1
|
Kruger K, Myeonghyun Y, van der Wielen N, Kok DE, Hooiveld GJ, Keshtkar S, Diepeveen-de Bruin M, Balvers MGJ, Grootte-Bromhaar M, Mudde K, Ly NTHN, Vermeiren Y, de Groot LCPGM, de Vos RCH, Gonzales GB, Steegenga WT, van Trijp MPH. Evaluation of inter- and intra-variability in gut health markers in healthy adults using an optimised faecal sampling and processing method. Sci Rep 2024; 14:24580. [PMID: 39427011 PMCID: PMC11490648 DOI: 10.1038/s41598-024-75477-z] [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: 07/26/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Despite advances in gut health research, the variability of important gut markers within individuals over time remains underexplored. We investigated the intra-individual variation of various faecal gut health markers using an optimised processing protocol aimed at reducing variability. Faecal samples from ten healthy adults over three consecutive days demonstrated marker-specific intra-individual coefficients of variation (CV%), namely: stool consistency (16.5%), water content (5.7%), pH (3.9%), total SCFAs (17.2%), total BCFAs (27.4%), total bacteria and fungi copies (40.6% and 66.7%), calprotectin and myeloperoxidase (63.8% and 106.5%), and untargeted metabolites (on average 40%). For thirteen microbiota genera, including Bifidobacterium and Akkermansia, variability exceeded 30%, whereas microbiota diversity was less variable (Phylogenetic Diversity 3.3%, Inverse Simpson 17.2%). Mill-homogenisation of frozen faeces significantly reduced the replicates CV% for total SCFAs (20.4-7.5%) and total BCFAs (15.9-7.8%), and untargeted metabolites compared to faecal hammering only, without altering mean concentrations. Our results show the potential need for repeated sampling to accurately represent specific gut health markers. We also demonstrated the effectiveness of optimised preprocessing of human stool samples in reducing overall analytical variability.
Collapse
Affiliation(s)
- Kirsten Kruger
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Yoou Myeonghyun
- Clinical Microbiomics, Copenhagen, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Nicky van der Wielen
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Dieuwertje E Kok
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Guido J Hooiveld
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Shohreh Keshtkar
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Michiel G J Balvers
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Mechteld Grootte-Bromhaar
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Karin Mudde
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Nhien T H N Ly
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Yannick Vermeiren
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Lisette C P G M de Groot
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Ric C H de Vos
- Bioscience, Wageningen Plant Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Gerard Bryan Gonzales
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Wilma T Steegenga
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Mara P H van Trijp
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
2
|
Hosomi K, Hatanaka N, Hinenoya A, Adachi J, Tojima Y, Furuta M, Uchiyama K, Morita M, Nagatake T, Saika A, Kawai S, Yoshii K, Kondo S, Yamasaki S, Kunisawa J. QcrC is a potential target for antibody therapy and vaccination to control Campylobacter jejuni infection by suppressing its energy metabolism. Front Microbiol 2024; 15:1415893. [PMID: 39015740 PMCID: PMC11250076 DOI: 10.3389/fmicb.2024.1415893] [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: 04/11/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Introduction Campylobacter spp. are a public health concern, yet there is still no effective vaccine or medicine available. Methods Here, we developed a Campylobacter jejuni-specific antibody and found that it targeted a menaquinol cytochrome c reductase complex QcrC. Results The antibody was specifically reactive to multiple C. jejuni strains including clinical isolates from patients with acute enteritis and was found to inhibit the energy metabolism and growth of C. jejuni. Different culture conditions produced different expression levels of QcrC in C. jejuni, and these levels were closely related not only to the energy metabolism of C. jejuni but also its pathogenicity. Furthermore, immunization of mice with recombinant QcrC induced protective immunity against C. jejuni infection. Discussion Taken together, our present findings highlight a possible antibody- or vaccination-based strategy to prevent or control Campylobacter infection by targeting the QcrC-mediated metabolic pathway.
Collapse
Affiliation(s)
- Koji Hosomi
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Noritoshi Hatanaka
- Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
- Asian Health Science Research Institute, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Atsushi Hinenoya
- Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
- Asian Health Science Research Institute, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, NIBIOHN, Osaka, Japan
| | - Yoko Tojima
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Mari Furuta
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Keita Uchiyama
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Makiko Morita
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Functional Anatomy, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Japan
| | - Azusa Saika
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Soichiro Kawai
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Ken Yoshii
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Saki Kondo
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Shinji Yamasaki
- Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
- Asian Health Science Research Institute, Osaka Metropolitan University, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Graduate School of Dentistry, Osaka University, Osaka, Japan
- Graduate School of Science, Osaka University, Osaka, Japan
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| |
Collapse
|
3
|
Diez-Martin E, Hernandez-Suarez L, Muñoz-Villafranca C, Martin-Souto L, Astigarraga E, Ramirez-Garcia A, Barreda-Gómez G. Inflammatory Bowel Disease: A Comprehensive Analysis of Molecular Bases, Predictive Biomarkers, Diagnostic Methods, and Therapeutic Options. Int J Mol Sci 2024; 25:7062. [PMID: 39000169 PMCID: PMC11241012 DOI: 10.3390/ijms25137062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/15/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
In inflammatory bowel diseases (IBDs), such as Crohn's disease (CD) and ulcerative colitis (UC), the immune system relentlessly attacks intestinal cells, causing recurrent tissue damage over the lifetime of patients. The etiology of IBD is complex and multifactorial, involving environmental, microbiota, genetic, and immunological factors that alter the molecular basis of the organism. Among these, the microbiota and immune cells play pivotal roles; the microbiota generates antigens recognized by immune cells and antibodies, while autoantibodies target and attack the intestinal membrane, exacerbating inflammation and tissue damage. Given the altered molecular framework, the analysis of multiple molecular biomarkers in patients proves exceedingly valuable for diagnosing and prognosing IBD, including markers like C reactive protein and fecal calprotectin. Upon detection and classification of patients, specific treatments are administered, ranging from conventional drugs to new biological therapies, such as antibodies to neutralize inflammatory molecules like tumor necrosis factor (TNF) and integrin. This review delves into the molecular basis and targets, biomarkers, treatment options, monitoring techniques, and, ultimately, current challenges in IBD management.
