1
|
Razzak A, Saha O, Sultana KF, Amin MR, Zahid AB, Sultana A, Bristi UP, Rajia S, Sarker N, Rahaman MM, Bahadur NM, Hossen F. Development of a Novel mRNA Vaccine Against Shigella Pathotypes Causing Widespread Shigellosis Endemic: An In-Silico Immunoinformatic Approach. Bioinform Biol Insights 2025; 19:11779322251328302. [PMID: 40160890 PMCID: PMC11951904 DOI: 10.1177/11779322251328302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Shigellosis remains a major global health concern, particularly in regions with poor sanitation and limited access to clean water. This study used immunoinformatics and reverse vaccinology to design a potential mRNA vaccine targeting Shigella pathotypes out of 4071 proteins from Shigella sonnei str. Ss046, 4 key antigenic candidates were identified: putative outer membrane protein (Q3YZL0), PapC-like porin protein (Q3YZM5), putative fimbrial-like protein (Q3Z3I2), and lipopolysaccharide (LPS)-assembly protein LptD (Q3Z5V5), ensuring broad pathotype coverage. A multitope vaccine was designed incorporating cytotoxic T lymphocyte, helper T lymphocyte, and B-cell epitopes, linked with suitable linkers and adjuvants to enhance immunogenicity. Computational analyses predicted vaccine's favorable antigenicity, solubility, and stability, while molecular docking and dynamic simulations demonstrated strong binding affinity and stability with Toll-like receptor 4 (TLR-4), indicating potential for robust immune activation. Immune simulations predicted strong humoral and cellular immune responses, characterized by significant cytokine production and long-term immune memory. Structural evaluations of the complex, including radius of gyration, root mean square deviation, root mean square fluctuation, and solvent accessibility, confirmed the vaccine's structural integrity, and stability under physiological conditions. This research contributes to the ongoing effort to alleviate the global burden of Shigella infections, providing a foundation for future wet laboratory investigations aimed at vaccine development.
Collapse
Affiliation(s)
- Abdur Razzak
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Otun Saha
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - Mohammad Ruhul Amin
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Abdullah bin Zahid
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Afroza Sultana
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Uditi Paul Bristi
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Sultana Rajia
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Nikkon Sarker
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - Newaz Mohammed Bahadur
- Department of Chemistry, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Foysal Hossen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
2
|
Haldar R, Halder P, Koley H, Miyoshi SI, Das S. A newly developed oral infection mouse model of shigellosis for immunogenicity and protective efficacy studies of a candidate vaccine. Infect Immun 2025; 93:e0034624. [PMID: 39692481 PMCID: PMC11784180 DOI: 10.1128/iai.00346-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024] Open
Abstract
Shigella infection poses a significant public health challenge in the developing world. However, lack of a widely available mouse model that replicates human shigellosis creates a major bottleneck to better understanding of disease pathogenesis and development of newer drugs and vaccines. BALB/c mice pre-treated with streptomycin and iron (FeCl3) plus desferrioxamine intraperitoneally followed by oral infection with virulent Shigella flexneri 2a resulted in diarrhea, loss of body weight, bacterial colonization and progressive colitis characterized by disruption of epithelial lining, loss of crypt architecture with goblet cell depletion, increased polymorphonuclear infiltration into the mucosa, submucosal swelling (edema), and raised proinflammatory cytokines and chemokines in the large intestine. To evaluate the usefulness of the model for vaccine efficacy studies, mice were immunized intranasally with a recombinant protein vaccine containing Shigella invasion protein invasion plasmid antigen B (IpaB). Vaccinated mice conferred protection against Shigella, indicating that the model is suitable for testing of vaccine candidates. To protect both Shigella and Salmonella, a chimeric recombinant vaccine (rIpaB-T2544) was developed by fusing IpaB with Salmonella outer membrane protein T2544. Vaccinated mice developed antigen-specific serum IgG and IgA antibodies and a balanced Th1/Th2 response and were protected against oral challenge with Shigella (S. flexneri 2a, Shigella dysenteriae, and Shigella sonnei) using our present mouse model and Salmonella (Salmonella Typhi and Paratyphi) using an iron overload mouse model. We describe here the development of an oral Shigella infection model in wild-type mouse. This model was successfully used to demonstrate the immunogenicity and protective efficacy of a candidate protein subunit vaccine against Shigella.
Collapse
MESH Headings
- Animals
- Dysentery, Bacillary/prevention & control
- Dysentery, Bacillary/immunology
- Dysentery, Bacillary/microbiology
- Disease Models, Animal
- Shigella Vaccines/immunology
- Shigella Vaccines/administration & dosage
- Mice
- Mice, Inbred BALB C
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- Shigella flexneri/immunology
- Female
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Adhesins, Bacterial/immunology
- Adhesins, Bacterial/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Immunoglobulin G/blood
- Immunoglobulin A/immunology
- Cytokines/metabolism
- Vaccine Efficacy
- Bacterial Proteins
Collapse
Affiliation(s)
- Risha Haldar
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Shin-ichi Miyoshi
- Division of Medicine, Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Okayama, Japan
| | - Santasabuj Das
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
- Division of Biological Science, ICMR-National Institute of Occupational Health, Ahmedabad, Gujarat, India
| |
Collapse
|
3
|
Yi D, Liu X, Wang M, Zhao L, Liu Y, Xu Z, Peng Y, Zhang R, Wei Q, Liang Z, He J. Rosmarinic acid alleviated intestinal barrier damage caused by Escherichia coli by regulating the gut microbiota and inhibiting the NF-κB signalling pathway in mice. Food Funct 2024; 15:11740-11756. [PMID: 39540591 DOI: 10.1039/d4fo02654c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Escherichia coli (E. coli) is a common zoonotic foodborne pathogen that poses a major threat to public health and economic development. Rosmarinic acid (RA) can inhibit intestinal inflammation; however, the protective effect of RA against the intestinal barrier damage induced by E. coli in mice and the underlying mechanism have not been elucidated. In this study, mice were orally administered with RA (20 mg kg-1) by gavage for one week and then were intraperitoneally challenged with E. coli. Mouse colonic epithelial cells (MCECs) were pretreated with RA for 6 h and challenged with E. coli (MOI = 1000) for 3 h. The results revealed that RA alleviated E. coli-induced weight loss in mice; reduced the increase in the levels of TNF-α, IL-6 and IL-1β in the serum; alleviated the decrease in ZO-1 protein expression; and increased intestinal permeability by inhibiting the NF-κB signalling pathway both in vivo and in vitro. Moreover, RA relieved the increase in intestinal permeability, reversed the structural damage to the mouse gut microbiota caused by E. coli, and increased the abundance of beneficial bacteria, including Lachnospiraceae_NK4136_group. Additionally, RA lost its protective function against E. coli infection in a pseudosterile mouse model, suggesting that the protection induced by RA was dependent on the gut microbiota. In conclusion, these results indicate that RA alleviates E. coli-induced inflammatory damage to the intestinal barrier by inhibiting the NF-κB signalling pathway and maintaining gut microbiota homeostasis. These findings provide new ideas and foundations for the application of RA as protection against E. coli.
Collapse
Affiliation(s)
- Dandan Yi
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Xia Liu
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Menghui Wang
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Linyi Zhao
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Yu Liu
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Zhiran Xu
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Ying Peng
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Rui Zhang
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Qianyin Wei
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
| | - Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, 530004, PR China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530004, PR China
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, Room 307, 100 Daxue Road, Xixiangtang District, Nanning, Guangxi 530004, P. R. China.
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, 530004, PR China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530004, PR China
| |
Collapse
|
4
|
Huang F, Wang Q, Wang Z, Lv L, Feng J. Effects of Organic Zinc on the Growth Performance of Weanling Pigs: A Meta-analysis. Biol Trace Elem Res 2024; 202:5051-5060. [PMID: 38253801 DOI: 10.1007/s12011-024-04070-2] [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: 11/09/2023] [Accepted: 01/15/2024] [Indexed: 01/24/2024]
Abstract
Supplementation of feed with organic zinc (Zn) has long been discussed as an alternative to inorganic Zn in pigs, but its effects on growth performance are mixed. This meta-analysis was conducted to provide a comprehensive evaluation of the influence of organic Zn on the growth performance of weanling pigs, on the basis of average daily gain (ADG), average daily feed intake (ADFI), and feed to gain ratio (F/G). We screened the PubMed and Web of Science databases (published before December 31, 2022; limited to English) systematically and contrasted organic Zn supplementation with inorganic Zn supplementation. There were 680 retrievals of studies, of which 16 (1389 pigs, 37 records) were eligible to analyze. Weighted mean differences (WMDs) and 95% confidence intervals (CIs) were calculated using a random-effects model. The subgroup analysis was classified as organic Zn source (Zn-amino acid (Zn-AA), Zn-glycine (Zn-Gly), Zn-methionine (Zn-Met), Zn-Lysine (Zn-Lys), proteinate complex Zn (Zn-Pro), chitosan-Zn (Zn-CS) or Zn-lactate (Zn-Lac)) and Zn additive dose (low, medium, or high, i.e., lower than, equal to or higher than the requirement of NRC). Organic Zn addition in the weaning phase increased the ADG (P < 0.001) and the ADFI (P = 0.023) and decreased the F/G (P < 0.001). Specifically, for the organic sources, only Zn-CS supplementation presented significant effects on the ADG (P < 0.001), ADFI (P = 0.011), and F/G (P < 0.001). Moreover, medium-dose organic Zn supplementation had positive effects on ADG (P = 0.012), ADFI (P = 0.018), and F/G (P < 0.001). Our results indicate that organic Zn added to diets greatly improves the growth performance of weanling pigs.
