Kpong, Ghana
- The Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | | | - Stephen Manortey
- Department of Community Health, Ensign Global College, Kpong, Ghana
| |
Collapse
|
30
|
Gobert AP, Hawkins CV, Williams KJ, Snyder LA, Barry DP, Asim M, Allaman MM, McNamara KM, Delgado AG, Wang Y, Zhao S, Rose KL, Piazuelo MB, Wilson KT. Hypusination in intestinal epithelial cells protects mice from infectious colitis. Gut Microbes 2024; 16:2438828. [PMID: 39673545 DOI: 10.1080/19490976.2024.2438828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is a bacterium that causes attaching/effacing (A/E) lesions and serious diarrheal disease, a major health issue in developing countries. EPEC pathogenicity results from the effect of virulence factors and dysregulation of host responses. Polyamines, including spermidine, play a major role in intestinal homeostasis. Spermidine is the substrate for deoxyhypusine synthase (DHPS), which catalyzes the conjugation of the amino acid hypusine to eukaryotic translation initiation factor 5A (EIF5A); hypusinated EIF5A (EIF5AHyp) binds specific mRNAs and initiates translation. Our aim was to determine the role of hypusination during infection with A/E pathogens. We found that DHPS and EIF5AHyp levels are induced in i) a colonic epithelial cell line and human-derived colon organoids infected with EPEC, and ii) the colon of mice infected with Citrobacter rodentium, the rodent equivalent of EPEC. Specific deletion of Dhps in intestinal epithelial cells worsened clinical, histological, and pro-inflammatory parameters in C. rodentium-infected mice. These animals also exhibited an exacerbated pathogenic transcriptome in their colon. Furthermore, infected mice with specific Dhps deletion exhibited reduced levels of proteins involved in detoxification of tissue-damaging reactive aldehydes and consequently increased electrophile adducts in the colon. Thus, hypusination in intestinal epithelial cells protects from infectious colitis mediated by A/E pathogens.
Collapse
Affiliation(s)
- Alain P Gobert
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caroline V Hawkins
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kamery J Williams
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lydia A Snyder
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel P Barry
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Asim
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Margaret M Allaman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kara M McNamara
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberto G Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yu Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kristie L Rose
- Department of Biochemistry, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - M Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
31
|
Mohammad N, Tak V, Bohra GK, Gadepalli R, Sharma A, Singh K, Nag VL. Molecular appraisal of Giardia intestinalis from Western India: A prospective observational study. Trop Parasitol 2024; 14:36-44. [PMID: 38444794 PMCID: PMC10911180 DOI: 10.4103/tp.tp_44_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/24/2023] [Accepted: 11/03/2023] [Indexed: 03/07/2024] Open
Abstract
Background Giardia intestinalis is an intestinal protozoan which commonly causes parasitic gastroenteritis globally. It is a species complex consisting of at least eight assemblages (genotypes). In India, Giardia is mostly underreported and missed in asymptomatic cases. Aim The aim of this study was to genotype the G. intestinalis isolates from stool samples of patients at a tertiary care center in Rajasthan, India, and to clinically correlate it. Methods This prospective pilot cross-sectional study was conducted from 2019 to 2021 in a tertiary care center in western India. Patients who were microscopically positive for giardiasis were enrolled. DNA was extracted from their stool samples and amplified by polymerase chain reaction (PCR) using 4E1-HP as the target sequence. Anthropometric measurements and analysis were done for children by using Anthrocal application. Results A total of 50 patients were enrolled. Diarrhea was present in 18 patients (36%). Among these, 6 were immunocompromised and had different comorbidities. Among the children <12 years of age, 55.17% (n = 16/29) were stunted (<-2 S.D.), and among <5 years, 44.4% (n = 4/9) showed wasting (<-2 S.D.). A PCR product corresponding to assemblage B of G. intestinalis was amplified in 47 stool specimens. Only three stool samples were negative for both assemblages A and B and posed an interesting enigma. Conclusion In this study, a predominance of assemblage B of G. intestinalis was detected in 94% of the isolates. Furthermore, the possibility of zoonotic transmission could not be ruled out.
Collapse
Affiliation(s)
- Naila Mohammad
- Department of Microbiology, AIIMS, Jodhpur, Rajasthan, India
| | - Vibhor Tak
- Department of Microbiology, AIIMS, Jodhpur, Rajasthan, India
| | | | | | - Anuradha Sharma
- Department of Microbiology, AIIMS, Jodhpur, Rajasthan, India
| | - Kuldeep Singh
- Department of Paediatrics, AIIMS, Jodhpur, Rajasthan, India
| | | |
Collapse
|
32
|
Ando H, Ahmed W, Okabe S, Kitajima M. Tracking the effects of the COVID-19 pandemic on viral gastroenteritis through wastewater-based retrospective analyses. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:166557. [PMID: 37633393 DOI: 10.1016/j.scitotenv.2023.166557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
The COVID-19 pandemic possibly disrupted the circulation and seasonality of gastroenteritis viruses (e.g., Norovirus (NoV), Sapovirus (SaV), group A rotavirus (ARoV), and Aichivirus (AiV)). Despite the growing application of wastewater-based epidemiology (WBE), there remains a lack of sufficient investigations into the actual impact of the COVID-19 pandemic on the prevalence of gastroenteritis viruses. In this study, we measured NoV GI and GII, SaV, ARoV, and AiV RNA concentrations in 296 influent wastewater samples collected from three wastewater treatment plants (WWTPs) in Sapporo, Japan between October 28, 2018 and January 12, 2023 using the highly sensitive EPISENS™ method. The detection ratios of SaV and ARoV after May 2020 (SaV: 49.8 % (134/269), ARoV: 57.4 % (151/263)) were significantly lower than those before April 2020 (SaV: 93.9 % (31/33), ARoV: 97.0 % (32/33); SaV: p < 3.5×10-7, ARoV: p < 1.5×10-6). Furthermore, despite comparable detection ratios before (88.5 %, 23/26) and during (66.7 %, 80/120) the COVID-19 pandemic (p = 0.032), the concentrations of NoV GII revealed a significant decrease after the onset of the pandemic (p < 1.5×10-7, Cliff's delta = 0.72). NoV GI RNA were sporadically detected (24.7 %, 8/33) before April 2020 and after May 2020 (6.5 %, 17/263), whereas AiV was consistently (100 %, 33/33) detected from wastewater throughout the study period (95.8 %, 252/263). The WBE results demonstrated the significant influence of COVID-19 countermeasures on the circulation of gastroenteritis viruses, with variations observed in the magnitude of their impact across different types of viruses. These epidemiological findings highlight that the hygiene practices implemented to prevent COVID-19 infections may also be effective for controlling the prevalence of gastroenteritis viruses, providing invaluable insights for public health units and the development of effective disease management guidelines.