Collapse
Affiliation(s)
- Eguzkiñe Diez-Martin
- Research and Development Department, IMG Pharma Biotech S.L., 48170 Zamudio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Leidi Hernandez-Suarez
- Research and Development Department, IMG Pharma Biotech S.L., 48170 Zamudio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Carmen Muñoz-Villafranca
- Department of Gastroenterology, University Hospital of Basurto, Avda Montevideo 18, 48013 Bilbao, Spain
| | - Leire Martin-Souto
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Egoitz Astigarraga
- Research and Development Department, IMG Pharma Biotech S.L., 48170 Zamudio, Spain
| | - Andoni Ramirez-Garcia
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | | |
Collapse
|
4
|
Müller-Hauser AA, Huda TMN, Sobhan S, Lambrecht NJ, Waid JL, Wendt AS, Ali S, Rahman M, Gabrysch S. Effect of a Homestead Food Production and Food Hygiene Intervention on Biomarkers of Environmental Enteric Dysfunction in Children Younger Than 24 Months in Rural Bangladesh: A Cluster-Randomized Controlled Trial. Am J Trop Med Hyg 2023; 109:1166-1176. [PMID: 37783459 PMCID: PMC10622486 DOI: 10.4269/ajtmh.23-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/08/2023] [Indexed: 10/04/2023] Open
Abstract
Poor sanitation and hygiene practices and inadequate diets can contribute to environmental enteric dysfunction (EED). We evaluated the impact of a combined homestead food production and food hygiene intervention on EED biomarkers in young children in rural Bangladesh. The analysis was conducted within the Food and Agricultural Approaches to Reducing Malnutrition (FAARM) cluster-randomized trial in Sylhet, Bangladesh. The FAARM trial enrolled 2,705 married women and their children younger than 3 years of age in 96 settlements (geographic clusters): 48 intervention and 48 control. The 3-year intervention (2015-2018) included training on gardening, poultry rearing, and improved nutrition practices and was supplemented by an 8-month food hygiene behavior change component, implemented from mid-2017. We analyzed data on 574 children age 0 to 24 months with multilevel linear regression. We assessed fecal myeloperoxidase (MPO), neopterin (NEO), and alpha-1-antitrypsin (AAT) as biomarkers of EED, and serum C-reactive protein (CRP) and alpha-1-acid glycoprotein (AGP) as biomarkers of systemic inflammation, using ELISA. There was no intervention effect on NEO, AAT, CRP, and AGP concentrations, but, surprisingly, MPO levels were increased in children of the intervention group (0.11 log ng/mL; 95% CI, 0.001-0.22). This increase was greater with increasing child age and among intervention households with poultry that were not kept in a shed. A combined homestead food production and food hygiene intervention did not decrease EED in children in our study setting. Small-scale poultry rearing promoted by the intervention might be a risk factor for EED.
Collapse
Affiliation(s)
- Anna A. Müller-Hauser
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Public Health, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
| | - Tarique Md. Nurul Huda
- Department of Public Health, College of Public Health and Health Informatics, Qassim University, Al Bukairiyah, Saudi Arabia
- Environmental Interventions Unit, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Shafinaz Sobhan
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Public Health, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
| | - Nathalie J. Lambrecht
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Public Health, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
| | - Jillian L. Waid
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
- Heidelberg Institute of Global Health, Heidelberg University, Heidelberg, Germany
| | - Amanda S. Wendt
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
| | - Shahjahan Ali
- Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
- Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mahbubur Rahman
- Environmental Interventions Unit, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Sabine Gabrysch
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Public Health, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
- Heidelberg Institute of Global Health, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
5
|
Koenderman L, Tesselaar K, Vrisekoop N. Human neutrophil kinetics: a call to revisit old evidence. Trends Immunol 2022; 43:868-876. [PMID: 36243621 DOI: 10.1016/j.it.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 01/12/2023]
Abstract
The half-life of human neutrophils is still controversial, with estimates ranging from 7-9 h to 3.75 days. This debate should be settled to understand neutrophil production in the bone marrow (BM) and the potential and limitations of emergency neutropoiesis following infection or trauma. Furthermore, cellular lifespan greatly influences the potential effect(s) neutrophils have on the adaptive immune response. We posit that blood neutrophils are in exchange with different tissues, but particularly the BM, as it contains the largest pool of mature neutrophils. Furthermore, we propose that the oldest neutrophils are the first to die following a so-called conveyor belt model. These guiding principles shed new light on our interpretation of existing neutrophil lifespan data and offer recommendations for future research.
Collapse
Affiliation(s)
- Leo Koenderman
- Department of Respiratory Medicine, University Medical Center Utrecht, The Netherlands; Center for Translational Immunology, University Medical Center Utrecht, The Netherlands.
| | - Kiki Tesselaar
- Center for Translational Immunology, University Medical Center Utrecht, The Netherlands; Department of Immunology, University Medical Center Utrecht, The Netherlands
| | - Nienke Vrisekoop
- Department of Respiratory Medicine, University Medical Center Utrecht, The Netherlands; Center for Translational Immunology, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
6
|
Diaz JN, Dulience SJL, Wolthausen N, Jiang X, Gyimah E, Marhône Pierre FJ, Kuhlmann FM, Iannotti LL. Choline, DHA, and Diarrheal Disease Associated with Growth Faltering in a Case-Control Study. Curr Dev Nutr 2022; 6:nzac140. [PMID: 36204326 PMCID: PMC9529221 DOI: 10.1093/cdn/nzac140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/17/2022] [Accepted: 09/01/2022] [Indexed: 12/03/2022] Open
Abstract
Background Children with recurrent infectious diarrhea are susceptible to growth faltering. DHA and choline may play a role in this relationship due to their involvement in lipid metabolism, gut immunity, and inflammatory pathways. Objectives This study aimed to characterize the contributions made by DHA and choline status and enteric damage in young children in the association between diarrheal illness and child growth. Methods A longitudinal case-control study was conducted among children aged 6-36 mo (N = 195) in Cap-Haitien, Haiti. Mother-child dyads were recruited from community health posts and outpatient clinics. Cases were defined as children experiencing acute diarrhea within the last 3 d and matched to healthy controls. Child anthropometry, dietary intake, and blood and stool samples were collected at baseline and follow-up. Plasma DHA, choline, and betaine were determined by LC-MS/MS methods (n = 49) and intestinal fatty acid-binding protein (I-FABP) by ELISA (n = 183). Multivariate regression models were applied with mediation analyses to examine associations and adjust for confounding factors. Results At baseline, mean plasma DHA concentrations (1.03 µg/mL; 95% CI: 0.91, 1.15) were not significantly different between cases and controls, nor was there a difference in mean plasma choline concentrations (4.5 µg/mL; 95% CI: 3.8, 5.1). Mean plasma I-FABP concentrations were significantly higher at follow-up in cases (3.34; 95% CI: 3.28, 3.40) than controls (3.20; 95% CI: 3.13, 3.27; P = 0.002). In adjusted multilinear regression models, higher plasma DHA concentrations at follow-up were associated with a negative change in weight-age z score (P = 0.016), and follow-up I-FABP was inversely associated with height-age z score (P = 0.035). No interaction or mediation effects were found. Conclusions I-FABP concentrations were significantly higher in cases as compared with controls at follow-up, suggesting ongoing enteric damage and increased risk for malnutrition. Plasma DHA and I-FABP may have a role in childhood growth outcomes.