Collapse
Affiliation(s)
- Feifei Huang
- Key Laboratory of Animal Nutrition and Feed Sciences of Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Qiwen Wang
- Key Laboratory of Animal Nutrition and Feed Sciences of Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Zhonghang Wang
- Key Laboratory of Animal Nutrition and Feed Sciences of Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Liangkang Lv
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jie Feng
- Key Laboratory of Animal Nutrition and Feed Sciences of Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
5
|
Yang C, Xiao J, Xu Z, Wang Z. Gut Microbiota Changes and Its Potential Relations with Thyroid Disorders: From Composition to Therapeutic Targets. Int J Gen Med 2024; 17:3719-3731. [PMID: 39219667 PMCID: PMC11363920 DOI: 10.2147/ijgm.s481183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Composed of over 1200 species of anaerobes and aerobes bacteria along with bacteriophages, viruses, and fungal species, the human gut microbiota (GM) is vital to health, including digestive equilibrium, immunologic, hormonal, and metabolic homeostasis. Micronutrients, usually refer to trace elements (copper, iodine, iron, selenium, zinc) and vitamins (A, C, D, E), interact with the GM to influence host immune metabolism. So far, microbiome studies have revealed an association between disturbances in the microbiota and various pathological disorders, such as anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis, anxiety, depression, early-onset cancers, type 1 diabetes (T1D) and type 2 diabetes (T2D). As common conditions, thyroid diseases, encompassing Graves' disease (GD), Graves' orbitopathy (GO), Hashimoto's thyroiditis (HT), benign nodules, and papillary thyroid cancer (TC), have negative impacts on the health of all populations. Following recent studies, GM might play an integral role in triggering diseases of the thyroid gland. Not only do environmental triggers and genetic predisposing background lead to auto-aggressive damage, involving cellular and humoral networks of the immune system, but the intestinal microbiota interacts with distant organs by signals that may be part of the bacteria themselves or their metabolites. The review aims to describe the current knowledge about the GM in the metabolism of thyroid hormones and the pathogenesis of thyroid diseases and its involvement in the appearance of benign nodules and papillary TC. We further focused on the reciprocal interaction between GM composition and the most used treatment drugs for thyroid disorders. However, the exact etiology has not yet been known. To elucidate more precisely the mechanism for GM involvement in the development of thyroid diseases, future work is needed.
Collapse
Affiliation(s)
- Cai Yang
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Jiafeng Xiao
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Zibei Xu
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Zehong Wang
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| |
Collapse
|
6
|
Wang Y, Xiao J, Wei S, Su Y, Yang X, Su S, Lan L, Chen X, Huang T, Shan Q. Protective effect of zinc gluconate on intestinal mucosal barrier injury in antibiotics and LPS-induced mice. Front Microbiol 2024; 15:1407091. [PMID: 38855764 PMCID: PMC11157515 DOI: 10.3389/fmicb.2024.1407091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
Objective The aim of the study is to investigate the function and mechanism of Zinc Gluconate (ZG) on intestinal mucosal barrier damage in antibiotics and Lipopolysaccharide (LPS)-induced mice. Methods We established a composite mouse model by inducing intestinal mucosal barrier damage using antibiotics and LPS. The animals were divided into five groups: Control (normal and model) and experimental (low, medium, and high-dose ZG treatments). We evaluated the intestinal mucosal barrier using various methods, including monitoring body weight and fecal changes, assessing pathological damage and ultrastructure of the mouse ileum, analyzing expression levels of tight junction (TJ)-related proteins and genes, confirming the TLR4/NF-κB signaling pathway, and examining the structure of the intestinal flora. Results In mice, the dual induction of antibiotics and LPS led to weight loss, fecal abnormalities, disruption of ileocecal mucosal structure, increased intestinal barrier permeability, and disorganization of the microbiota structure. ZG restored body weight, alleviated diarrheal symptoms and pathological damage, and maintained the structural integrity of intestinal epithelial cells (IECs). Additionally, ZG reduced intestinal mucosal permeability by upregulating TJ-associated proteins (ZO-1, Occludin, Claudin-1, and JAM-A) and downregulating MLCK, thereby repairing intestinal mucosal barrier damage induced by dual induction of antibiotics and LPS. Moreover, ZG suppressed the TLR4/NF-κB signaling pathway, demonstrating anti-inflammatory properties and preserving barrier integrity. Furthermore, ZG restored gut microbiota diversity and richness, evidenced by increased Shannon and Observed features indices, and decreased Simpson's index. ZG also modulated the relative abundance of beneficial human gut bacteria (Bacteroidetes, Firmicutes, Verrucomicrobia, Parabacteroides, Lactobacillus, and Akkermansia) and harmful bacteria (Proteobacteria and Enterobacter), repairing the damage induced by dual administration of antibiotics and LPS. Conclusion ZG attenuates the dual induction of antibiotics and LPS-induced intestinal barrier damage and also protects the intestinal barrier function in mice.
Collapse
Affiliation(s)
- Yongcai Wang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Dazhou Central Hospital, Dazhou, China
| | - Juan Xiao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Sumei Wei
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ying Su
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xia Yang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shiqi Su
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liancheng Lan
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiuqi Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ting Huang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Qingwen Shan
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
7
|
Babb C, Badji H, Bhuiyan MTR, Cornick J, Qureshi S, Sonye C, Shapiama Lopez WV, Adnan M, Atlas HE, Begum K, Brennhofer SA, Ceesay BE, Ceesay AK, Cunliffe NA, Garcia Bardales PF, Haque S, Horne B, Hossain MJ, Iqbal J, Islam MT, Islam S, Khanam F, Kotloff KL, Malemia T, Manzanares Villanueva K, Million GM, Munthali V, Ochieng JB, Ogwel B, Paredes Olortegui M, Omore R, Pavlinac PB, Platts-Mills JA, Sears KT, Secka O, Tennant SM, Peñataro Yori P, Yousafzai MT, Jere KC, Kosek MN, Munga S, Ikumapayi UN, Qadri F, Qamar FN, Rogawski McQuade ET. Evaluation of Fecal Inflammatory Biomarkers to Identify Bacterial Diarrhea Episodes: Systematic Review and Protocol for the Enterics for Global Health Shigella Surveillance Study. Open Forum Infect Dis 2024; 11:S65-S75. [PMID: 38532957 PMCID: PMC10962755 DOI: 10.1093/ofid/ofad652] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024] Open
Abstract
Background The measurement of fecal inflammatory biomarkers among individuals presenting to care with diarrhea could improve the identification of bacterial diarrheal episodes that would benefit from antibiotic therapy. We reviewed prior literature in this area and describe our proposed methods to evaluate 4 biomarkers in the Enterics for Global Health (EFGH) Shigella surveillance study. Methods We systematically reviewed studies since 1970 from PubMed and Embase that assessed the diagnostic characteristics of inflammatory biomarkers to identify bacterial diarrhea episodes. We extracted sensitivity and specificity and summarized the evidence by biomarker and diarrhea etiology. In EFGH, we propose using commercial enzyme-linked immunosorbent assays to test for myeloperoxidase, calprotectin, lipocalin-2, and hemoglobin in stored whole stool samples collected within 24 hours of enrollment from participants in the Bangladesh, Kenya, Malawi, Pakistan, Peru, and The Gambia sites. We will develop clinical prediction scores that incorporate the inflammatory biomarkers and evaluate their ability to identify Shigella and other bacterial etiologies of diarrhea as determined by quantitative polymerase chain reaction (qPCR). Results Forty-nine studies that assessed fecal leukocytes (n = 39), red blood cells (n = 26), lactoferrin (n = 13), calprotectin (n = 8), and myeloperoxidase (n = 1) were included in the systematic review. Sensitivities were high for identifying Shigella, moderate for identifying any bacteria, and comparable across biomarkers. Specificities varied depending on the outcomes assessed. Prior studies were generally small, identified red and white blood cells by microscopy, and used insensitive gold standard diagnostics, such as conventional bacteriological culture for pathogen detection. Conclusions Our evaluation of inflammatory biomarkers to distinguish diarrhea etiologies as determined by qPCR will provide an important addition to the prior literature, which was likely biased by the limited sensitivity of the gold standard diagnostics used. We will determine whether point-of-care biomarker tests could be a viable strategy to inform treatment decision making and increase appropriate targeting of antibiotic treatment to bacterial diarrhea episodes.