Collapse
Affiliation(s)
- Hiroki Ando
- Division of Environmental Engineering, Faculty of Engineering, Hokkaido University, North 13 West 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| | - Warish Ahmed
- CSIRO Land and Water, Ecosciences Precinct, 41 Boggo Road, QLD 4102, Australia
| | - Satoshi Okabe
- Division of Environmental Engineering, Faculty of Engineering, Hokkaido University, North 13 West 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| | - Masaaki Kitajima
- Division of Environmental Engineering, Faculty of Engineering, Hokkaido University, North 13 West 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan.
| |
Collapse
|
33
|
Sarmento SK, de Andrade JDSR, Malta FC, Fialho AM, Mello MDS, Burlandy FM, Fumian TM. Norovirus Epidemiology and Genotype Circulation during the COVID-19 Pandemic in Brazil, 2019-2022. Pathogens 2023; 13:3. [PMID: 38276149 PMCID: PMC10818385 DOI: 10.3390/pathogens13010003] [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: 11/23/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/27/2024] Open
Abstract
Norovirus stands out as a leading cause of acute gastroenteritis (AGE) worldwide, affecting all age groups. In the present study, we investigated fecal samples from medically attended AGE patients received from nine Brazilian states, from 2019 to 2022, including the COVID-19 pandemic period. Norovirus GI and GII were detected and quantified using RT-qPCR, and norovirus-positive samples underwent genotyping through sequencing the ORF1/2 junction region. During the four-year period, norovirus prevalence was 37.2%, varying from 20.1% in 2020 to 55.4% in 2021. GII genotypes dominated, being detected in 92.9% of samples. GII-infected patients had significantly higher viral concentrations compared to GI-infected patients (median of 3.8 × 107 GC/g and 6.7 × 105 GC/g, respectively); and patients aged >12-24 months showed a higher median viral load (8 × 107 GC/g) compared to other age groups. Norovirus sequencing revealed 20 genotypes by phylogenetic analysis of RdRp and VP1 partial regions. GII.4 Sydney[P16] was the dominant genotype (57.3%), especially in 2019 and 2021, followed by GII.2[P16] (14.8%) and GII.6[P7] (6.3%). The intergenogroup recombinant genotype, GIX.1[GII.P15], was detected in five samples. Our study is the first to explore norovirus epidemiology and genotype distribution in Brazil during COVID-19, and contributes to understanding the epidemiological dynamics of norovirus and highlighting the importance of continuing to follow norovirus surveillance programs in Brazil.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tulio Machado Fumian
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21045-900, RJ, Brazil (F.M.B.)
| |
Collapse
|
34
|
Gharbi-Khelifi H, Jmii H, Mosbahi M, Hamdi S, Hamdi R, Brahmi J, Loukil S, Chamkha M, Sayadi S, Aouni M, Barreiro A, Fernández-Sanjurjo MJ, Núñez-Delgado A, Rodríguez EÁ. Microbiological and physicochemical quality enhancement of treated wastewater using raw and chemically modified clays from Sidi Bouzid region, Tunisia. ENVIRONMENTAL RESEARCH 2023; 239:117391. [PMID: 37852463 DOI: 10.1016/j.envres.2023.117391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Environmental discharge of wastewater represents a source of chemical and biological pollutants. This study firstly evaluates the microbiological and physicochemical quality of treated wastewaters collected from two wastewater treatment plants (WWTPs) located in two different Tunisian cities namely Sidi Bouzid (SB) and Gafsa (G). Then, the capacity of three raw and acid/base-activated local clays to enhance the quality of wastewaters was assessed. The results indicate that the quantities of enteric bacteria (oscillating from 1.381 × 103 to 1.4 × 108 CFU/100 mL), fungi (between 1.331 × 103 and 1.781 × 104 CFU/100 mL), as well as SARS-CoV-2 (between 4.25 × 103 and 5.05 × 105 CFU/100 mL) and Hepatitis A virus RNA (form 4.25 × 103 to 7.4 × 104 CFU/100 mL) detected in effluent wastewaters were not in compliance with the Tunisian standards for both studied WWTPs. Likewise for other indicators such as electrical conductivity (ranging 4.9-5.4 mS/cm), suspended matter (145-160 g l-1), chemical oxygen demand (123-160 mg l-1), biological oxygen demand 5 (172-195 mg l-1), chloride, Total Kjeldahl nitrogen (TKN) and phosphorus contents (710, 58-66 and 9.47-10.83 mg l-1 respectively), the registered values do not agree with the set standards established for wastewater treatment. On the other hand, the pH values fitted (oscillating from 6.86 (at G) to 7.24 (at SB) with the Tunisian standards for both WWTPs. After treatment, wastewaters showed better values for the microbiological parameters, especially for the clays designed as AM and HJ1, which eliminated 100% of viruses. In addition, when acid-activated AM clays were applied, a marked improvement in the quality of physicochemical parameters was obtained, especially for suspended matter (2 and 4 g l-1 for SB and G, respectively), TKN (5.2 (SB) and 6.40 (G) mg/l), phosphorus (1.01 (SB) and 0.81 (G) mg/l). Our results open perspectives for the possibility of efficiently using these specific clays in the enhancement of the quality of treated wastewaters.
Collapse
Affiliation(s)
- Hakima Gharbi-Khelifi
- LR99-ES27, Faculty of Pharmacy of Monastir, University of Monastir, Avenue Avicenne, 5000 Monastir, Tunisia; Department of Biotechnology, Faculty of Science and Technology of Sidi Bouzid, University of Kairouan, 9100 Sidi Bouzid, Tunisia.