Collapse
Affiliation(s)
- Jenna N Diaz
- Department of Pediatrics, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | | | - Noah Wolthausen
- Brown School, Institute for Public Health, Washington University in St Louis, St Louis, Missouri, USA
| | - Xuntian Jiang
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Emmanuel Gyimah
- Brown School, Institute for Public Health, Washington University in St Louis, St Louis, Missouri, USA
| | - Francesca J Marhône Pierre
- Unité de Coordination du Programme National d'Alimentation et de Nutrition, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| | - F Matthew Kuhlmann
- Department of Medicine, Washington University School of Medicine in St Louis, St Louis, Missouri, USA
| | - Lora L Iannotti
- Brown School, Institute for Public Health, Washington University in St Louis, St Louis, Missouri, USA
| |
Collapse
|
7
|
Hasan MM, Gazi MA, Das S, Fahim SM, Hossaini F, Khan AR, Ferdous J, Alam MA, Mahfuz M, Ahmed T. Gut biomolecules (I-FABP, TFF3 and lipocalin-2) are associated with linear growth and biomarkers of environmental enteric dysfunction (EED) in Bangladeshi children. Sci Rep 2022; 12:13905. [PMID: 35974137 PMCID: PMC9381788 DOI: 10.1038/s41598-022-18141-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
In the current world, a major challenge to diagnose environmental enteric dysfunction (EED) is the lack of validated non-invasive biomarkers. Intestine derived molecules, including intestinal fatty acid binding protein (I-FABP), trefoil factor-3 (TFF3), lactoferrin, lipocalin-2 (LCN2), and mucin-2, have been reported as indicators of intestinal inflammation and gut health. Therefore, we aimed to investigate the levels of these bio-molecules as biomarkers of EED among under-2 children in Bangladesh. A total of 140 children were recruited in a case-control design. All the biomarkers were measured by ELISA. Spearman's rank correlation was performed to see the correlation between the biomarkers and the EED score. Moreover, multivariable linear regression was performed to investigate the association of biomarkers with length-for-age z-score (LAZ). TFF3 correlates positively with myeloperoxidase (r = 0.26, p < 0.05) and EED score (r = 0.17, p < 0.05). Likewise, LCN2 correlates positively with myeloperoxidase (r = 0.37, p < 0.05), neopterin (r = 0.33, p < 0.05) and EED score (r = 0.31, p < 0.05). Moreover, multivariable linear regression revealed a negative association of I-FABP with LAZ of the study participants. Our results imply that TFF3 and LCN2 might be promising biomarkers to diagnose intestinal inflammation and EED, while I-FABP is negatively associated with linear growth of Bangladeshi children.
Collapse
Affiliation(s)
- Md Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh.
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Farzana Hossaini
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Ar-Rafi Khan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Jafrin Ferdous
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Md Ashraful Alam
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
- James P. Grant School of Public Health, BRAC University, Dhaka, Bangladesh
- Department of Global Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
8
|
Ledwaba SE, Bolick DT, de Medeiros PHQS, Kolling GL, Traore AN, Potgieter N, Nataro JP, Guerrant RL. Enteropathogenic Escherichia coli (EPEC) expressing a non-functional bundle-forming pili (BFP) also leads to increased growth failure and intestinal inflammation in C57BL/6 mice. Braz J Microbiol 2022; 53:1781-1787. [PMID: 35882715 PMCID: PMC9679052 DOI: 10.1007/s42770-022-00802-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/17/2022] [Indexed: 01/13/2023] Open
Abstract
Bundle-forming pili (BFP) are implicated in the virulence of typical enteropathogenic E. coli (EPEC), resulting in enhanced colonization and mild to severe disease outcomes; hence, non-functional BFP may have a major influence on disease outcomes in vivo. Weaned antibiotic pre-treated C57BL/6 mice were orally infected with EPEC strain UMD901 (E2348/69 bfpA C129S); mice were monitored daily for body weight; stool specimens were collected daily; and intestinal tissues were collected at the termination of the experiment on day 3 post-infection. Real-time PCR was used to quantify fecal shedding and tissue burden. Intestinal inflammatory biomarkers lipocalin-2 (LCN-2) and myeloperoxidase (MPO) were also assessed. Infection caused substantial body weight loss, bloody diarrhea, and intestinal colonization with fecal and intestinal tissue inflammatory biomarkers that were comparable to those previously published with the wild-type typical EPEC strain. Here we further report on the evaluation of an EPEC infection model, showing how disruption of bfp function does not impair, and may even worsen diarrhea, colonization, and intestinal disruption and inflammation. More research is needed to understand the role of bfp in pathogenicity of EPEC infections in vivo.
Collapse
Affiliation(s)
- Solanka Ellen Ledwaba
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Thohoyandou, Limpopo Province South Africa
| | - David Thomas Bolick
- Division of Infectious Disease and International Health, School of Medicine, University of Virginia, Charlottesville, VA USA
| | | | - Glynis Luanne Kolling
- Division of Infectious Disease and International Health, School of Medicine, University of Virginia, Charlottesville, VA USA ,Department of Biomedical Engineering, University of Virgina, Charlottesville, VA USA
| | - Afsatou Ndama Traore
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Thohoyandou, Limpopo Province South Africa
| | - Natasha Potgieter
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Thohoyandou, Limpopo Province South Africa
| | - James Paul Nataro
- Department of Pediatrics, School of Medicine, University of Virginia, Charlottesville, VA USA
| | - Richard Littleton Guerrant
- Division of Infectious Disease and International Health, School of Medicine, University of Virginia, Charlottesville, VA USA
| |
Collapse
|
9
|
Gleeson JP, Chaudhary N, Fein KC, Doerfler R, Hredzak-Showalter P, Whitehead KA. Profiling of mature-stage human breast milk cells identifies six unique lactocyte subpopulations. SCIENCE ADVANCES 2022; 8:eabm6865. [PMID: 35767604 PMCID: PMC9242445 DOI: 10.1126/sciadv.abm6865] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/11/2022] [Indexed: 06/15/2023]
Abstract
Breast milk is chock-full of nutrients, immunological factors, and cells that aid infant development. Maternal cells are the least studied breast milk component, and their unique properties are difficult to identify using traditional techniques. Here, we characterized the cells in mature-stage breast milk from healthy donors at the protein, gene, and transcriptome levels. Holistic analysis of flow cytometry, quantitative polymerase chain reaction, and single-cell RNA sequencing data identified the predominant cell population as epithelial with smaller populations of macrophages and T cells. Two percent of epithelial cells expressed four stem cell markers: SOX2, TRA-1-60, NANOG, and SSEA4. Furthermore, milk contained six distinct epithelial lactocyte subpopulations, including three previously unidentified subpopulations programmed toward mucosal defense and intestinal development. Pseudotime analysis delineated the differentiation pathways of epithelial progenitors. Together, these data define healthy human maternal breast milk cells and provide a basis for their application in maternal and infant medicine.