Collapse
Affiliation(s)
- Courtney Babb
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Henry Badji
- Medical Research Council Unit The Gambia, London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Md Taufiqur Rahman Bhuiyan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Jennifer Cornick
- Institute of Infection, Veterinary and Ecological Sciences, Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
| | - Sonia Qureshi
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Catherine Sonye
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | | | - Mehreen Adnan
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Hannah E Atlas
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Kehkashan Begum
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Stephanie A Brennhofer
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Bubacarr E Ceesay
- Medical Research Council Unit The Gambia, London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Abdoulie K Ceesay
- Medical Research Council Unit The Gambia, London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Nigel A Cunliffe
- Institute of Infection, Veterinary and Ecological Sciences, Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | | | - Shahinur Haque
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Bri’Anna Horne
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - M Jahangir Hossain
- Medical Research Council Unit The Gambia, London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Junaid Iqbal
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Md Taufiqul Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Sadia Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Farhana Khanam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | - Billy Ogwel
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | | | - Richard Omore
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Patricia B Pavlinac
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Khandra T Sears
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ousman Secka
- Medical Research Council Unit The Gambia, London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Sharon M Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Pablo Peñataro Yori
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | | | - Khuzwayo C Jere
- Institute of Infection, Veterinary and Ecological Sciences, Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Department of Medical Laboratory Sciences, Kamuzu University of Health Sciences, School of Life Sciences and Health Professions, Blantyre, Malawi
| | - Margaret N Kosek
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Stephen Munga
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Usman N Ikumapayi
- Medical Research Council Unit The Gambia, London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Firdausi Qadri
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Farah Naz Qamar
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | | |
Collapse
|
8
|
Clarkson KA, Porter CK, Talaat KR, Kapulu MC, Chen WH, Frenck RW, Bourgeois AL, Kaminski RW, Martin LB. Shigella-Controlled Human Infection Models: Current and Future Perspectives. Curr Top Microbiol Immunol 2024; 445:257-313. [PMID: 35616717 PMCID: PMC7616482 DOI: 10.1007/82_2021_248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Shigella-controlled human infection models (CHIMs) are an invaluable tool utilized by the vaccine community to combat one of the leading global causes of infectious diarrhea, which affects infants, children and adults regardless of socioeconomic status. The impact of shigellosis disproportionately affects children in low- and middle-income countries (LMICs) resulting in cognitive and physical stunting, perpetuating a cycle that must be halted. Shigella-CHIMs not only facilitate the early evaluation of enteric countermeasures and up-selection of the most promising products but also provide insight into mechanisms of infection and immunity that are not possible utilizing animal models or in vitro systems. The greater understanding of shigellosis obtained in CHIMs builds and empowers the development of new generation solutions to global health issues which are unattainable in the conventional laboratory and clinical settings. Therefore, refining, mining and expansion of safe and reproducible infection models hold the potential to create effective means to end diarrheal disease and associated co-morbidities associated with Shigella infection.
Collapse
Affiliation(s)
- Kristen A Clarkson
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Chad K Porter
- Enteric Disease Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 624 North Broadway Street Hampton House, Baltimore, MD, 21205, USA
| | - Melissa C Kapulu
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi County Hospital, Off Bofa Road, Kilifi, 80108, Kenya
| | - Wilbur H Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Robert W Frenck
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - A Louis Bourgeois
- PATH Center for Vaccine Innovation and Access, 455 Massachusetts Avenue NW, Washington, DC, 20001, USA
| | - Robert W Kaminski
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Laura B Martin
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy.
| |
Collapse
|
9
|
Matanza XM, Clements A. Pathogenicity and virulence of Shigella sonnei: A highly drug-resistant pathogen of increasing prevalence. Virulence 2023; 14:2280838. [PMID: 37994877 PMCID: PMC10732612 DOI: 10.1080/21505594.2023.2280838] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/01/2023] [Indexed: 11/24/2023] Open
Abstract
Shigella spp. are the causative agent of shigellosis (or bacillary dysentery), a diarrhoeal disease characterized for the bacterial invasion of gut epithelial cells. Among the 4 species included in the genus, Shigella flexneri is principally responsible for the disease in the developing world while Shigella sonnei is the main causative agent in high-income countries. Remarkably, as more countries improve their socioeconomic conditions, we observe an increase in the relative prevalence of S. sonnei. To date, the reasons behind this change in aetiology depending on economic growth are not understood. S. flexneri has been widely used as a model to study the pathogenesis of the genus, but as more research data are collected, important discrepancies with S. sonnei have come to light. In comparison to S. flexneri, S. sonnei can be differentiated in numerous aspects; it presents a characteristic O-antigen identical to that of one serogroup of the environmental bacterium Plesiomonas shigelloides, a group 4 capsule, antibacterial mechanisms to outcompete and displace gut commensal bacteria, and a poorer adaptation to an intracellular lifestyle. In addition, the World Health Organization (WHO) have recognized the significant threat posed by antibiotic-resistant strains of S. sonnei, demanding new approaches. This review gathers knowledge on what is known about S. sonnei within the context of other Shigella spp. and aims to open the door for future research on understanding the increasing spread of this pathogen.
Collapse
Affiliation(s)
- Xosé M. Matanza
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London, UK
| | - Abigail Clements
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London, UK
| |
Collapse
|
10
|
Alphonse N, Odendall C. Animal models of shigellosis: a historical overview. Curr Opin Immunol 2023; 85:102399. [PMID: 37952487 DOI: 10.1016/j.coi.2023.102399] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023]
Abstract
Shigella spp. are major causative agents of bacillary dysentery, a severe enteric disease characterized by destruction and inflammation of the colonic epithelium accompanied by acute diarrhea, fever, and abdominal pain. Although antibiotics have traditionally been effective, the prevalence of multidrug-resistant strains is increasing, stressing the urgent need for a vaccine. The human-specific nature of shigellosis and the absence of a dependable animal model have posed significant obstacles in understanding Shigella pathogenesis and the host immune response, both of which are crucial for the development of an effective vaccine. Efforts have been made over time to develop a physiological model that mimics the pathological features of the human disease with limited success until the recent development of genetically modified mouse models. In this review, we provide an overview of Shigella pathogenesis and chronicle the historical development of various shigellosis models, emphasizing their strengths and weaknesses.
Collapse
Affiliation(s)
- Noémie Alphonse
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK; Immunoregulation Laboratory, Francis Crick Institute, London, UK.
| | - Charlotte Odendall
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London, UK.
| |
Collapse
|
11
|
Fan L, Xia Y, Wang Y, Han D, Liu Y, Li J, Fu J, Wang L, Gan Z, Liu B, Fu J, Zhu C, Wu Z, Zhao J, Han H, Wu H, He Y, Tang Y, Zhang Q, Wang Y, Zhang F, Zong X, Yin J, Zhou X, Yang X, Wang J, Yin Y, Ren W. Gut microbiota bridges dietary nutrients and host immunity. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2466-2514. [PMID: 37286860 PMCID: PMC10247344 DOI: 10.1007/s11427-023-2346-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023]
Abstract
Dietary nutrients and the gut microbiota are increasingly recognized to cross-regulate and entrain each other, and thus affect host health and immune-mediated diseases. Here, we systematically review the current understanding linking dietary nutrients to gut microbiota-host immune interactions, emphasizing how this axis might influence host immunity in health and diseases. Of relevance, we highlight that the implications of gut microbiota-targeted dietary intervention could be harnessed in orchestrating a spectrum of immune-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yaoyao Xia
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Youxia Wang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Jiahuan Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie Fu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Leli Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Zhending Gan
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Bingnan Liu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Fu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenhua Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hui Han
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yiwen He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingzhuo Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yibin Wang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Fan Zhang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China
| | - Xin Zong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, 712100, China.
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
| | - Wenkai Ren
- Guangdong Laboratory of Lingnan Modern Agriculture, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
12
|
Abdelfattah A, Samir R, Amin HM. Production of highly immunogenic and safe Triton X-100 produced bacterial ghost vaccine against Shigella flexneri 2b serotype. Gut Pathog 2023; 15:41. [PMID: 37679798 PMCID: PMC10483756 DOI: 10.1186/s13099-023-00568-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Bacterial ghost cells (BGCs) are cells were drained of their genetic and cytoplasmic components. This work aimed to develop vaccine candidates against the Shigella flexneri (S. flexneri) 2b serotype using the BGCs approach. For the first time, (S. flexneri) 2b serotype BGCs vaccine was prepared by incubation with Triton X-100 (TX100) for only 12 h. Its safety and immunogenicity were compared to another vaccine produced using a previously used surfactant, namely Tween 80 (TW80). Scanning electron microscopy (SEM), cellular DNA, protein contents measurements, and ghost cell re-cultivation were used to confirm the successful generation of the BGCs. Immunogenicity was assessed through mice's intraperitoneal (IP) immunization followed by infection with S. flexneri ATCC 12022. Finally, histopathological examination was carried out. RESULTS Viable colony forming units (CFUs) of S. flexneri were counted from stool samples as well as homogenized colon tissues of the non-immunized challenged group. Immunized mice sera showed a significant increase in serum bactericidal activity of both preparations (TX100 = 40% and TW80 = 56%) compared to the non-immunized challenged group (positive control). The IgG levels of the bacterial ghost-vaccinated groups were four and three times greater for the TX100 and TW80 ghost vaccines, respectively, compared to that of the positive control; both bacterial ghost vaccines (BGVs) were safe and effective, according to the results of the safety check tests and histopathological analysis. CONCLUSIONS When comparing the BGVs prepared using TX100 and TW80 methods, the use of TX100 as a new chemical treating agent for BGC production attained robust results in terms of shorter incubation time with the targeted cells and a strong immune response against S. flexneri 2b serotype ATCC 12022 in the IP challenge test. However, a clinical study is needed to confirm the efficacy and total safety of this novel vaccine.