| | - Habib Jmii
- LR99-ES27, Faculty of Pharmacy of Monastir, University of Monastir, Avenue Avicenne, 5000 Monastir, Tunisia
| | - Mohamed Mosbahi
- Laboratory of Georessources CERTE, Technopole Borj Cedria, Tunisia
| | - Samiha Hamdi
- Department of Biotechnology, Faculty of Science and Technology of Sidi Bouzid, University of Kairouan, 9100 Sidi Bouzid, Tunisia; Department of Soil Science and Agricultural Chemistry, Engineering Polytechnic School, University of Santiago de Compostela, 27002 Lugo, Spain
| | - Rawand Hamdi
- LR99-ES27, Faculty of Pharmacy of Monastir, University of Monastir, Avenue Avicenne, 5000 Monastir, Tunisia
| | - Jihen Brahmi
- Department of Biotechnology, Faculty of Science and Technology of Sidi Bouzid, University of Kairouan, 9100 Sidi Bouzid, Tunisia
| | - Slim Loukil
- Laboratory of Environmental Bioprocesses, Centre of Biotechnology of Sfax, Sfax University, PO Box 1177, 3018 Sfax, Tunisia
| | - Mohamed Chamkha
- Laboratory of Environmental Bioprocesses, Centre of Biotechnology of Sfax, Sfax University, PO Box 1177, 3018 Sfax, Tunisia
| | - Sami Sayadi
- Biotechnology Program, Center for Sustainable Development, College of Arts and Sciences, Qatar University, Doha 2713, Qatar
| | - Mahjoub Aouni
- LR99-ES27, Faculty of Pharmacy of Monastir, University of Monastir, Avenue Avicenne, 5000 Monastir, Tunisia
| | - Ana Barreiro
- Department of Soil Science and Agricultural Chemistry, Engineering Polytechnic School, University of Santiago de Compostela, 27002 Lugo, Spain
| | - Maria J Fernández-Sanjurjo
- Department of Soil Science and Agricultural Chemistry, Engineering Polytechnic School, University of Santiago de Compostela, 27002 Lugo, Spain
| | - Avelino Núñez-Delgado
- Department of Soil Science and Agricultural Chemistry, Engineering Polytechnic School, University of Santiago de Compostela, 27002 Lugo, Spain
| | - Esperanza Álvarez Rodríguez
- Department of Soil Science and Agricultural Chemistry, Engineering Polytechnic School, University of Santiago de Compostela, 27002 Lugo, Spain
| |
Collapse
|
35
|
Islam D, Ruamsap N, Imerbsin R, Khanijou P, Gonwong S, Wegner MD, McVeigh A, Poly FM, Crawford JM, Swierczewski BE, Kaminski RW, Laird RM. Bioactivity and efficacy of a hyperimmune bovine colostrum product- Travelan, against shigellosis in a non-Human primate model (Macaca mulatta). PLoS One 2023; 18:e0294021. [PMID: 38091314 PMCID: PMC10718440 DOI: 10.1371/journal.pone.0294021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 10/16/2023] [Indexed: 12/18/2023] Open
Abstract
Infectious diarrhea is a World Health Organization public health priority area due to the lack of effective vaccines and an accelerating global antimicrobial resistance crisis. New strategies are urgently needed such as immunoprophylactic for prevention of diarrheal diseases. Hyperimmune bovine colostrum (HBC) is an established and effective prophylactic for infectious diarrhea. The commercial HBC product, Travelan® (Immuron Ltd, Australia) targets multiple strains of enterotoxigenic Escherichia coli (ETEC) is highly effective in preventing diarrhea in human clinical studies. Although Travelan® targets ETEC, preliminary studies suggested cross-reactivity with other Gram-negative enteric pathogens including Shigella and Salmonella species. For this study we selected an invasive diarrheal/dysentery-causing enteric pathogen, Shigella, to evaluate the effectiveness of Travelan®, both in vitro and in vivo. Here we demonstrate broad cross-reactivity of Travelan® with all four Shigella spp. (S. flexneri, S. sonnei, S. dysenteriae and S. boydii) and important virulence factor Shigella antigens. Naïve juvenile rhesus macaques (NJRM) were randomized, 8 dosed with Travelan® and 4 with a placebo intragastrically twice daily over 6 days. All NJRM were challenged with S. flexneri 2a strain 2457T on the 4th day of treatment and monitored for diarrheal symptoms. All placebo-treated NJRM displayed acute dysentery symptoms within 24-36 hours of challenge. Two Travelan®-treated NJRM displayed dysentery symptoms and six animals remained healthy and symptom-free post challenge; resulting in 75% efficacy of prevention of shigellosis (p = 0.014). These results strongly indicate that Travelan® is functionally cross-reactive and an effective prophylactic for shigellosis. This has positive implications for the prophylactic use of Travelan® for protection against both ETEC and Shigella spp. diarrheal infections. Future refinement and expansion of pathogens recognized by HBC including Travelan® could revolutionize current management of gastrointestinal infections and outbreaks in travelers' including military, peacekeepers, humanitarian workers and in populations living in endemic regions of the world.
Collapse
Affiliation(s)
- Dilara Islam
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Nattaya Ruamsap
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Rawiwan Imerbsin
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Patchariya Khanijou
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Siriphan Gonwong
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Matthew D. Wegner
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Annette McVeigh
- Henry M. Jackson Foundation for Military Medicine (HJF), Bethesda, Maryland, United States of America
- Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Frédéric M. Poly
- Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - John M. Crawford
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Brett E. Swierczewski
- US Army Medical Directorate of the Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Robert W. Kaminski
- Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, United States of America
| | - Renee M. Laird
- Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, United States of America
| |
Collapse
|
36
|
Guo H, Liang J, Lin H, Zhou X, Zhang Z, Lai L, Zhang T, Wang Z, Zhou J, Sun J, Liao J, Jiang M, Yang Z. Differentiate Clinical Characteristics Between Viral Pneumonia and Mycoplasma pneumoniae and Nomograms for Predicting Mycoplasma pneumoniae : A Retrospective Study in Primary Hospitals. Pediatr Infect Dis J 2023; 42:1035-1040. [PMID: 37820276 PMCID: PMC10629606 DOI: 10.1097/inf.0000000000004082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 10/13/2023]
Abstract
OBJECTIVE To identify the difference in clinical characteristics between viral pneumonia and Mycoplasma pneumoniae , providing cues on their differential diagnosis for primary hospitals with the insufficient pathogen detection capacity. METHODS We retrospectively reviewed the medical records of hospitalized children with acute respiratory tract infections, and pathogenic microbes test results were analyzed. Clinical characteristics, routine blood parameters and hospitalization duration and fee were compared between M. pneumoniae and viral pneumonia. We used in the multivariable logistic regression to predict the probability of children with M. pneumoniae and graphically represented by a dynamic nomogram. The discrimination and clinical utility of the model were confirmed by receiver operating characteristic and decision curve analysis curves. RESULT A total of 375 children with community-acquired pneumonia were included. Mycoplasma infection accounted for the largest proportion (22.13%). The incidence of both hypothermia and vomiting was lower in M. pneumoniae compared to viral pneumonia (hypothermia: 10.50% vs. 0.00%; vomiting: 7.90% vs. 0.00%). The prevalence of hyperthermia was higher in M. pneumoniae (hyperthermia: 89.5% vs. 100%). Procalcitonin, peripheral blood white blood cell count and lymphocyte levels were higher in the viral pneumonia group, and eosinophil levels were conversely lower. As for the duration of illness, the mean length of stay was 5.20 ± 2.12 (viral pneumonia) and 6.27 ± 2.48 days ( M. pneumoniae ). Children with M. pneumoniae had higher overall hospital costs and required more medical treatment. The above were all statistically significant with a P < 0.05. The scoring system was established based on the above results. Receiver operating characteristic curves showed good model-discrimination ability with 0.844 of the area under the curve in the training set and 0.778 in the test set. Decision curve analysis curves demonstrated the discriminative superiority of this model. The web-based dynamic nomogram calculator is accessible at https://zhxylxy0160128.shinyapps.io/Nomogram/ . CONCLUSION Nomograms have satisfactory discrimination, and clinical utility may benefit in predicting the probability of developing M. pneumoniae in children. Children with M. pneumoniae have a higher burden than those with viral pneumonia and may require more intensive in-hospital monitoring.