Collapse
Affiliation(s)
- John P. Gleeson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Katherine C. Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Rose Doerfler
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
10
|
El Wakeel MA, El-Kassas GM, Fouad Ahmed G, Ali WH, Elsheikh EM, El-Zayat SR, Fadl NN, Kamel EH, Rabah TM. Fecal Markers of Environmental Enteric Dysfunction and their Relation to Faltering Growth in a Sample of Egyptian Children. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Chronic malnutrition is a long-term health condition that has threatening effects on children’s health. Environmental enteric dysfunction (EED) is a subclinical disorder affecting the small intestine that may occur due to exposure to environmental pathogens and toxins.
AIM: The present research was intended to detect the value of fecal biomarkers of intestinal epithelial damage alpha-1anti-trypsin (AAT) and intestinal inflammation Myeloperoxidase (MPO) and Neopetrin (NEO), also to quantify their association with faltering growth in stunted and underweight children.
PATIENTS AND METHODS: This case–control study included 105 children with moderate malnutrition as a case group and 100 children of normal body weight and height as a control group. Quantification of fecal markers levels of intestinal permeability AAT and intestinal inflammation (NEO and MPO) along with serum micronutrients levels (iron and zinc) in children with malnutrition in comparison to controls.
RESULTS: Fecal markers of intestinal permeability AAT and intestinal inflammation NEO had statistically significant higher levels in children with malnutrition, while serum micronutrients (iron and zinc) had statistically significant lower levels in children with malnutrition.
CONCLUSION: Faltering growth is associated with elevated fecal markers of intestinal permeability AAT and intestinal inflammation NEO. EED may be a cause for faltering growth.
Collapse
|
11
|
Hasan MM, Gazi MA, Das S, Fahim SM, Hossaini F, Alam MA, Mahfuz M, Ahmed T. Association of lipocalin-2 and low-density lipoprotein receptor-related protein-1 (LRP1) with biomarkers of environmental enteric dysfunction (EED) among under 2 children in Bangladesh: results from a community-based intervention study. BMJ Paediatr Open 2021; 5:e001138. [PMID: 34423140 PMCID: PMC8340289 DOI: 10.1136/bmjpo-2021-001138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/17/2021] [Indexed: 11/03/2022] Open
Abstract
Background Environmental enteric dysfunction (EED) is thought to occur from persistent intestinal inflammation. Studies also revealed the association of lipocalin-2 (LCN2) and low-density lipoprotein receptor-related protein-1 (LRP1) with intestinal inflammation. Therefore, we intended to explore the relationship of LCN2 and LRP1 with gut inflammation and biomarkers of EED in Bangladeshi malnourished children. Methods A total of 222 children (length-for-age z-score (LAZ) <-1) aged 12-18 months were enrolled in this study in a cross-sectional manner. Among the participants, 115 were stunted (LAZ <-2) and 107 were at risk of being stunted (LAZ -1 to -2) children. Plasma and faecal biomarkers were measured using ELISA. Spearman's rank correlation was done to see the correlation among LCN2, LRP1 and biological biomarkers. Results LCN2 correlates positively with myeloperoxidase (r=0.19, p=0.005), neopterin (r=0.20, p=0.004), calprotectin (r=0.3, p=0.0001), Reg1B (r=0.20, p=0.003) and EED score (r=0.20, p=0.003). Whereas, LRP1 correlates negatively with myeloperoxidase (r = -0.18, p=0.006), neopterin (r = -0.30, p=0.0001), alpha-1-antitrypsin (r = -0.18, p=0.006), Reg1B (r=-0.2, p=0.003) and EED score (r = -0.29, p=0.0001). Conclusions Our findings imply that LCN2 might be a promising biomarker to predict gut inflammation and EED. Whereas, increased level of LRP1 may contribute to alleviating intestinal inflammation.
Collapse
Affiliation(s)
- Md. Mehedi Hasan
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Md. Amran Gazi
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Subhasish Das
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Shah Mohammad Fahim
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Farzana Hossaini
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Md. Ashraful Alam
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh (icddr, b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division (NCSD), International Centre for Diarrhoeal Disease Research Bangladesh (icddr, b), Dhaka, Bangladesh
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
12
|
Lactoferrin and Its Detection Methods: A Review. Nutrients 2021; 13:nu13082492. [PMID: 34444652 PMCID: PMC8398339 DOI: 10.3390/nu13082492] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Lactoferrin (LF) is one of the major functional proteins in maintaining human health due to its antioxidant, antibacterial, antiviral, and anti-inflammatory activities. Abnormal levels of LF in the human body are related to some serious diseases, such as inflammatory bowel disease, Alzheimer’s disease and dry eye disease. Recent studies indicate that LF can be used as a biomarker for diagnosis of these diseases. Many methods have been developed to detect the level of LF. In this review, the biofunctions of LF and its potential to work as a biomarker are introduced. In addition, the current methods of detecting lactoferrin have been presented and discussed. We hope that this review will inspire efforts in the development of new sensing systems for LF detection.