Collapse
Affiliation(s)
- Amany Abdelfattah
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 26 July Mehwar Road Intersection With Wahat Road, 6Th of October, 12451 Giza Egypt
| | - Reham Samir
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Nile Corniche, El Sayeda Zeinab, Cairo, 11562 Egypt
| | - Heba M. Amin
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 26 July Mehwar Road Intersection With Wahat Road, 6Th of October, 12451 Giza Egypt
| |
Collapse
|
13
|
Bagamian KH, Anderson IV JD, Blohm G, Scheele S. Shigella and childhood stunting: Evidence, gaps, and future research directions. PLoS Negl Trop Dis 2023; 17:e0011475. [PMID: 37699032 PMCID: PMC10497124 DOI: 10.1371/journal.pntd.0011475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023] Open
Abstract
Early childhood growth deficits have been shown to have lifelong health and economic impacts, yet their connection to one of their underlying causes, diarrheal diseases, has remained difficult to characterize. Identifying the processes and mechanisms that underlie this link has remained a challenge due to the complexity of the relationship and limitations in access to more advanced laboratory methods. In recent years, however, several large-scale, multisite studies have extensively investigated and reported the prevalence, etiology, and impacts of diarrheal diseases in children under 5 years (CU5) in low- to middle-income countries (LMICs). These studies, in combination with several single-site studies, have applied more advanced laboratory methods to uncover the etiology, true prevalence, infection mechanisms, and inflammation biomarkers of diarrheal disease. Of the multiple pathogens that have been shown to be strongly associated with diarrheal disease in CU5, Shigella is one of the more prevalent and impactful of these pathogens. In this narrative review, we highlight key insights from these studies and identify knowledge gaps and directions for future research. According to these studies, Shigella is most commonly detected in toddlers and young children; however, it can cause more severe disease and has a greater impact on linear growth for infants. Shigella often has a stronger relationship to linear growth faltering (LGF) than other enteropathogens, with higher Shigella loads resulting in greater growth deficits. Future studies should employ more Shigella-specific molecular assays and identify diarrheal etiologies using standardized diagnostics to improve child anthropometric and Shigella surveillance. Also, they should focus on uncovering the mechanisms of the relationship underlying Shigella and growth faltering to better characterize the role of asymptomatic infections and intestinal inflammation in this relationship.
Collapse
Affiliation(s)
- Karoun H. Bagamian
- Bagamian Scientific Consulting, LLC, Gainesville, Florida, United States of America
- Department of Environmental and Global Health, University of Florida, Gainesville, Florida, United States of America
| | - John D. Anderson IV
- Bagamian Scientific Consulting, LLC, Gainesville, Florida, United States of America
- Health Affairs Institute, West Virginia University, Morgantown, West Virginia, United States of America
| | - Gabriela Blohm
- Bagamian Scientific Consulting, LLC, Gainesville, Florida, United States of America
| | - Suzanne Scheele
- Center for Vaccine Innovation and Access, Washington, District of Columbia, United States of America
| |
Collapse
|
14
|
Huh E, Choi JG, Lee MY, Kim JH, Choi Y, Ju IG, Eo H, Park MG, Kim DH, Park HJ, Lee CH, Oh MS. Peripheral metabolic alterations associated with pathological manifestations of Parkinson's disease in gut-brain axis-based mouse model. Front Mol Neurosci 2023; 16:1201073. [PMID: 37635904 PMCID: PMC10447900 DOI: 10.3389/fnmol.2023.1201073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction Parkinson's disease (PD) is a representative neurodegenerative disease, and its diagnosis relies on the evaluation of clinical manifestations or brain neuroimaging in the absence of a crucial noninvasive biomarker. Here, we used non-targeted metabolomics profiling to identify metabolic alterations in the colon and plasma samples of Proteus mirabilis (P. mirabilis)-treated mice, which is a possible animal model for investigating the microbiota-gut-brain axis. Methods We performed gas chromatography-mass spectrometry to analyze the samples and detected metabolites that could reflect P. mirabilis-induced disease progression and pathology. Results and discussion Pattern, correlation and pathway enrichment analyses showed significant alterations in sugar metabolism such as galactose metabolism and fructose and mannose metabolism, which are closely associated with energy metabolism and lipid metabolism. This study indicates possible metabolic factors for P. mirabilis-induced pathological progression and provides evidence of metabolic alterations associated with P. mirabilis-mediated pathology of brain neurodegeneration.
Collapse
Affiliation(s)
- Eugene Huh
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Jin Gyu Choi
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Mee Youn Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jin Hee Kim
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Yujin Choi
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - In Gyoung Ju
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Hyeyoon Eo
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Myoung Gyu Park
- MetaCen Therapeutics Inc. R&D Center, Suwon, Republic of Korea
| | - Dong-Hyun Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Hi-Joon Park
- Acupuncture and Meridian Science Research Center (AMSRC), College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Myung Sook Oh
- Department of Oriental Pharmaceutical Science and Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Boero E, Vezzani G, Micoli F, Pizza M, Rossi O. Functional assays to evaluate antibody-mediated responses against Shigella: a review. Front Cell Infect Microbiol 2023; 13:1171213. [PMID: 37260708 PMCID: PMC10227456 DOI: 10.3389/fcimb.2023.1171213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/27/2023] [Indexed: 06/02/2023] Open
Abstract
Shigella is a major global pathogen and the etiological agent of shigellosis, a diarrheal disease that primarily affects low- and middle-income countries. Shigellosis is characterized by a complex, multistep pathogenesis during which bacteria use multiple invasion proteins to manipulate and invade the intestinal epithelium. Antibodies, especially against the O-antigen and some invasion proteins, play a protective role as titres against specific antigens inversely correlate with disease severity; however, the context of antibody action during pathogenesis remains to be elucidated, especially with Shigella being mostly an intracellular pathogen. In the absence of a correlate of protection, functional assays rebuilding salient moments of Shigella pathogenesis can improve our understanding of the role of protective antibodies in blocking infection and disease. In vitro assays are important tools to build correlates of protection. Only recently animal models to recapitulate human pathogenesis, often not in full, have been established. This review aims to discuss in vitro assays to evaluate the functionality of anti-Shigella antibodies in polyclonal sera in light of the multistep and multifaced Shigella infection process. Indeed, measurement of antibody level alone may limit the evaluation of full vaccine potential. Serum bactericidal assay (SBA), and other functional assays such as opsonophagocytic killing assays (OPKA), and adhesion/invasion inhibition assays (AIA), are instead physiologically relevant and may provide important information regarding the role played by these effector mechanisms in protective immunity. Ultimately, the review aims at providing scientists in the field with new points of view regarding the significance of functional assays of choice which may be more representative of immune-mediated protection mechanisms.
Collapse
Affiliation(s)
- Elena Boero
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Giacomo Vezzani
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
16
|
Abstract
The major function of the mammalian immune system is to prevent and control infections caused by enteropathogens that collectively have altered human destiny. In fact, as the gastrointestinal tissues are the major interface of mammals with the environment, up to 70% of the human immune system is dedicated to patrolling them The defenses are multi-tiered and include the endogenous microflora that mediate colonization resistance as well as physical barriers intended to compartmentalize infections. The gastrointestinal tract and associated lymphoid tissue are also protected by sophisticated interleaved arrays of active innate and adaptive immune defenses. Remarkably, some bacterial enteropathogens have acquired an arsenal of virulence factors with which they neutralize all these formidable barriers to infection, causing disease ranging from mild self-limiting gastroenteritis to in some cases devastating human disease.
Collapse
Affiliation(s)
- Micah J. Worley
- Department of Biology, University of Louisville, Louisville, Kentucky, USA,CONTACT Micah J. Worley Department of Biology, University of Louisville, 139 Life Sciences Bldg, Louisville, Kentucky, USA
| |
Collapse
|
17
|
Ensemble Machine Learning Model to Predict the Waterborne Syndrome. ALGORITHMS 2022. [DOI: 10.3390/a15030093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The COVID-19 epidemic has highlighted the significance of sanitization and maintaining hygienic access to clean water to reduce mortality and morbidity cases worldwide. Diarrhea is one of the prevalent waterborne diseases caused due to contaminated water in many low-income countries with similar living conditions. According to the latest statistics from the World Health Organization (WHO), diarrhea is among the top five primary causes of death worldwide in low-income nations. The condition affects people in every age group due to a lack of proper water used for daily living. In this study, a stacking ensemble machine learning model was employed against traditional models to extract clinical knowledge for better understanding patients’ characteristics; disease prevalence; hygienic conditions; quality of water used for cooking, bathing, and toiletries; chemicals used; therapist’s medications; and symptoms that are reflected in the field study data. Results revealed that the ensemble model provides higher accuracy with 98.90% as part of training and testing phases when experimented against frequently used J48, Naïve Bayes, SVM, NN, PART, Random Forest, and Logistic Regression models. Managing outcomes of this research in the early stages could assist people in low-income countries to have a better lifestyle, fewer infections, and minimize expensive hospital visits.