Collapse
Affiliation(s)
- Huixian Guo
- From the Department of Respiration, Guangzhou Yuexiu District Children’s Hospital, Guangzhou, China
| | - Jingyi Liang
- Department of Respiratory Research, National Center for Respiratory medicine, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Haowen Lin
- Department of Clinical Medicine, The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xingyou Zhou
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhou Zhang
- From the Department of Respiration, Guangzhou Yuexiu District Children’s Hospital, Guangzhou, China
| | - Laiqing Lai
- From the Department of Respiration, Guangzhou Yuexiu District Children’s Hospital, Guangzhou, China
| | - Tao Zhang
- From the Department of Respiration, Guangzhou Yuexiu District Children’s Hospital, Guangzhou, China
| | - Zhufeng Wang
- Department of Respiratory Research, National Center for Respiratory medicine, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Junhou Zhou
- Department of Respiratory Research, National Center for Respiratory medicine, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Jiaxi Sun
- Department of Respiratory Research, National Center for Respiratory medicine, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Jiayi Liao
- From the Department of Respiration, Guangzhou Yuexiu District Children’s Hospital, Guangzhou, China
| | - Mei Jiang
- Department of Respiratory Research, National Center for Respiratory medicine, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zifeng Yang
- Department of Respiratory Research, National Center for Respiratory medicine, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
37
|
Zenebe T, Eguale T, Desalegn Z, Beshah D, Gebre-Selassie S, Mihret A, Abebe T. Distribution of ß-Lactamase Genes Among Multidrug-Resistant and Extended-Spectrum ß-Lactamase-Producing Diarrheagenic Escherichia coli from Under-Five Children in Ethiopia. Infect Drug Resist 2023; 16:7041-7054. [PMID: 37954506 PMCID: PMC10637226 DOI: 10.2147/idr.s432743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023] Open
Abstract
Purpose Escherichia coli strains that produce extended-spectrum ß-lactamase (ESBL) and carbapenemase are among the major threats to global health. The objective of the present study was to determine the distribution of ß-lactamase genes among multidrug-resistant (MDR) and ESBL-producing Diarrheagenic E. coli (DEC) pathotypes isolated from under-five children in Ethiopia. Patients and Methods A cross-sectional study was conducted in Addis Ababa and Debre Berhan, Ethiopia. It was a health-facility-based study and conducted between December 2020 and August 2021. A total of 476 under-five children participated in the study. DEC pathotypes were detected by conventional Polymerase Chain Reaction (PCR) assay. After evaluating the antimicrobial susceptibility profile of the DEC strains by disk diffusion method, confirmation test was done for ESBL and carbapenemase production. ß-lactamase encoding genes were identified from phenotypically ESBLs and carbapenemase positive DEC strains using PCR assay. Results In total, 183 DEC pathotypes were isolated from the 476 under-five children. Seventy-nine (43%, 79/183) MDR-DEC pathotypes were identified. MDR was common among enteroaggregative E. coli (EAEC) (58%, 44/76), followed by enterotoxigenic E. coli (ETEC) (44%, 17/39)) and enteroinvasive E. coli (EIEC) (30%, 7/23). Phenotypically, a total of 30 MDR-DEC pathotypes (16.4%, 30/183) were tested positive for ESBLs. Few ETEC (5.1%, 2/39) and EAEC (2.6%, 2/76) were carbapenemase producers. The predominant β-lactamase genes identified was blaTEM (80%, 24/30) followed by blaCTX-M (73%, 22/30), blaSHV (60%, 18/30), blaNDM (13%, 4/30), and blaOXA-48 (13%, 4/30). Majority of the ß-lactamase encoding genes were detected in EAEC (50%) and ETEC (20%). Co-existence of different β-lactamase genes was found in the present study. Conclusion The blaTEM, blaCTX-M, blaSHV, blaNDM, and blaOXA-48, that are associated with serious and urgent threats globally, were detected in diarrheagenic E. coli isolates from under-five children in Ethiopia. This study also revealed the coexistence of the β-lactamase genes.
Collapse
Affiliation(s)
- Tizazu Zenebe
- Department of Microbiology, Immunology and Parasitology, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Medical Laboratory Science, Debre Berhan University, Debre Berhan, Ethiopia
| | - Tadesse Eguale
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
- Ohio State University, Global One Health LLC, Addis Ababa, Ethiopia
| | - Zelalem Desalegn
- Department of Microbiology, Immunology and Parasitology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Daniel Beshah
- Department of Medical Laboratory, Tikur Anbessa Specialized Hospital, Addis Ababa University, Addis Ababa, Ethiopia
| | - Solomon Gebre-Selassie
- Department of Microbiology, Immunology and Parasitology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Adane Mihret
- Department of Microbiology, Immunology and Parasitology, Addis Ababa University, Addis Ababa, Ethiopia
- Bacterial and Viral Disease Research Directorate, Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Tamrat Abebe
- Department of Microbiology, Immunology and Parasitology, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
38
|
Armah G, Lopman BA, Vinjé J, O'Ryan M, Lanata CF, Groome M, Ovitt J, Marshall C, Sajewski E, Riddle MS. Vaccine value profile for norovirus. Vaccine 2023; 41 Suppl 2:S134-S152. [PMID: 37951692 PMCID: PMC10710898 DOI: 10.1016/j.vaccine.2023.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/11/2023] [Accepted: 03/16/2023] [Indexed: 11/14/2023]
Abstract
Norovirus is attributed to nearly 1 out of every 5 episodes of diarrheal disease globally and is estimated to cause approximately 200,000 deaths annually worldwide, with 70,000 or more among children in developing countries. Noroviruses remain a leading cause of sporadic disease and outbreaks of acute gastroenteritis even in industrialized settings, highlighting that improved hygiene and sanitation alone may not be fully effective in controlling norovirus. Strengths in global progress towards a Norovirus vaccine include a diverse though not deep pipeline which includes multiple approaches, including some with proven technology platforms (e.g., VLP-based HPV vaccines). However, several gaps in knowledge persist, including a fulsome mechanistic understanding of how the virus attaches to human host cells, internalizes, and induces disease.