Collapse
|
13
|
Uddin MI, Hossain M, Islam S, Akter A, Nishat NS, Nila TA, Rafique TA, Leung DT, Calderwood SB, Ryan ET, Harris JB, LaRocque RC, Bhuiyan TR, Qadri F. An assessment of potential biomarkers of environment enteropathy and its association with age and microbial infections among children in Bangladesh. PLoS One 2021; 16:e0250446. [PMID: 33886672 PMCID: PMC8061931 DOI: 10.1371/journal.pone.0250446] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 04/06/2021] [Indexed: 01/13/2023] Open
Abstract
Interventional studies targeting environment enteropathy (EE) are impeded by the lack of appropriate, validated, non-invasive biomarkers of EE. Thus, we aimed to validate the association of potential biomarkers for EE with enteric infections and nutritional status in a longitudinal birth cohort study. We measured endotoxin core antibody (EndoCab) and soluble CD14 (sCD14) in serum, and myeloperoxidase (MPO) in feces using commercially available enzyme-linked immunosorbent assay (ELISA) kits. We found that levels of serum EndoCab and sCD14 increase with the cumulative incidence of enteric infections. We observed a significant correlation between the fecal MPO level in the children at 24 months of age with the total number of bacterial and viral infections, the total number of parasitic infections, and the total number of diarrheal episodes and diarrheal duration. We observed that the levels of serum EndoCab, sCD14, and fecal MPO at 3 months of age were significantly associated with whether children were malnourished at 18 months of age or not. Biomarkers such as fecal MPO, serum EndoCab and sCD14 in children at an early age may be useful as a measure of cumulative burden of preceding enteric infections, which are predictive of subsequent malnutrition status and may be useful non-invasive biomarkers for EE.
Collapse
Affiliation(s)
| | | | - Shahidul Islam
- Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Aklima Akter
- Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | | | | | | | - Daniel T. Leung
- Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
- Division of Infectious Diseases, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Stephen B. Calderwood
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Edward T. Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jason B. Harris
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Regina C. LaRocque
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Firdausi Qadri
- Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
- * E-mail: (FQ); (TRB)
| |
Collapse
|
14
|
Park H, Yeo S, Kang S, Huh CS. Longitudinal Microbiome Analysis in a Dextran Sulfate Sodium-Induced Colitis Mouse Model. Microorganisms 2021; 9:370. [PMID: 33673349 PMCID: PMC7917662 DOI: 10.3390/microorganisms9020370] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
The role of the gut microbiota in the pathogenesis of inflammatory bowel disease (IBD) has been in focus for decades. Although metagenomic observations in patients/animal colitis models have been attempted, the microbiome results were still indefinite and broad taxonomic presumptions were made due to the cross-sectional studies. Herein, we conducted a longitudinal microbiome analysis in a dextran sulfate sodium (DSS)-induced colitis mouse model with a two-factor design based on serial DSS dose (0, 1, 2, and 3%) and duration for 12 days, and four mice from each group were sacrificed at two-day intervals. During the colitis development, a transition of the cecal microbial diversity from the normal state to dysbiosis and dynamic changes of the populations were observed. We identified genera that significantly induced or depleted depending on DSS exposure, and confirmed the correlations of the individual taxa to the colitis severity indicated by inflammatory biomarkers (intestinal bleeding and neutrophil-derived indicators). Of note, each taxonomic population showed its own susceptibility to the changing colitis status. Our findings suggest that an understanding of the individual susceptibility to colitis conditions may contribute to identifying the role of the gut microbes in the pathogenesis of IBD.
Collapse
Affiliation(s)
- Hyunjoon Park
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon 25354, Korea;
- Advanced Green Energy and Environment Institute, Handong Global University, Pohang 37554, Korea
| | - Soyoung Yeo
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea;
| | - Seokwon Kang
- Department of Life Sciences, Handong Global University, Pohang 37554, Korea;
| | - Chul Sung Huh
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon 25354, Korea;
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Gangwon 25354, Korea
| |
Collapse
|
15
|
Brubaker J, Zhang X, Bourgeois AL, Harro C, Sack DA, Chakraborty S. Intestinal and systemic inflammation induced by symptomatic and asymptomatic enterotoxigenic E. coli infection and impact on intestinal colonization and ETEC specific immune responses in an experimental human challenge model. Gut Microbes 2021; 13:1-13. [PMID: 33645430 PMCID: PMC7919917 DOI: 10.1080/19490976.2021.1891852] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/02/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Recent studies have gained a better appreciation of the potential impacts of enteric infections beyond symptomatic diarrhea. It is recognized that infections by several enteropathogens could be associated with growth deficits in children and intestinal and systemic inflammation may play an important underlying role. With enterotoxigenic E. coli (ETEC) being one of the leading causes of diarrhea among children in the developing world and important contributor to stunting, a better understanding of the impact of ETEC infection beyond diarrhea is timely and greatly needed. To address this, we evaluated if ETEC infection induces intestinal and systemic inflammation and its impact on colonization and immune responses to ETEC vaccine-specific antigens in a dose descending experimental human challenge model using ETEC strain H10407. This study demonstrates that the concentrations of myeloperoxidase (MPO) in stool and intestinal fatty acid-binding protein (an indicator of compromised intestinal epithelial integrity) in serum, significantly increased following ETEC infection in both diarrhea and asymptomatic cases and the magnitudes and kinetics of MPO are dose and clinical outcome dependent. Cytokines IL-17A and IFN-γ were significantly increased in serum post-ETEC challenge. In addition, higher pre-challenge concentrations of cytokines IL-10 and GM-CSF were associated with protection from ETEC diarrhea. Interestingly, higher MPO concentrations were associated with higher intestinal colonization of ETEC and lower seroconversions of colonization factor I antigen, but the reverse was noted for seroconversions to heat-labile toxin B-subunit. Together this study has important implications for understanding the acute and long-term negative health outcomes associated with ETEC infection.
Collapse
Affiliation(s)
- Jessica Brubaker
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Xueyan Zhang
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - A. Louis Bourgeois
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- PATH, Center for Vaccine Innovation and Access, Washington, DC, 20001, USA
| | - Clayton Harro
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - David A Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
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: 12] [Impact Index Per Article: 2.4] [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
|
17
|
Kaminski RW, Pasetti MF, Aguilar AO, Clarkson KA, Rijpkema S, Bourgeois AL, Cohen D, Feavers I, MacLennan CA. Consensus Report on Shigella Controlled Human Infection Model: Immunological Assays. Clin Infect Dis 2020; 69:S596-S601. [PMID: 31816067 PMCID: PMC6901123 DOI: 10.1093/cid/ciz909] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Moderate to severe diarrhea caused by Shigella is a global health concern due to its substantial contribution to morbidity and mortality in children aged <5 years in low- and middle-income countries. Although antibiotic treatment can be effective, emerging antimicrobial resistance, limited access, and cost affirm the role of vaccines as the most attractive countermeasure. Controlled human infection models (CHIMs) represent a valuable tool for assessing vaccine efficacy and potentially accelerating licensure. Currently, immunological analysis during CHIM studies is customized based on vaccine type, regimen, and administration route. Additionally, differences in type of immunoassays and procedures used limit comparisons across studies. In November 2017, an expert working group reviewed Shigella CHIM studies performed to date and developed consensus guidelines on prioritization of immunoassays, specimens, and collection time points. Immunoassays were ranked into 3 tiers, with antibodies to Shigella lipopolysaccharide (LPS) being the highest priority. To facilitate comparisons across clinical studies, a second workshop was conducted in December 2017, which focused on the pathway toward a recognized enzyme-linked immunosorbent assay (ELISA) to determine serum immunoglobulin G titers against Shigella LPS. The consensus of the meeting was to establish a consortium of international institutions with expertise in Shigella immunology that would work with the National Institute for Biological Standards and Control to establish a harmonized ELISA, produce a reference sera, and identify a reliable source of Shigella LPS for global utilization. Herein we describe efforts toward establishing common procedures to advance Shigella vaccine development, support licensure, and ultimately facilitate vaccine deployment and uptake.