Collapse
|
18
|
An in silico hierarchal approach for drug candidate mining and validation of natural product inhibitors against pyrimidine biosynthesis enzyme in the antibiotic-resistant Shigella flexneri. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 98:105233. [PMID: 35104682 DOI: 10.1016/j.meegid.2022.105233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023]
Abstract
Shigella flexneri is the main causative agent of the communicable diarrheal disease, shigellosis. It is estimated that about 80-165 million cases and > 1 million deaths occur every year due to this disease. S. flexneri causes dysentery mostly in young children, elderly and immunocompromised patients, all over the globe. Recently, due to the emergence of S. flexneri antibiotic resistance strains, it is a dire need to predict novel therapeutic drug targets in the bacterium and screen natural products against it, which could eliminate the curse of antibiotic resistance. Therefore, in current study, available antibiotic-resistant genomes (n = 179) of S. flexneri were downloaded from PATRIC database and a pan-genome and resistome analysis was conducted. Around 5059 genes made up the accessory, 2469 genes made up the core, and 1558 genes made up the unique genome fraction, with 44, 34, and 13 antibiotic-resistant genes in each fraction, respectively. Core genome fraction (27% of the pan-genome), which was common to all strains, was used for subtractive genomics and resulted in 384 non-homologous, and 85 druggable targets. Dihydroorotase was chosen for further analysis and docked with natural product libraries (Ayurvedic and Streptomycin compounds), while the control was orotic acid or vitamin B13 (which is a natural binder of this protein). Dynamics simulation of 50 ns was carried out to validate findings for top-scored inhibitors. The current study proposed dihydroorotase as a significant drug target in S. flexneri and 4-tritriacontanone & patupilone compounds as potent drugs against shigellosis. Further experiments are required to ascertain validity of our findings.
Collapse
|
19
|
Nozaki K, Li L, Miao EA. Innate Sensors Trigger Regulated Cell Death to Combat Intracellular Infection. Annu Rev Immunol 2022; 40:469-498. [PMID: 35138947 PMCID: PMC9614550 DOI: 10.1146/annurev-immunol-101320-011235] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intracellular pathogens pose a significant threat to animals. In defense, innate immune sensors attempt to detect these pathogens using pattern recognition receptors that either directly detect microbial molecules or indirectly detect their pathogenic activity. These sensors trigger different forms of regulated cell death, including pyroptosis, apoptosis, and necroptosis, which eliminate the infected host cell niche while simultaneously promoting beneficial immune responses. These defenses force intracellular pathogens to evolve strategies to minimize or completely evade the sensors. In this review, we discuss recent advances in our understanding of the cytosolic pattern recognition receptors that drive cell death, including NLRP1, NLRP3, NLRP6, NLRP9, NLRC4, AIM2, IFI16, and ZBP1. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kengo Nozaki
- Department of Immunology and Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA;
| | - Lupeng Li
- Department of Immunology and Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA; .,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Edward A Miao
- Department of Immunology and Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA;
| |
Collapse
|
20
|
Tian H, Li B, Xu T, Yu H, Chen J, Yu H, Li S, Zeng L, Huang X, Liu Q. Outer Membrane Vesicles Derived from Salmonella enterica Serotype Typhimurium Can Deliver Shigella flexneri 2a O-Polysaccharide Antigen To Prevent Shigella flexneri 2a Infection in Mice. Appl Environ Microbiol 2021; 87:e0096821. [PMID: 34319809 PMCID: PMC8432525 DOI: 10.1128/aem.00968-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/19/2021] [Indexed: 11/20/2022] Open
Abstract
Shigellosis has become a serious threat to health in many developing countries due to the severe diarrhea it causes. Shigella flexneri 2a is the principal species responsible for this endemic disease. Despite multiple attempts to design a vaccine against shigellosis, no effective vaccine has been developed yet. Lipopolysaccharide (LPS) is both an essential virulence factor and an antigen protective against Shigella, due to its outer domain, termed O-polysaccharide antigen. In the present study, S. flexneri 2a O-polysaccharide antigen was innovatively biosynthesized in Salmonella and attached to core-lipid A via the ligase WaaL, with purified outer membrane vesicles (OMVs) utilized as vaccine vectors. Here, we identified the expression of the heterologous O-antigen and have described the isolation, characterization, and immune protection efficiency of the OMV vaccine. Furthermore, the results of animal experiments indicated that immunization of mice with the OMV vaccine induced significant specific anti-Shigella LPS antibodies in the serum, with similar trends in IgA levels from vaginal secretions and fluid from bronchopulmonary lavage, both intranasally and intraperitoneally. The OMV vaccine derived from both routes of administration provided significant protection against virulent S. flexneri 2a infection, as judged by a serum bactericidal assay, opsonization assay, and challenge test. This vaccination strategy represents a novel and improved approach to control shigellosis by the combination of Salmonella glycosyl carrier lipid bioconjugation with OMVs. IMPORTANCEShigella, the cause of shigellosis or bacillary dysentery, is a major public health concern, especially for children in developing countries. An effective vaccine would control the spread of the disease to some extent. However, no licensed vaccine against Shigella infection in humans has so far been developed. The Shigella O-antigen polysaccharide is effective in stimulating the production of protective antibodies and so could represent a vaccine antigen candidate. In addition, bacterial outer membrane vesicles (OMVs) have been used as antigen delivery platforms due to their nanoscale properties and ease of antigen delivery to trigger an immune response. Therefore, the present study provides a new strategy for vaccine design, combining a glycoconjugated vaccine with OMVs. The design concept of this strategy is the expression of Shigella O-antigen via the LPS synthesis pathway in recombinant Salmonella, from which the OMV vaccine is then isolated. Based on these findings, we believe that the novel vaccine design strategy in which polysaccharide antigens are delivered via bacterial OMVs will be effective for the development and clinical application of an effective Shigella vaccine.
Collapse
Affiliation(s)
- Huizhen Tian
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Biaoxian Li
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Tian Xu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Haolin Yu
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, China
| | - Jingxuan Chen
- School of Ophthalmology and Optometry, Nanchang University, Nanchang, China
| | - Haiyan Yu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Shan Li
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Lingbing Zeng
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
21
|
Walker R, Kaminski RW, Porter C, Choy RKM, White JA, Fleckenstein JM, Cassels F, Bourgeois L. Vaccines for Protecting Infants from Bacterial Causes of Diarrheal Disease. Microorganisms 2021; 9:1382. [PMID: 34202102 PMCID: PMC8303436 DOI: 10.3390/microorganisms9071382] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022] Open
Abstract
The global diarrheal disease burden for Shigella, enterotoxigenic Escherichia coli (ETEC), and Campylobacter is estimated to be 88M, 75M, and 75M cases annually, respectively. A vaccine against this target trio of enteric pathogens could address about one-third of diarrhea cases in children. All three of these pathogens contribute to growth stunting and have demonstrated increasing resistance to antimicrobial agents. Several combinations of antigens are now recognized that could be effective for inducing protective immunity against each of the three target pathogens in a single vaccine for oral administration or parenteral injection. The vaccine combinations proposed here would result in a final product consistent with the World Health Organization's (WHO) preferred product characteristics for ETEC and Shigella vaccines, and improve the vaccine prospects for support from Gavi, the Vaccine Alliance, and widespread uptake by low- and middle-income countries' (LMIC) public health stakeholders. Broadly protective antigens will enable multi-pathogen vaccines to be efficiently developed and cost-effective. This review describes how emerging discoveries for each pathogen component of the target trio could be used to make vaccines, which could help reduce a major cause of poor health, reduced cognitive development, lost economic productivity, and poverty in many parts of the world.
Collapse
Affiliation(s)
- Richard Walker
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA;
| | - Robert W. Kaminski
- Department of Diarrheal Disease Research, Walter Reed Institute of Research, Silver Spring, MD 20910, USA;
| | - Chad Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910, USA;
| | - Robert K. M. Choy
- Center for Vaccine Innovation and Access, PATH, San Francisco, CA 94108, USA;
| | - Jessica A. White
- Center for Vaccine Innovation and Access, PATH, Seattle, WA 98121, USA; (J.A.W.); (F.C.)
| | - James M. Fleckenstein
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Medicine Service, Saint Louis VA Health Care System, St. Louis, MO 63106, USA
| | - Fred Cassels
- Center for Vaccine Innovation and Access, PATH, Seattle, WA 98121, USA; (J.A.W.); (F.C.)
| | - Louis Bourgeois
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA;
| |
Collapse
|
22
|
Guerrant RL, Bolick DT, Swann JR. Modeling Enteropathy or Diarrhea with the Top Bacterial and Protozoal Pathogens: Differential Determinants of Outcomes. ACS Infect Dis 2021; 7:1020-1031. [PMID: 33901398 PMCID: PMC8154416 DOI: 10.1021/acsinfecdis.0c00831] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Developing effective
therapeutics or preventive interventions for
important health threats is greatly enhanced whenever accessible models
can enable the assessment of clinically important outcomes. While
no non-human model is ever perfect, inexpensive in vivo small animal models in such as mice are often of great help in assessing
the relevant efficacy of potential interventions. In addition to acute
diarrhea, the long-term growth and developmental effects of enteric
infections, with or without overt diarrhea, are increasingly recognized.