Collapse
Affiliation(s)
- George Armah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Ben A Lopman
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Jan Vinjé
- Division of Viral Diseases, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Miguel O'Ryan
- Microbiology and Mycology Program, Faculty of Medicine, University of Chile and Instituto de Sistemas Complejos de Ingenierìa (ISCI), Santiago, Chile
| | | | - Michelle Groome
- National Institute for Communicable Diseases, National Health Laboratory Services, Johannesburg, South Africa; School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jared Ovitt
- Office of Medical Research, University of Nevada, Reno School of Medicine, Reno, Nevada, USA
| | | | - Elizabeth Sajewski
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Mark S Riddle
- Office of Medical Research, University of Nevada, Reno School of Medicine, Reno, Nevada, USA.
| |
Collapse
|
39
|
Latifi T, Jalilvand S, Golsaz-Shirazi F, Arashkia A, Kachooei A, Afchangi A, Zafarian S, Roohvand F, Shoja Z. Characterization and immunogenicity of a novel chimeric hepatitis B core-virus like particles (cVLPs) carrying rotavirus VP8*protein in mice model. Virology 2023; 588:109903. [PMID: 37832344 DOI: 10.1016/j.virol.2023.109903] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/23/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
Given the efficacy and safety issues of the WHO for approved/prequalified live attenuated rotavirus (RV) vaccines, studies on alternative non-replicating modals and proper RV antigens are actively undertaken. Herein, we report the novel chimeric hepatitis B core-virus like particles (VLPs) carrying RV VP8*26-231 protein of a P [8] strain (cVLPVP8*), as a parenteral VLP RV vaccine candidate. SDS-PAGE and Western blotting analyses indicated the expected size of the E. coli-derived HBc-VP8* protein that self-assembled to cVLPVP8* particles. Immunization in mice indicated development of higher levels of IgG and IgA as well as higher IgG1/IgG2a ratios by cVLPVP8* vaccination compared to the VP8* alone. Assessment of neutralizing antibodies (nAbs) indicated development of heterotypic nAbs with cross-reactivity to a heterotypic RV strain by cVLPVP8* immunization compared to VP8* alone. The observed anti-VP8* cross-reactivity might indicate the possibility of developing a Pan-genomic RVA vaccine based on the cVLPVP8* formulation that deserves further challenge studies.
Collapse
Affiliation(s)
- Tayebeh Latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Somayeh Jalilvand
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Forough Golsaz-Shirazi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Arashkia
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Atefeh Kachooei
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atefeh Afchangi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Saman Zafarian
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Department of Microbial Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Zabihollah Shoja
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran; Research Center for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
40
|
Hogben E, Khamrin P, Kumthip K, Yodmeeklin A, Maneekarn N. Distribution and molecular characterization of saffold virus and human cosavirus in children admitted to hospitals with acute gastroenteritis in Thailand, 2017-2022. J Med Virol 2023; 95:e29159. [PMID: 37805831 DOI: 10.1002/jmv.29159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/29/2023] [Accepted: 09/21/2023] [Indexed: 10/09/2023]
Abstract
Saffold virus (SAFV) and human cosavirus (HCoSV) are emerging viruses of the Picornaviridae family. They have been shown to associate with gastrointestinal infection and more recently these viruses have also been demonstrated to associate with other clinical infections such as the respiratory tract, cardiovascular system, and the cerebral ventricular system. In this study, 2459 stool specimens collected from pediatric patients admitted to hospitals with acute gastroenteritis from January 2017 to December 2022, were screened for SAFV and HCoSV utilizing reverse transcription-polymerase chain reaction. Positive samples were then characterized into genotypes via nucleotide sequencing and bioinformatic analysis. Of the 2459 samples, 21 and 39 were positive for SAFV (0.9%) and HCoSV (1.6%), respectively. Three genotypes of SAFV were identified-SAFV-1 (38%), SAFV-2 (24%), and SAFV-3 (38%). Two genetic groups of HCoSV were identified-HCoSV-C (97%) and HCoSV-A (3%), demonstrating a large increase of HCoSV-C as compared to those reported previously from the same geographical region in Thailand. This study provides the prevalence of SAFV and HCoSV genotypes in Chiang Mai, Thailand during a period of 6 years from 2017 to 2022.
Collapse
Affiliation(s)
- Emily Hogben
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pattara Khamrin
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Emerging and Re-emerging Diarrheal Viruses, Chiang Mai University, Chiang Mai, Thailand
| | - Kattareeya Kumthip
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Emerging and Re-emerging Diarrheal Viruses, Chiang Mai University, Chiang Mai, Thailand
| | - Arpaporn Yodmeeklin
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Emerging and Re-emerging Diarrheal Viruses, Chiang Mai University, Chiang Mai, Thailand
| | - Niwat Maneekarn
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Emerging and Re-emerging Diarrheal Viruses, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
41
|
Prabakaran M, Weible LJ, Champlain JD, Jiang RY, Biondi K, Weil AA, Van Voorhis WC, Ojo KK. The Gut-Wrenching Effects of Cryptosporidiosis and Giardiasis in Children. Microorganisms 2023; 11:2323. [PMID: 37764167 PMCID: PMC10538111 DOI: 10.3390/microorganisms11092323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Cryptosporidium species and Giardia duodenalis are infectious intestinal protozoan pathogens that cause alarming rates of morbidity and mortality worldwide. Children are more likely to have clinical symptoms due to their less developed immune systems and factors such as undernutrition, especially in low- and middle-income countries. The severity of the symptoms and clinical manifestations in children may vary from asymptomatic to life-threatening depending on the Cryptosporidium species/G. duodenalis strains and the resulting complex stepwise interactions between the parasite, the host nutritional and immunologic status, and the gut microbiome profile. Structural damages inflicted by both parasites to epithelial cells in the large and small intestines could severely impair children's gut health, including the ability to absorb nutrients, resulting in stunted growth, diminished neurocognitive development, and other long-term effects. Clinically approved cryptosporidiosis and giardiasis drugs have broad antimicrobial effects that have incomprehensible impacts on growing children's gut health.