Collapse
Affiliation(s)
- Robert W Kaminski
- Subunit Enteric Vaccines and Immunology, Department of Enteric Infections, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring
| | - Marcela F Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore
| | | | - Kristen A Clarkson
- Subunit Enteric Vaccines and Immunology, Department of Enteric Infections, Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring
| | - Sjoerd Rijpkema
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | | | - Dani Cohen
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Ian Feavers
- Division of Bacteriology, National Institute for Biological Standards and Control, Potters Bar, United Kingdom
| | | |
Collapse
|
18
|
Ozkul C, Ruiz VE, Battaglia T, Xu J, Roubaud-Baudron C, Cadwell K, Perez-Perez GI, Blaser MJ. A single early-in-life antibiotic course increases susceptibility to DSS-induced colitis. Genome Med 2020; 12:65. [PMID: 32711559 PMCID: PMC7382806 DOI: 10.1186/s13073-020-00764-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Background There is increasing evidence that the intestinal microbiota plays a crucial role in the maturation of the immune system and the prevention of diseases during childhood. Early-life short-course antibiotic use may affect the progression of subsequent disease conditions by changing both host microbiota and immunologic development. Epidemiologic studies provide evidence that early-life antibiotic exposures predispose to inflammatory bowel disease (IBD). Methods By using a murine model of dextran sodium sulfate (DSS)-induced colitis, we evaluated the effect on disease outcomes of early-life pulsed antibiotic treatment (PAT) using tylosin, a macrolide and amoxicillin, a beta-lactam. We evaluated microbiota effects at the 16S rRNA gene level, and intestinal T cells by flow cytometry. Antibiotic-perturbed or control microbiota were transferred to pups that then were challenged with DSS. Results A single PAT course early-in-life exacerbated later DSS-induced colitis by both perturbing the microbial community and altering mucosal immune cell composition. By conventionalizing germ-free mice with either antibiotic-perturbed or control microbiota obtained 40 days after the challenge ended, we showed the transferrable and direct effect of the still-perturbed microbiota on colitis severity in the DSS model. Conclusions The findings in this experimental model provide evidence that early-life microbiota perturbation may increase risk of colitis later in life.
Collapse
Affiliation(s)
- Ceren Ozkul
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Hacettepe University, Sihhiye, Ankara, Turkey.,Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Victoria E Ruiz
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA.,Department of Biology, St. Francis College, Brooklyn, New York, USA
| | - Thomas Battaglia
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Joseph Xu
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Claire Roubaud-Baudron
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA.,CHU Bordeaux, Pôle de Gérontologie Clinique, Bordeaux, France.,INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, F-33000, Bordeaux, France
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY, 10016, USA.,Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA.,Division of Gastroenterology and Hepatology, Department of Medicine, New York University Langone Health, New York, NY, 10016, USA
| | - Guillermo I Perez-Perez
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA
| | - Martin J Blaser
- Departments of Medicine and Microbiology, New York University School of Medicine (NYUSM), New York, NY, 10016, USA. .,Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
19
|
Yeo S, Park H, Seo E, Kim J, Kim BK, Choi IS, Huh CS. Anti-Inflammatory and Gut Microbiota Modulatory Effect of Lactobacillus rhamnosus Strain LDTM 7511 in a Dextran Sulfate Sodium-Induced Colitis Murine Model. Microorganisms 2020; 8:E845. [PMID: 32512895 PMCID: PMC7356973 DOI: 10.3390/microorganisms8060845] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a group of conditions involving chronic relapsing-remitting inflammation of the gastrointestinal tract with an unknown etiology. Although the cause-effect relationship between gut microbiota and IBD has not been clearly established, emerging evidence from experimental models supports the idea that gut microbes play a fundamental role in the pathogenesis of IBD. As microbiome-based therapeutics for IBD, the beneficial effects of probiotics have been found in animal colitis models and IBD patients. In this study, based on the dextran sulfate sodium (DSS)-induced colitis mouse model, we investigated Lactobacillus rhamnosus strain LDTM 7511 originating from Korean infant feces as a putative probiotic strain for IBD. The strain LDTM 7511 not only alleviated the release of inflammatory mediators, but also induced the transition of gut microbiota from dysbiotic conditions, exhibiting the opposite pattern in the abundance of DSS colitis-associated bacterial taxa to the DSS group. Our findings suggest that the strain LDTM 7511 has the potential to be used as a probiotic treatment for IBD patients in comparison to L. rhamnosus GG (ATCC 53103), which has been frequently used for IBD studies.
Collapse
Affiliation(s)
- Soyoung Yeo
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (S.Y.); (E.S.); (J.K.)
| | - Hyunjoon Park
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea;
| | - Eunsol Seo
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (S.Y.); (E.S.); (J.K.)
| | - Jihee Kim
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea; (S.Y.); (E.S.); (J.K.)