To address these diverse effects, inexpensive animal models are proving
to be very helpful. Herein, we review the major clinical concerns
with enteric parasitic and bacterial infections that are extremely
common worldwide, especially in vulnerable young children living in
impoverished areas, and the recently published murine models of these
infections and their outcomes. We find that common dietary deficiencies
seen in children in developing areas have striking effects on diarrhea
and enteropathy outcomes in mice. However, these effects differ with
different pathogens. Specifically, the effects of protein or zinc
deficiency differ considerably with different major protozoal and
bacterial pathogens, suggesting different pathogenetic pathways and
intervention effects. The pathogens reviewed are the seven top parasitic
and bacterial pathogens seen in children, namely, Cryptosporidium, Giardia, Campylobacter, Shigella, enterotoxigenic Escherichia coli (ETEC), enteroaggregative E. coli (EAEC), and enteropathogenic E. coli (EPEC).
Collapse
Affiliation(s)
- Richard L. Guerrant
- Center for Global Health Equity, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - David T. Bolick
- Center for Global Health Equity, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Jonathan R. Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, United Kingdom
- Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
23
|
Ledwaba SE, Costa DVS, Bolick DT, Giallourou N, Medeiros PHQS, Swann JR, Traore AN, Potgieter N, Nataro JP, Guerrant RL. Enteropathogenic Escherichia coli Infection Induces Diarrhea, Intestinal Damage, Metabolic Alterations, and Increased Intestinal Permeability in a Murine Model. Front Cell Infect Microbiol 2020; 10:595266. [PMID: 33392105 PMCID: PMC7773950 DOI: 10.3389/fcimb.2020.595266] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Enteropathogenic E. coli (EPEC) are recognized as one of the leading bacterial causes of infantile diarrhea worldwide. Weaned C57BL/6 mice pretreated with antibiotics were challenged orally with wild-type EPEC or escN mutant (lacking type 3 secretion system) to determine colonization, inflammatory responses and clinical outcomes during infection. Antibiotic disruption of intestinal microbiota enabled efficient colonization by wild-type EPEC resulting in growth impairment and diarrhea. Increase in inflammatory biomarkers, chemokines, cellular recruitment and pro-inflammatory cytokines were observed in intestinal tissues. Metabolomic changes were also observed in EPEC infected mice with changes in tricarboxylic acid (TCA) cycle intermediates, increased creatine excretion and shifts in gut microbial metabolite levels. In addition, by 7 days after infection, although weights were recovering, EPEC-infected mice had increased intestinal permeability and decreased colonic claudin-1 levels. The escN mutant colonized the mice with no weight loss or increased inflammatory biomarkers, showing the importance of the T3SS in EPEC virulence in this model. In conclusion, a murine infection model treated with antibiotics has been developed to mimic clinical outcomes seen in children with EPEC infection and to examine potential roles of selected virulence traits. This model can help in further understanding mechanisms involved in the pathogenesis of EPEC infections and potential outcomes and thus assist in the development of potential preventive or therapeutic interventions.
Collapse
Affiliation(s)
- Solanka E. Ledwaba
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - Deiziane V. S. Costa
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - David T. Bolick
- Center for Global Health, Division of Infectious Disease and International Health, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Natasa Giallourou
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, London, England
| | | | - Jonathan R. Swann
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College, London, England
| | - Afsatou N. Traore
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - Natasha Potgieter
- Department of Microbiology, University of Venda, Thohoyandou, South Africa
| | - James P. Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Richard L. Guerrant
- Center for Global Health, Division of Infectious Disease and International Health, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
24
|
Mitchell PS, Roncaioli JL, Turcotte EA, Goers L, Chavez RA, Lee AY, Lesser CF, Rauch I, Vance RE. NAIP-NLRC4-deficient mice are susceptible to shigellosis. eLife 2020; 9:e59022. [PMID: 33074100 PMCID: PMC7595732 DOI: 10.7554/elife.59022] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022] Open
Abstract
Bacteria of the genus Shigella cause shigellosis, a severe gastrointestinal disease that is a major cause of diarrhea-associated mortality in humans. Mice are highly resistant to Shigella and the lack of a tractable physiological model of shigellosis has impeded our understanding of this important human disease. Here, we propose that the differential susceptibility of mice and humans to Shigella is due to mouse-specific activation of the NAIP-NLRC4 inflammasome. We find that NAIP-NLRC4-deficient mice are highly susceptible to oral Shigella infection and recapitulate the clinical features of human shigellosis. Although inflammasomes are generally thought to promote Shigella pathogenesis, we instead demonstrate that intestinal epithelial cell (IEC)-specific NAIP-NLRC4 activity is sufficient to protect mice from shigellosis. In addition to describing a new mouse model of shigellosis, our results suggest that the lack of an inflammasome response in IECs may help explain the susceptibility of humans to shigellosis.
Collapse
Affiliation(s)
- Patrick S Mitchell
- Division of Immunology & Pathogenesis, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Justin L Roncaioli
- Division of Immunology & Pathogenesis, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Elizabeth A Turcotte
- Division of Immunology & Pathogenesis, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Lisa Goers
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Department of Medicine, Division of Infectious Diseases, Massachusetts General HospitalBostonUnited States
| | - Roberto A Chavez
- Division of Immunology & Pathogenesis, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Angus Y Lee
- Cancer Research Laboratory, University of California, BerkeleyBerkeleyUnited States
| | - Cammie F Lesser
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Department of Medicine, Division of Infectious Diseases, Massachusetts General HospitalBostonUnited States
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health and Science UniversityPortlandUnited States
| | - Russell E Vance
- Division of Immunology & Pathogenesis, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Cancer Research Laboratory, University of California, BerkeleyBerkeleyUnited States
- Immunotherapeutics and Vaccine Research Initiative, University of California, BerkeleyBerkeleyUnited States
- Howard Hughes Medical Institute, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
25
|
Wei M, Deng Z, Liu B, Ye W, Fan Y, Liu R, Li J. Investigation of amino acids and minerals in Chinese breast milk. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:3920-3931. [PMID: 32329067 DOI: 10.1002/jsfa.10434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/13/2020] [Accepted: 04/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND The nutrients in human milk, particularly amino acids and minerals, are important for infant growth and development. Since there are few reports of amino acids and minerals in Chinese breast milk, we conducted this study to establish a representative preliminary database of breast milk nutrients in Chinese breast milk. In this study, we collected breast milk from healthy mothers in seven cities in western, southern and central China. The composition, content and proportion of total amino acids and ten elements (potassium, sodium, calcium, magnesium, iron, zinc, manganese, copper, selenium and phosphorus) in human milk in different lactation stages were investigated. RESULTS In this study, it was found that the content of total essential amino acids (671.47 mg 100 mL-1 ) in Chinese breast milk was higher compared with the European Society for Paediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) (574 mg 100 mL-1 ), but the content of leucine (LEU) (129.01 mg 100 mL-1 ) and cysteine (CYS) (20.31 mg 100 mL-1 ) was much lower than that recommended by ESPGHAN. Moreover, it was found that the content of most of these ten elements decreased during lactation, and the content of calcium in Chinese breast milk was lower compared with ESPGHAN. In addition, the content of selenium (7.23-20.55 mg 1000 mL-1 ) in breast milk from the three cities Nanchang, Shanghai and Guangzhou in China was much higher than that recommended by ESPGHAN. CONCLUSIONS In a word, amino acids and minerals in Chinese human milk showed a significant difference from other countries. Human milk meal or infant food should be regulated to meet the requirements of the infant and to maintain the balance of the amino acids and minerals. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Meng Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Biao Liu
- Yili Group Milk Powder Division Research and Development, Hohhot, China
| | - Wenhui Ye
- Yili Group Milk Powder Division Research and Development, Hohhot, China
| | - Yawei Fan
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Rong Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Jing Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Cai R, Cheng C, Chen J, Xu X, Ding C, Gu B. Interactions of commensal and pathogenic microorganisms with the mucus layer in the colon. Gut Microbes 2020; 11:680-690. [PMID: 32223365 PMCID: PMC7524288 DOI: 10.1080/19490976.2020.1735606] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/17/2020] [Accepted: 02/17/2020] [Indexed: 02/03/2023] Open
Abstract
The intestinal mucosal barrier, which is composed of epithelial cells and mucus layers secreted by goblet cells and contains commensal bacteria, constitutes the first line of defense against pathogenic gut microbiota. However, homeostasis between the microbiota and mucus layer is easily disrupted by certain factors, resulting in alteration of the gut microbiota and entry of pathogens to the intestinal mucosal barrier. In this review, we describe the structures and functions of the mucus layer, expound several crucial influencing factors, including diet styles, medications and host genetics, and discuss how pathogenic microorganisms interact with the mucus layer and commensal microbiota, with the understanding that unraveling their complex interactions under homeostatic and dysbiosis conditions in the colon would help reveal some underlying pathogenic mechanisms and thus develop new strategies to prevent pathogenic microbiological colonization.