Collapse
Affiliation(s)
- Mayuri Prabakaran
- Center for Emerging and Reemerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (M.P.); (L.J.W.); (J.D.C.); (R.Y.J.); (A.A.W.); (W.C.V.V.)
| | - Lyssa J. Weible
- Center for Emerging and Reemerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (M.P.); (L.J.W.); (J.D.C.); (R.Y.J.); (A.A.W.); (W.C.V.V.)
| | - Joshua D. Champlain
- Center for Emerging and Reemerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (M.P.); (L.J.W.); (J.D.C.); (R.Y.J.); (A.A.W.); (W.C.V.V.)
| | - Ryan Ye Jiang
- Center for Emerging and Reemerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (M.P.); (L.J.W.); (J.D.C.); (R.Y.J.); (A.A.W.); (W.C.V.V.)
| | - Katalina Biondi
- Human Center for Artificial Intelligence, Department of Computer Science, University of Central Florida, Orlando, FL 32816, USA;
| | - Ana A. Weil
- Center for Emerging and Reemerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (M.P.); (L.J.W.); (J.D.C.); (R.Y.J.); (A.A.W.); (W.C.V.V.)
| | - Wesley C. Van Voorhis
- Center for Emerging and Reemerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (M.P.); (L.J.W.); (J.D.C.); (R.Y.J.); (A.A.W.); (W.C.V.V.)
| | - Kayode K. Ojo
- Center for Emerging and Reemerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (M.P.); (L.J.W.); (J.D.C.); (R.Y.J.); (A.A.W.); (W.C.V.V.)
| |
Collapse
|
42
|
Ranasinghe S, Aspinall S, Beynon A, Ash A, Lymbery A. Traditional medicinal plants in the treatment of gastrointestinal parasites in humans: A systematic review and meta-analysis of clinical and experimental evidence. Phytother Res 2023; 37:3675-3687. [PMID: 37230485 DOI: 10.1002/ptr.7895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/22/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
Gastrointestinal (GI) parasites cause significant morbidity and mortality worldwide. The use of conventional antiparasitic drugs is often inhibited due to limited availability, side effects or parasite resistance. Medicinal plants can be used as alternatives or adjuncts to current antiparasitic therapies. This systematic review and meta-analysis aimed to critically synthesise the literature on the efficacy of different plants and plant compounds against common human GI parasites and their toxicity profiles. Searches were conducted from inception to September 2021. Of 5393 screened articles, 162 were included in the qualitative synthesis (159 experimental studies and three randomised control trials [RCTs]), and three articles were included in meta-analyses. A total of 507 plant species belonging to 126 families were tested against different parasites, and most of these (78.4%) evaluated antiparasitic efficacy in vitro. A total of 91 plant species and 34 compounds were reported as having significant in vitro efficacy against parasites. Only a few plants (n = 57) were evaluated for their toxicity before testing their antiparasitic effects. The meta-analyses revealed strong evidence of the effectiveness of Lepidium virginicum L. against Entamoeba histolytica with a pooled mean IC50 of 198.63 μg/mL (95% CI 155.54-241.72). We present summary tables and various recommendations to direct future research.
Collapse
Affiliation(s)
- Sandamalie Ranasinghe
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| | - Sasha Aspinall
- School of Allied Health, College of Health and Education, Murdoch University, Perth, Western Australia, Australia
| | - Amber Beynon
- Department of Chiropractic, Faculty of Medicine, Health and Human Sciences, Macquarie University, Macquarie Park, New South Wales, Australia
| | - Amanda Ash
- Centre for Biosecurity and One Health, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| | - Alan Lymbery
- Centre for Sustainable Aquatic Ecosystems, Harry Butler Institute, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
43
|
Gaona-López C, Martínez-Vázquez AV, Villalobos-Rocha JC, Juárez-Rendón KJ, Rivera G. Analysis of Giardia lamblia Nucleolus as Drug Target: A Review. Pharmaceuticals (Basel) 2023; 16:1168. [PMID: 37631082 PMCID: PMC10457859 DOI: 10.3390/ph16081168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Giardia lamblia (G. lamblia) is the main causative agent of diarrhea worldwide, affecting children and adults alike; in the former, it can be lethal, and in the latter a strong cause of morbidity. Despite being considered a predominant disease in low-income and developing countries, current migratory flows have caused an increase in giardiasis cases in high-income countries. Currently, there is a wide variety of chemotherapeutic treatments to combat this parasitosis, most of which have potentially serious side effects, such as genotoxic, carcinogenic, and teratogenic. The necessity to create novel treatments and discover new therapeutic targets to fight against this illness is evident. The current review centers around the controversial nucleolus of G. lamblia, providing a historical perspective that traces its apparent absence to the present evidence supporting its existence as a subnuclear compartment in this organism. Additionally, possible examples of ncRNAs and proteins ubiquitous to the nucleolus that can be used as targets of different therapeutic strategies are discussed. Finally, some examples of drugs under research that could be effective against G. lamblia are described.
Collapse
Affiliation(s)
- Carlos Gaona-López
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| | | | - Juan Carlos Villalobos-Rocha
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico;
| | - Karina Janett Juárez-Rendón
- Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (A.V.M.-V.); (K.J.J.-R.)
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico
| |
Collapse
|
44
|
Pham NTK, Khamrin P, Shimizu-Onda Y, Hoque SA, Trinh QD, Komine-Aizawa S, Okitsu S, Maneekarn N, Hayakawa S, Yoshimune K, Ushijima H. Genetic diversity and declining norovirus prevalence in infants and children during Japan's COVID-19 pandemic: a three-year molecular surveillance. Arch Virol 2023; 168:231. [PMID: 37584776 DOI: 10.1007/s00705-023-05856-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/18/2023] [Indexed: 08/17/2023]
Abstract
Noroviruses (NoVs) are a global concern, causing widespread outbreaks and sporadic acute gastroenteritis (AGE) cases across all age groups. Recent research has shed light on the emergence of novel recombinant strains of NoV in various countries. To delve deeper into this phenomenon, we extensively analyzed 1,175 stool samples collected from Japanese infants and children with AGE from six different prefectures in Japan over three years, from July 2018 to June 2021. Our investigation aimed to determine the prevalence and genetic characteristics of NoV associated with sporadic AGE while exploring the possibility of detecting NoV recombination events. Among the analyzed samples, we identified 355 cases positive for NoV, 11 cases attributed to GI genotypes, and 344 associated with GII genotypes. Notably, we discovered four distinct GI genotypes (GI.2, GI.3, GI.4, and GI.6) and seven diverse GII genotypes (GII.2, GII.3, GII.4, GII.6, GII.7, GII.14, and GII.17). The predominant genotypes were GII.4 (56.4%; 194 out of 344), followed by GII.2 and GII.3. Through dual genotyping based on sequencing of the ORF1/ORF2 junction region, we identified a total of 14 different RdRp/capsid genotypes. Of particular interest were the prevalent recombinant genotypes GII.4[P31] and GII.2[P16]. Notably, our study revealed a decrease in the number of children infected with NoV during and after the COVID-19 pandemic. These findings underscore the importance of continuous NoV surveillance efforts.