- Chong Kun Dang Bio Research Institute, Chong Kun Dang Bio Research Institute, Ansan 15604, Korea; (B.K.K.); (I.S.C.)
| | - Byoung Kook Kim
- Chong Kun Dang Bio Research Institute, Chong Kun Dang Bio Research Institute, Ansan 15604, Korea; (B.K.K.); (I.S.C.)
| | - In Suk Choi
- Chong Kun Dang Bio Research Institute, Chong Kun Dang Bio Research Institute, Ansan 15604, Korea; (B.K.K.); (I.S.C.)
| | - Chul Sung Huh
- Research Institute of Eco-Friendly Livestock Science, Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea;
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang 25354, Korea
| |
Collapse
|
20
|
Glucagon-like Peptide 2 Concentrations Vary in Zambian Children During Diarrhoea, in Malnutrition and Seasonally. J Pediatr Gastroenterol Nutr 2020; 70:513-520. [PMID: 32044830 PMCID: PMC7340486 DOI: 10.1097/mpg.0000000000002633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Glucagon-like peptide 2 (GLP-2) is a 33 amino acid peptide hormone released from enteroendocrine L-cells following nutrient ingestion. It has been shown to exert trophic effects on the gut. We set out to measure GLP-2 concentrations in blood in children with diarrhoea and malnutrition. METHODS GLP-2 levels were measured in blood samples collected from 5 different groups of children (n = 324) at different time points: those with acute diarrhoea, during illness and 3 weeks after recovery; persistent diarrhoea and severe acute malnutrition; controls contemporaneous for diarrhoea; stunted children from the community; and controls contemporaneous for the stunted children. Stool biomarkers and pathogen analysis were carried out on the children with stunting. RESULTS GLP-2 concentrations were higher during acute diarrhoea (median 3.1 ng/mL, interquartile range 2.1, 4.4) than on recovery (median 1.8, interquartile range 1.4, 3.1; P = 0.001), but were not elevated in children with persistent diarrhoea and severe acute malnutrition. In stunted children, there was a progressive decline in GLP-2 levels from 3.2 ng/mL (1.9, 4.9) to 1.0 (0.0, 2.0; P < 0.001) as the children became more stunted. Measures of seasonality (rainfall, temperature,Food Price Index, and Shiga toxin-producing Escherichia coli) were found to be significantly associated with GLP-2 concentrations in multivariable analysis. We also found a correlation between stool inflammatory biomarkers and GLP-2. CONCLUSIONS In diarrhoea, GLP-2 levels increased in acute but not persistent diarrhoea. Malnutrition was associated with reduced concentrations. GLP-2 displayed seasonal variation consistent with variations in nutrient availability.
Collapse
|
21
|
Q.S. Medeiros PH, Ledwaba SE, Bolick DT, Giallourou N, Yum LK, Costa DV, Oriá RB, Barry EM, Swann JR, Lima AÂM, Agaisse H, Guerrant RL. A murine model of diarrhea, growth impairment and metabolic disturbances with Shigella flexneri infection and the role of zinc deficiency. Gut Microbes 2019; 10:615-630. [PMID: 30712505 PMCID: PMC6748602 DOI: 10.1080/19490976.2018.1564430] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Shigella is one of the major enteric pathogens worldwide. We present a murine model of S. flexneri infection and investigate the role of zinc deficiency (ZD). C57BL/6 mice fed either standard chow (HC) or ZD diets were pretreated with an antibiotic cocktail and received S. flexneri strain 2457T orally. Antibiotic pre-treated ZD mice showed higher S. flexneri colonization than non-treated mice. ZD mice showed persistent colonization for at least 50 days post-infection (pi). S. flexneri-infected mice showed significant weight loss, diarrhea and increased levels of fecal MPO and LCN in both HC and ZD fed mice. S. flexneri preferentially colonized the colon, caused epithelial disruption and inflammatory cell infiltrate, and promoted cytokine production which correlated with weight loss and histopathological changes. Infection with S. flexneri ΔmxiG (critical for type 3 secretion system) did not cause weight loss or diarrhea, and had decreased stool shedding duration and tissue burden. Several biochemical changes related to energy, inflammation and gut-microbial metabolism were observed. Zinc supplementation increased weight gains and reduced intestinal inflammation and stool shedding in ZD infected mice. In conclusion, young antibiotic-treated mice provide a new model of oral S. flexneri infection, with ZD promoting prolonged infection outcomes.
Collapse
Affiliation(s)
- Pedro Henrique Q.S. Medeiros
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil,CONTACT Pedro Henrique Q.S. Medeiros Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, 345 Crispell Drive, MR6 Room 2711, Charlottesville, VA, USA
| | - Solanka E. Ledwaba
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | - David T. Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | - Natasa Giallourou
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Lauren K. Yum
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Deiziane V.S. Costa
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Reinaldo B. Oriá
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Eileen M. Barry
- Center for Vaccine Development, University of Maryland, Baltimore, USA
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Hervé Agaisse
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Richard L. Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| |
Collapse
|
22
|
Bartelt LA, Bolick DT, Guerrant RL. Disentangling Microbial Mediators of Malnutrition: Modeling Environmental Enteric Dysfunction. Cell Mol Gastroenterol Hepatol 2019; 7:692-707. [PMID: 30630118 PMCID: PMC6477186 DOI: 10.1016/j.jcmgh.2018.12.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022]
Abstract
Environmental enteric dysfunction (EED) (also referred to as environmental enteropathy) is a subclinical chronic intestinal disorder that is an emerging contributor to early childhood malnutrition. EED is common in resource-limited settings, and is postulated to consist of small intestinal injury, dysfunctional nutrient absorption, and chronic inflammation that results in impaired early child growth attainment. Although there is emerging interest in the hypothetical potential for chemical toxins in the environmental exposome to contribute to EED, the propensity of published data, and hence the focus of this review, implicates a critical role of environmental microbes. Early childhood malnutrition and EED are most prevalent in resource-limited settings where food is limited, and inadequate access to clean water and sanitation results in frequent gastrointestinal pathogen exposures. Even as overt diarrhea rates in these settings decline, silent enteric infections and faltering growth persist. Furthermore, beyond restricted physical growth, EED and/or enteric pathogens also associate with impaired oral vaccine responses, impaired cognitive development, and may even accelerate metabolic syndrome and its cardiovascular consequences. As these potentially costly long-term consequences of early childhood enteric infections increasingly are appreciated, novel therapeutic strategies that reverse damage resulting from nutritional deficiencies and microbial insults in the developing small intestine are needed. Given the inherent limitations in investigating how specific intestinal pathogens directly injure the small intestine in children, animal models provide an affordable and controlled opportunity to elucidate causal sequelae of specific enteric infections, to differentiate consequences of defined nutrient deprivation alone from co-incident enteropathogen insults, and to correlate the resulting gut pathologies with their functional impact during vulnerable early life windows.