Collapse
Affiliation(s)
- Rui Cai
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chen Cheng
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | | | | | - Chao Ding
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bing Gu
- Medical Technology School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
27
|
Costa PN, Soares AM, Filho JQ, Junior FS, Ambikapathi R, Rogawski McQuade ET, Guerrant RL, Caulfield LE, Lima AAM, Maciel BLL. Dietary intake from complementary feeding is associated with intestinal barrier function and environmental enteropathy in Brazilian children from the MAL-ED cohort study. Br J Nutr 2020; 123:1003-1012. [PMID: 31964426 PMCID: PMC7282865 DOI: 10.1017/s0007114520000215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/26/2019] [Accepted: 01/07/2020] [Indexed: 12/19/2022]
Abstract
A child's diet contains nutrients and other substances that influence intestinal health. The present study aimed to evaluate the relations between complementary feeding, intestinal barrier function and environmental enteropathy (EE) in infants. Data from 233 children were obtained from the Brazilian site of the Etiology, Risk Factors, and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development Project cohort study. Habitual dietary intake from complementary feeding was estimated using seven 24-h dietary recalls, from 9 to 15 months of age. Intestinal barrier function was assessed using the lactulose-mannitol test (L-M), and EE was determined as a composite measure using faecal biomarkers concentrations - α-1-antitrypsin, myeloperoxidase (MPO) and neopterin (NEO) at 15 months of age. The nutrient adequacies explored the associations between dietary intake and the intestinal biomarkers. Children showed adequate nutrient intakes (with the exception of fibre), impaired intestinal barrier function and intestinal inflammation. There was a negative correlation between energy adequacy and L-M (ρ = -0·19, P < 0·05) and between folate adequacy and NEO concentrations (ρ = -0·21, P < 0·01). In addition, there was a positive correlation between thiamine adequacy and MPO concentration (ρ = 0·22, P < 0·01) and between Ca adequacy and NEO concentration (ρ = 0·23; P < 0·01). Multiple linear regression models showed that energy intakes were inversely associated with intestinal barrier function (β = -0·19, P = 0·02), and fibre intake was inversely associated with the EE scores (β = -0·20, P = 0·04). Findings suggest that dietary intake from complementary feeding is associated with decreased intestinal barrier function and EE in children.
Collapse
Affiliation(s)
- P. N. Costa
- Nutrition Post-Graduation Program, Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - A. M. Soares
- Department of Physiology and Pharmacology, INCT – Instituto de Biomedicina do Semiárido Brasileiro (IBISAB), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - J. Q. Filho
- Department of Physiology and Pharmacology, INCT – Instituto de Biomedicina do Semiárido Brasileiro (IBISAB), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - F. S. Junior
- Department of Physiology and Pharmacology, INCT – Instituto de Biomedicina do Semiárido Brasileiro (IBISAB), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - R. Ambikapathi
- Department of Public Health, Purdue University, West Lafayette, IN, USA
| | | | - R. L. Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - L. E. Caulfield
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - A. A. M. Lima
- Department of Public Health, Purdue University, West Lafayette, IN, USA
| | - B. L. L. Maciel
- Nutrition Post-Graduation Program, Department of Nutrition, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| |
Collapse
|
28
|
Kiefer MC, Motyka NI, Clements JD, Bitoun JP. Enterotoxigenic Escherichia coli Heat-Stable Toxin Increases the Rate of Zinc Release from Metallothionein and Is a Zinc- and Iron-Binding Peptide. mSphere 2020; 5:e00146-20. [PMID: 32238569 PMCID: PMC7113584 DOI: 10.1128/msphere.00146-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 03/12/2020] [Indexed: 12/16/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major diarrheal pathogen in children in low- to middle-income countries. Previous studies have identified heat-stable enterotoxin (ST)-producing ETEC as one of the major diarrhea-causing pathogens in children younger than five years. In this study, we examined iron and zinc binding by both human and porcine ST variants and determined how host metallothionein could detoxify ST. We found that ST purified from ETEC culture supernatants eluted as a doublet during C18 reverse-phase chromatography. Leading edge fractions of the ST doublet were found to be devoid of iron, while trailing edge fractions of the ST doublet were found to contain measurable iron. Next, we found that purified ST could be reconstituted with iron under reducing and anaerobic conditions, and iron-bound ST attenuated the induction of cGMP in T84 epithelial cells. Moreover, we demonstrated that supernatants of ETEC 214-4 grown under increasing iron concentrations were only able to induce cGMP at iron concentrations greater than 5 μM. In vitro studies also demonstrated that ST binds zinc, and once bound, zinc removal from ST required denaturing conditions. Zinc-bound ST also failed to induce cGMP. We found that ST contributes disulfide bonds to the perceived oxidized glutathione pool, increases the rate of zinc release from metallothionein, and can be detoxified by metallothionein. Lastly, we showed ST induces transcriptional changes in genes previously shown to be regulated by deferoxamine. These studies demonstrate ST ETEC pathogenesis may be tied intimately to host mucosal metal status.IMPORTANCE Enterotoxigenic Escherichia coli (ETEC) is a major diarrheal pathogen in children in low- to middle-income countries, deployed military personnel, and travelers to regions of endemicity. The heat-stable toxin (ST) is a small nonimmunogenic secreted peptide with 3 disulfide bonds. It has been appreciated that dietary disulfides modulate intestinal redox potential and that ST could be detoxified using exogenous reductants. Using biochemical and spectroscopic approaches, we demonstrated that ST can separately bind iron and zinc under reducing conditions, thereby reducing ST toxicity. Moreover, we demonstrated that ST modulates the glutathione (GSH)/oxidized glutathione (GSSG) ratio and that ST should be considered a toxin oxidant. ST can be detoxified by oxidizing zinc-loaded metallothionine, causing free zinc to be released. These studies help lay a foundation to understand how diarrheal pathogens modulate intestinal redox potential and may impact how we design therapeutics and/or vaccines for the pathogens that produce them.
Collapse
Affiliation(s)
- Mallory C Kiefer
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Natalya I Motyka
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - John D Clements
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jacob P Bitoun
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
29
|
Wu YHS, Lin YL, Huang C, Chiu CH, Nakthong S, Chen YC. Cardiac protection of functional chicken-liver hydrolysates on the high-fat diet induced cardio-renal damages via sustaining autophagy homeostasis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:2443-2452. [PMID: 31951016 DOI: 10.1002/jsfa.10261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/02/2020] [Accepted: 01/10/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Cardio-renal syndrome (CRS) is an integrative problem related to chronic malnutrition, obesity, etc. Amino acids and peptides are regarded as protective and essential for tissues. Pepsin-digested chicken liver hydrolysates (CLHs), which are made from the byproducts of the poultry industry, are amino-acid based and of animal origin, and may be protective against the myocardial and renal damage induced by a high-fat diet (HFD). RESULTS Our results showed that CLHs contain large quantities of anserine, taurine, and branched-chain amino acids (BCAAs), and supplementing the diet with CLHs reduced (P < 0.05) weight gain, liver weight, peri-renal fat mass / adipocyte-area sizes, serum total cholesterol (TC), aspartate aminotransferase (AST), and low-density lipoprotein cholesterol (LDLC) levels in HFD-fed mice but increased (P < 0.05) serum high-density lipoprotein cholesterol (HDLC) levels. By histological analyses, CLHs alleviated (P < 0.05) renal lipid deposition and fibrosis, as well as cardiac fibrosis and inflammation of HFD-fed mice. Meanwhile, increased (P < 0.05) inflammatory and fibrotic cytokines levels in the myocardia of the HFD-fed mice were downregulated (P < 0.05) by CLH supplementation. Regarding autophagy-related protein levels, protective effects of CLHs on the myocardia against HFD feeding may result from the early blockade of the autophagy pathway to prevent autophagosome accumulation. CONCLUSION Functional CLHs could be a novel food ingredient as a cardio-renal protective agent against a high-fat dietary habit in a niche market. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yi-Hsieng Samuel Wu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Ling Lin
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Chien Huang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Chih-Hsien Chiu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Sasitorn Nakthong
- Department of Animal Science, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University, Nakhon Pathom, Thailand
| | - Yi-Chen Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
30
|
Medeiros PHQS, Bolick DT, Ledwaba SE, Kolling GL, Costa DVS, Oriá RB, Lima AÂM, Barry EM, Guerrant RL. A bivalent vaccine confers immunogenicity and protection against Shigella flexneri and enterotoxigenic Escherichia coli infections in mice. NPJ Vaccines 2020; 5:30. [PMID: 32257392 PMCID: PMC7101394 DOI: 10.1038/s41541-020-0180-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Vaccine studies for Shigella flexneri and enterotoxigenic Escherichia coli have been impaired by the lack of optimal animal models. We used two murine models to show that a S. flexneri 2a bivalent vaccine (CVD 1208S-122) expressing enterotoxigenic Escherichia coli colonization factor antigen-I (CFA/I) and the binding subunits A2 and B of heat labile-enterotoxin (LTb) is immunogenic and protects against weight loss and diarrhea. These findings document the immunogenicity and pre-clinical efficacy effects of CVD 1208S-122 vaccine and suggest that further work can help elucidate relevant immune responses and ultimately its clinical efficacy in humans.