Collapse
Affiliation(s)
- Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
- Department of Applied Molecular Chemistry, College of Industrial Technology, Nihon University, Chiba, Japan
| | - Pattara Khamrin
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Yuko Shimizu-Onda
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Sheikh Ariful Hoque
- Cell and Tissue Culture Laboratory, Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Shoko Okitsu
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Niwat Maneekarn
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Kazuaki Yoshimune
- Department of Applied Molecular Chemistry, College of Industrial Technology, Nihon University, Chiba, Japan
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Ohyaguchi Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan.
| |
Collapse
|
45
|
Al Wazni AB, Chapman MV, Ansong D, Tawfik L. Climate Change, Fragility, and Child Mortality; Understanding the Role of Water Access and Diarrheal Disease Amongst Children Under Five During the MDG Era. JOURNAL OF PREVENTION (2022) 2023; 44:409-419. [PMID: 37036450 DOI: 10.1007/s10935-023-00732-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/11/2023]
Abstract
The present study examined the influence of improvements to Water, Sanitation, and Hygiene (WASH) infrastructure on rates of under-five mortality specifically from diarrheal disease amongst children in fragile states. The World Bank's Millennium Development Goals and Sustainable Development Goals both include a specific target of reduction in preventable disease amongst children, as well as goal to improve WASH. Although gains have been made, children under the age of five remain particularly vulnerable to diarrheal mortality in states identified as fragile. Increasingly, climate change is placing undue pressure on states labeled fragile due to their inability to properly prepare for, or respond to, natural disasters that further compromise WASH development and water safety. The impact of climate change upon child health outcomes is neither direct nor linear and necessitates a linkage framework that can account for complex pathways between environmental pressures and public health outcomes. The World Health Organization's Drive Force-Pressure-State-Exposure-Effect-Action conceptual framework was used to draw the connections between seemingly disparate, and highly nuanced, environmental, and social measures. Using a multilevel hierarchical model, this analysis used a publicly available UNICEF data set that reported rates of mortality specifically from diarrheal disease amongst children age five and younger. All 171 formally recognized countries were included, which showed a decline in diarrheal disease over time when investments in WASH infrastructure are compared. As states experience increased pressure because of climate change, this area of intervention is key for immediate health and safety of children under-five, as well as assisting fragile states long-term as the move toward stability.
Collapse
Affiliation(s)
- Anderson B Al Wazni
- School of Social Work, University of North Carolina at Chapel Hill, Tate-Turner-Kuralt Building, 325 Pittsboro St. CB#3550, Chapel Hill, NC, 27599-3550, USA.
| | - Mimi V Chapman
- School of Social Work, University of North Carolina at Chapel Hill, Tate-Turner-Kuralt Building, 325 Pittsboro St. CB#3550, Chapel Hill, NC, 27599-3550, USA
| | - David Ansong
- School of Social Work, University of North Carolina at Chapel Hill, Tate-Turner-Kuralt Building, 325 Pittsboro St. CB#3550, Chapel Hill, NC, 27599-3550, USA
| | - Linda Tawfik
- UNC Gillings School of Global Public Health, WHO Collaborating Center for Research Evidence for Sexual and Reproductive Health, 407 Rosenau Hall, Chapel Hill, NC, 27599, USA
| |
Collapse
|
46
|
Chauwa A, Bosomprah S, Laban NM, Phiri B, Chibuye M, Chilyabanyama ON, Munsaka S, Simuyandi M, Mwape I, Mubanga C, Chobe MC, Chisenga C, Chilengi R. Maternal and Infant Histo-Blood Group Antigen (HBGA) Profiles and Their Influence on Oral Rotavirus Vaccine (Rotarix TM) Immunogenicity among Infants in Zambia. Vaccines (Basel) 2023; 11:1303. [PMID: 37631871 PMCID: PMC10458424 DOI: 10.3390/vaccines11081303] [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: 05/24/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Live-attenuated, oral rotavirus vaccines have significantly reduced rotavirus-associated diarrhoea morbidity and infant mortality. However, vaccine immunogenicity is diminished in low-income countries. We investigated whether maternal and infant intrinsic susceptibility to rotavirus infection via histo-blood group antigen (HBGA) profiles influenced rotavirus (ROTARIX®) vaccine-induced responses in Zambia. We studied 135 mother-infant pairs under a rotavirus vaccine clinical trial, with infants aged 6 to 12 weeks at pre-vaccination up to 12 months old. We determined maternal and infant ABO/H, Lewis, and secretor HBGA phenotypes, and infant FUT2 HBGA genotypes. Vaccine immunogenicity was measured as anti-rotavirus IgA antibody titres. Overall, 34 (31.3%) children were seroconverted at 14 weeks, and no statistically significant difference in seroconversion was observed across the various HBGA profiles in early infant life. We also observed a statistically significant difference in rotavirus-IgA titres across infant HBGA profiles at 12 months, though no statistically significant difference was observed between the study arms. There was no association between maternal HBGA profiles and infant vaccine immunogenicity. Overall, infant HBGAs were associated with RV vaccine immunogenicity at 12 months as opposed to in early infant life. Further investigation into the low efficacy of ROTARIX® and appropriate intervention is key to unlocking the full vaccine benefits for U5 children.
Collapse
Affiliation(s)
- Adriace Chauwa
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka P.O. Box 50110, Zambia;
| | - Samuel Bosomprah
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
- Department of Biostatistics, School of Public Health, University of Ghana, Accra P.O. Box LG13, Ghana
| | - Natasha Makabilo Laban
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Bernard Phiri
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
| | - Mwelwa Chibuye
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
- Department of Global Health, Amsterdam Institute for Global Health and Development (AIGHD), Amsterdam University Medical Centers, University of Amsterdam, 1012 WP Amsterdam, The Netherlands
| | - Obvious Nchimunya Chilyabanyama
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
| | - Sody Munsaka
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka P.O. Box 50110, Zambia;
| | - Michelo Simuyandi
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
| | - Innocent Mwape
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
| | - Cynthia Mubanga
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
| | - Masuzyo Chirwa Chobe
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
| | - Caroline Chisenga
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
| | - Roma Chilengi
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia; (S.B.); (N.M.L.); (B.P.); (M.C.); (O.N.C.); (M.S.); (I.M.); (C.M.); (M.C.C.); (C.C.); (R.C.)
| |
Collapse
|
47
|
Nandi I, Aroeti B. Mitogen-Activated Protein Kinases (MAPKs) and Enteric Bacterial Pathogens: A Complex Interplay. Int J Mol Sci 2023; 24:11905. [PMID: 37569283 PMCID: PMC10419152 DOI: 10.3390/ijms241511905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Diverse extracellular and intracellular cues activate mammalian mitogen-activated protein kinases (MAPKs). Canonically, the activation starts at cell surface receptors and continues via intracellular MAPK components, acting in the host cell nucleus as activators of transcriptional programs to regulate various cellular activities, including proinflammatory responses against bacterial pathogens. For instance, binding host pattern recognition receptors (PRRs) on the surface of intestinal epithelial cells to bacterial pathogen external components trigger the MAPK/NF-κB signaling cascade, eliciting cytokine production. This results in an innate immune response that can eliminate the bacterial pathogen. However, enteric bacterial pathogens evolved sophisticated mechanisms that interfere with such a response by delivering virulent proteins, termed effectors, and toxins into the host cells. These proteins act in numerous ways to inactivate or activate critical components of the MAPK signaling cascades and innate immunity. The consequence of such activities could lead to successful bacterial colonization, dissemination, and pathogenicity. This article will review enteric bacterial pathogens' strategies to modulate MAPKs and host responses. It will also discuss findings attempting to develop anti-microbial treatments by targeting MAPKs.