Collapse
Affiliation(s)
- Pedro Henrique Q S Medeiros
- 1Center for Global Health and Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA USA.,2Institute of Biomedicine, Federal University of Ceará, Fortaleza, CE Brazil
| | - David T Bolick
- 1Center for Global Health and Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA USA
| | - Solanka E Ledwaba
- 1Center for Global Health and Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA USA.,3Department of Microbiology, University of Venda, Thohoyandou, Limpopo province South Africa
| | - Glynis L Kolling
- 1Center for Global Health and Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA USA
| | - Deiziane V S Costa
- 1Center for Global Health and Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA USA.,2Institute of Biomedicine, Federal University of Ceará, Fortaleza, CE Brazil
| | - Reinaldo B Oriá
- 1Center for Global Health and Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA USA.,2Institute of Biomedicine, Federal University of Ceará, Fortaleza, CE Brazil
| | - Aldo Ângelo M Lima
- 2Institute of Biomedicine, Federal University of Ceará, Fortaleza, CE Brazil
| | - Eileen M Barry
- 4Center for Vaccine Development and Global Health, University of Maryland, Baltimore, MD USA
| | - Richard L Guerrant
- 1Center for Global Health and Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA USA
| |
Collapse
|
31
|
Yang J, Chen W, Xia P, Zhang W. Dynamic comparison of gut microbiota of mice infected with Shigella flexneri via two different infective routes. Exp Ther Med 2020; 19:2273-2281. [PMID: 32104294 PMCID: PMC7027338 DOI: 10.3892/etm.2020.8469] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
Abstract
Shigella is one of the main pathogens causing diarrheal disease, and is associated with high morbidity and mortality in developing countries. Previous clinical data and animal studies have shown that the outcomes of oral and peritoneal infections of Shigella differ, and that the latter is more serious. Furthermore, a variety of pathogenic bacteria are known to cause changes in intestinal flora after infection, and the influence of Shigella infection on intestinal flora remains poorly understood. In the present study, the 16S rRNA high-throughput sequencing method was used to compare the changes in gut microbiota profiles in feces of mice infected with Shigella via two routes. In addition, the present study investigated the association between the differences in infection performance and bacterial communities. The present results suggested that the intraperitoneal route induced a distinct decrease in α-diversity in the fecal microbiota when compared to the control at a later time, while the effect of the oral route on α-diversity was not obvious. Oral infection of Shigella had a rapid and significant effect on gut microbiota, mainly causing a decreased abundance of Lactobacillus and an increased abundance of Prevotella and Escherichia/Shigella in the early stage of infection. By contrast, the effect of intraperitoneal infection on the gut microbiota was relatively slow and small. The principal coordinate analysis results suggested that the dynamic profile of gut microbiota between the two infective routes was consistent with the infection process. Probiotics, such as Lactobacillus reuteri and Faecalitalea exhibited significantly reduced abundance after Shigella infection. Collectively, the present results suggested that gut microbiota may play a pivotal role in the pathogenesis of Shigella infection. Future studies should investigate the effect of Shigella infection on the interaction between pathogenic bacteria and intestinal flora. The present results suggested that the use of probiotics may facilitate the prevention and treatment of shigellosis.
Collapse
Affiliation(s)
- Jinsong Yang
- Fujian Provincial Key Laboratory of Environment Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, P.R. China
| | - Wei Chen
- Department for Viral Disease Control and Prevention, Fujian Center for Disease Control and Prevention, Fuzhou, Fujian 350001, P.R. China
| | - Pinchang Xia
- Department for Viral Disease Control and Prevention, Fujian Center for Disease Control and Prevention, Fuzhou, Fujian 350001, P.R. China
| | - Wenchang Zhang
- Fujian Provincial Key Laboratory of Environment Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
32
|
Mukhopadhyay S, Ganguli S, Chakrabarti S. <em>Shigella</em> pathogenesis: molecular and computational insights. AIMS MOLECULAR SCIENCE 2020. [DOI: 10.3934/molsci.2020007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
33
|
Anti-Shigellosis Activity of Cola anomala Water/Ethanol Pods Extract on Shigella flexneri-Induced Diarrhea in Rats. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6706230. [PMID: 31828116 PMCID: PMC6881571 DOI: 10.1155/2019/6706230] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/10/2019] [Accepted: 10/15/2019] [Indexed: 12/02/2022]
Abstract
This study was undertaken to evaluate the activities of water/ethanol Cola anomala pods extract. In vitro antimicrobial susceptibility was determined by the disk diffusion method; the minimum inhibitory concentration and minimum bactericidal concentration were determined by agar dilution technique. In vivo, shigellosis was induced in healthy Wistar albino rats by oral administration of Shigella flexneri inoculum, 12 × 108 CFU/mL. At the onset of diarrhea, infected and normal control animals were subdivided into various groups treated with distilled water, with water/ethanol Cola anomala pods extract at 25, 50, or 100 mg/kg, or with ciprofloxacin, 2.5 mg/kg. After one-week treatment, rats were sacrificed, and blood and colon were collected. Blood was used for blood cell count. A portion of the colon served for histological studies while homogenate from the remaining part was centrifuged and the supernatant was collected for the determination of NO, PGE2, IL-1β, and TNF-α levels. In vitro, water/ethanol Cola anomala pods extract showed to be bactericidal, with a minimum inhibitory concentration of 2.0 mg/mL and a minimum bactericidal concentration of 3.0 mg/mL. In diarrheic rats, the extract significantly (P < 0.01) increased the white blood cells and significantly (P < 0.01) decreased stool Shigella density from the first to the seventh day of treatment. It partially restored the structure of eroded intestine epithelium and prevented weight loss; the dose dependently and significantly (P < 0.001) decreased NO, IL-1β, and TNF-α production in the colon and was found to have no significant effect on PGE2 production. These results support the use of this plant in traditional medicine in the treatment of gastrointestinal ailments.
Collapse
|
34
|
Evaluation of in vitro and in vivo antibiotic efficacy against a novel bioluminescent Shigella flexneri. Sci Rep 2019; 9:13567. [PMID: 31537849 PMCID: PMC6753072 DOI: 10.1038/s41598-019-49729-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/30/2019] [Indexed: 12/13/2022] Open
Abstract
Shigella spp., the bacteria responsible for shigellosis, are one of the leading causes of diarrheal morbidity and mortality amongst children. There is a pressing need for the development of novel therapeutics, as resistance of Shigella to many currently used antibiotics is rapidly emerging. This paper describes the development of robust in vitro and in vivo tools to study antibiotic efficacy against Shigella flexneri. A novel bioluminescent S. flexneri strain (S. flexneri lux1) was generated, which can be used in a mammalian epithelial cell co-culture assay to evaluate antibiotic intracellular and extracellular efficacy. In addition, the S. flexneri lux1 strain was used with an intraperitoneal (IP) murine model of shigellosis to test the efficacy of ciprofloxacin and ampicillin. Both antibiotics significantly reduced the observed radiance from the gastrointestinal tissue of infected mice compared to vehicle control. Furthermore, plated gastrointestinal tissue homogenate confirmed antibiotic treatment significantly reduced the S. flexneri infection. However, in contrast to the results generated with tissue homogenate, the radiance data was not able to distinguish between the efficacy of ampicillin and ciprofloxacin. Compared to traditional methods, these models can be utilized for efficient screening of novel antibiotics aiding in the discovery of new treatments against shigellosis.
Collapse
|
35
|
Salameh E, Morel FB, Zeilani M, Déchelotte P, Marion-Letellier R. Animal Models of Undernutrition and Enteropathy as Tools for Assessment of Nutritional Intervention. Nutrients 2019; 11:nu11092233. [PMID: 31527523 PMCID: PMC6770013 DOI: 10.3390/nu11092233] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/24/2019] [Accepted: 09/08/2019] [Indexed: 02/06/2023] Open
Abstract
: Undernutrition is a major public health problem leading to 1 in 5 of all deaths in children under 5 years. Undernutrition leads to growth stunting and/or wasting and is often associated with environmental enteric dysfunction (EED). EED mechanisms leading to growth failure include intestinal hyperpermeability, villus blunting, malabsorption and gut inflammation. As non-invasive methods for investigating gut function in undernourished children are limited, pre-clinical models are relevant to elucidating the pathophysiological processes involved in undernutrition and EED, and to identifying novel therapeutic strategies. In many published models, undernutrition was induced using protein or micronutrient deficient diets, but these experimental models were not associated with EED. Enteropathy models mainly used gastrointestinal injury triggers. These models are presented in this review. We found only a few studies investigating the combination of undernutrition and enteropathy. This highlights the need for further developments to establish an experimental model reproducing the impact of undernutrition and enteropathy on growth, intestinal hyperpermeability and inflammation, that could be suitable for preclinical evaluation of innovative therapeutic intervention.
Collapse
Affiliation(s)
- Emmeline Salameh
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
- Nutriset SAS, 76770 Malaunay, France.
| | | | | | - Pierre Déchelotte
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
- Department of Nutrition, Rouen University Hospital, 76183 Rouen, France.
| | - Rachel Marion-Letellier
- UniRouen, Inserm UMR 1073 Nutrition, Inflammation and Gut-Brain Axis, Normandie University, 76183 Rouen, France.
| |
Collapse
|