Collapse
Affiliation(s)
| | - Benjamin Aroeti
- Department of Biological Chemistry, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 9190410, Israel;
| |
Collapse
|
48
|
Xu J, Xue Y, Bolinger AA, Li J, Zhou M, Chen H, Li H, Zhou J. Therapeutic potential of salicylamide derivatives for combating viral infections. Med Res Rev 2023; 43:897-931. [PMID: 36905090 PMCID: PMC10247541 DOI: 10.1002/med.21940] [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: 11/23/2021] [Revised: 11/09/2022] [Accepted: 02/26/2023] [Indexed: 03/12/2023]
Abstract
Since time immemorial human beings have constantly been fighting against viral infections. The ongoing and devastating coronavirus disease 2019 pandemic represents one of the most severe and most significant public health emergencies in human history, highlighting an urgent need to develop broad-spectrum antiviral agents. Salicylamide (2-hydroxybenzamide) derivatives, represented by niclosamide and nitazoxanide, inhibit the replication of a broad range of RNA and DNA viruses such as flavivirus, influenza A virus, and coronavirus. Moreover, nitazoxanide was effective in clinical trials against different viral infections including diarrhea caused by rotavirus and norovirus, uncomplicated influenza A and B, hepatitis B, and hepatitis C. In this review, we summarize the broad antiviral activities of salicylamide derivatives, the clinical progress, and the potential targets or mechanisms against different viral infections and highlight their therapeutic potential in combating the circulating and emerging viral infections in the future.
Collapse
Affiliation(s)
- Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Yu Xue
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Andrew A. Bolinger
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jun Li
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Mingxiang Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Haiying Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
49
|
Köffer J, Frontzek A, Eigner U. Development and validation of a bacterial gastrointestinal multiplex RT-PCR assay for use on a fully automated molecular system. J Microbiol Methods 2023; 210:106754. [PMID: 37263528 DOI: 10.1016/j.mimet.2023.106754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/03/2023]
Abstract
PCR-based enteric multiplex panels represent a rapid and reliable alternative to conventional "classical" phenotypic stool diagnostics. The aim of this study was to establish a laboratory-developed non-commercial multiplex Real-Time-PCR panel for the detection of the most important bacterial stool pathogens, Salmonella spp., Shigella spp., Yersinia enterocolitica/ pseudotuberculosis and Campylobacter jejuni/coli. on the "open" cobas omni Utility Channel (UC) of the cobas 6800 system (Roche). The aim was to replace the laborious phenotypical stool diagnostics with a high throughput Real-Time PCR method. The respective primers and probes were designed to cover conserved genomic regions of the pathogens and validated using Ultramer oligonucleotides, positive stool material and reference strains. To further validate the multiplex PCR-assay, the following parameters were evaluated: analytical-sensitivity and -specificity, cross-reactivity, linearity and inter- and intra-assay variance as well as limit of detection (LOD). In addition a retrospective analysis of culture positive and negative samples from daily routine was performed using 745 native stool samples. The Gastro assay was linear over a 5-log-unit and within the expected dynamic range with amplification efficiencies ranging from 94.6% to 120%. In addition, all targets showed excellent coefficients of repeatability (≤ 1.11%), intermediate precision (≤ 1.02%) and total variance (≤ 1.39%). In terms of analytical sensitivity the assay demonstrated detection limits ranging from 7.83 copies per reaction to 14.4 copies per reaction. The assay showed excellent agreement with culture methods (>95%) and a 100% sensitivity and specificity after resolution of discrepant results. The multiplex-PCR assay provides a comprehensive, rapid and sensitive alternative to conventional methods for the detection of the major bacterial stool pathogens in diagnostic laboratories.
Collapse
Affiliation(s)
| | - André Frontzek
- Roche Diagnostics (formerly, during study: Labor Stein, Mönchengladbach, Germany), Mannheim, Germany
| | | |
Collapse
|
50
|
Li TT, Xu Q, Liu MC, Wang T, Che TL, Teng AY, Lv CL, Wang GL, Hong F, Liu W, Fang LQ. Prevalence and Etiological Characteristics of Norovirus Infection in China: A Systematic Review and Meta-Analysis. Viruses 2023; 15:1336. [PMID: 37376635 DOI: 10.3390/v15061336] [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: 05/16/2023] [Revised: 06/04/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Norovirus is a common cause of sporadic cases and outbreaks of gastroenteritis worldwide, although its prevalence and the dominant genotypes responsible for gastroenteritis outbreaks remain obscure. A systematic review was conducted on norovirus infection in China between January 2009 and March 2021. A meta-analysis and beta-binomial regression model were used to explore the epidemiological and clinical characteristics of norovirus infection and the potential factors contributing to the attack rate of the norovirus outbreaks, respectively. A total of 1132 articles with 155,865 confirmed cases were included, with a pooled positive test rate of 11.54% among 991,786 patients with acute diarrhea and a pooled attack rate of 6.73% in 500 norovirus outbreaks. GII.4 was the predominant genotype in both the etiological surveillance and outbreaks, followed by GII.3 in the etiological surveillance, and GII.17 in the outbreaks, with the proportion of recombinant genotypes increasing in recent years. A higher attack rate in the norovirus outbreaks was associated with age group (older adults), settings (nurseries, primary schools, etc.) and region (North China). The nation-wide pooled positive rate in the etiological surveillance of norovirus is lower than elsewhere in the global population, while the dominant genotypes are similar in both the etiological surveillance and the outbreak investigations. This study contributes to the understanding of norovirus infection with different genotypes in China. The prevention and control of norovirus outbreaks during the cold season should be intensified, with special attention paid to and enhanced surveillance performed in nurseries, schools and nursing homes from November to March.
Collapse
Affiliation(s)
- Ting-Ting Li
- School of Public Health, Guizhou Medical University, Guiyang 550025, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qiang Xu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Mei-Chen Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Tao Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Tian-Le Che
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Ai-Ying Teng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Chen-Long Lv
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Guo-Lin Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Feng Hong
- School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- School of Public Health, Anhui Medical University, Hefei 230032, China
| | - Li-Qun Fang
- School of Public Health, Guizhou Medical University, Guiyang 550025, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| |
Collapse
|