Collapse
Affiliation(s)
- Luther A Bartelt
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - David T Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Richard L Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
23
|
Church JA, Parker EPK, Kosek MN, Kang G, Grassly NC, Kelly P, Prendergast AJ. Exploring the relationship between environmental enteric dysfunction and oral vaccine responses. Future Microbiol 2018; 13:1055-1070. [PMID: 29926747 PMCID: PMC6136084 DOI: 10.2217/fmb-2018-0016] [Citation(s) in RCA: 38] [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: 01/12/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022] Open
Abstract
Oral vaccines significantly underperform in low-income countries. One possible contributory factor is environmental enteric dysfunction (EED), a subclinical disorder of small intestinal structure and function among children living in poverty. Here, we review studies describing oral vaccine responses and EED. We identified eight studies evaluating EED and oral vaccine responses. There was substantial heterogeneity in study design and few consistent trends emerged. Four studies reported a negative association between EED and oral vaccine responses; two showed no significant association; and two described a positive correlation. Current evidence is therefore insufficient to determine whether EED contributes to oral vaccine underperformance. We identify roadblocks in the field and future research needs, including carefully designed studies those can investigate this hypothesis further.
Collapse
Affiliation(s)
- James A Church
- Zvitambo Institute for Maternal & Child Health Research, Harare, Zimbabwe
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK
| | - Edward PK Parker
- Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, UK
| | - Margaret N Kosek
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Gagandeep Kang
- Department of Gastrointestinal Sciences, Christian Medical College, Vellore, Tamil Nadu, India
| | - Nicholas C Grassly
- Department of Infectious Disease Epidemiology, St Mary's Campus, Imperial College London, London, UK
| | - Paul Kelly
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal & Child Health Research, Harare, Zimbabwe
- Centre for Genomics & Child Health, Blizard Institute, Queen Mary University of London, UK
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
24
|
Critical Role of Zinc in a New Murine Model of Enterotoxigenic Escherichia coli Diarrhea. Infect Immun 2018; 86:IAI.00183-18. [PMID: 29661930 PMCID: PMC6013668 DOI: 10.1128/iai.00183-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of traveler's diarrhea as well as of endemic diarrhea and stunting in children in developing areas. However, a small-mammal model has been badly needed to better understand and assess mechanisms, vaccines, and interventions. We report a murine model of ETEC diarrhea, weight loss, and enteropathy and investigate the role of zinc in the outcomes. ETEC strains producing heat-labile toxins (LT) and heat-stable toxins (ST) that were given to weaned C57BL/6 mice after antibiotic disruption of normal microbiota caused growth impairment, watery diarrhea, heavy stool shedding, and mild to moderate intestinal inflammation, the latter being worse with zinc deficiency. Zinc treatment promoted growth in zinc-deficient infected mice, and subinhibitory levels of zinc reduced expression of ETEC virulence genes cfa1, cexE, sta2, and degP but not of eltA in vitro Zinc supplementation increased shedding and the ileal burden of wild-type (WT) ETEC but decreased shedding and the tissue burden of LT knockout (LTKO) ETEC. LTKO ETEC-infected mice had delayed disease onset and also had less inflammation by fecal myeloperoxidase (MPO) assessment. These findings provide a new murine model of ETEC infection that can help elucidate mechanisms of growth, diarrhea, and inflammatory responses as well as potential vaccines and interventions.
Collapse
|
25
|
Giallourou N, Medlock GL, Bolick DT, Medeiros PHQS, Ledwaba SE, Kolling GL, Tung K, Guerry P, Swann JR, Guerrant RL. A novel mouse model of Campylobacter jejuni enteropathy and diarrhea. PLoS Pathog 2018; 14:e1007083. [PMID: 29791507 PMCID: PMC5988333 DOI: 10.1371/journal.ppat.1007083] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/05/2018] [Accepted: 05/09/2018] [Indexed: 01/31/2023] Open
Abstract
Campylobacter infections are among the leading bacterial causes of diarrhea and of 'environmental enteropathy' (EE) and growth failure worldwide. However, the lack of an inexpensive small animal model of enteric disease with Campylobacter has been a major limitation for understanding its pathogenesis, interventions or vaccine development. We describe a robust standard mouse model that can exhibit reproducible bloody diarrhea or growth failure, depending on the zinc or protein deficient diet and on antibiotic alteration of normal microbiota prior to infection. Zinc deficiency and the use of antibiotics create a niche for Campylobacter infection to establish by narrowing the metabolic flexibility of these mice for pathogen clearance and by promoting intestinal and systemic inflammation. Several biomarkers and intestinal pathology in this model also mimic those seen in human disease. This model provides a novel tool to test specific hypotheses regarding disease pathogenesis as well as vaccine development that is currently in progress.
Collapse
Affiliation(s)
- Natasa Giallourou
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Gregory L. Medlock
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - David T. Bolick
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Pedro HQS Medeiros
- Institute of Biomedicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Solanka E. Ledwaba
- Department of Microbiology, University of Venda, Thohoyandou, Limpopo, South Africa
| | - Glynis L. Kolling
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Kenneth Tung
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Patricia Guerry
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Richard L. Guerrant
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| |
Collapse
|
26
|
Denno DM, Tarr PI, Nataro JP. Environmental Enteric Dysfunction: A Case Definition for Intervention Trials. Am J Trop Med Hyg 2017; 97:1643-1646. [PMID: 29016294 PMCID: PMC5805039 DOI: 10.4269/ajtmh.17-0183] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Donna M Denno
- Department of Pediatrics, University of Washington, Seattle, Washington.,Department of Global Health, University of Washington, Seattle, Washington
| | - Phillip I Tarr
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - James P Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
27
|
Abstract
Environmental enteropathy is a chronic condition of the small intestine associated with increased intestinal permeability, mucosal inflammation, malabsorption, and systemic inflammation. It is commonly accompanied by enteric infections and is misleadingly considered a subclinical disease. Potential effects of enteric infections and enteropathy on vaccine responses, child growth, cognitive development, and even later life obesity, diabetes, and metabolic syndrome are increasingly being recognized. Herein, we review the evolving challenges to defining environmental enteropathy and enteric infections, current evidence for the magnitude and determinants of its burden, new assessment tools, and relevant interventions.
Collapse
Affiliation(s)
- Elizabeth T Rogawski
- Department of Public Health Sciences, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA; Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA.
| | - Richard L Guerrant
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, PO Box 801379, Carter Harrison Research Building MR-6, 345 Crispell Drive, Room 2520, Charlottesville, VA 22908-1379, USA
| |
Collapse
|
28
|
Bartelt LA, Bolick DT, Mayneris-Perxachs J, Kolling GL, Medlock GL, Zaenker EI, Donowitz J, Thomas-Beckett RV, Rogala A, Carroll IM, Singer SM, Papin J, Swann JR, Guerrant RL. Cross-modulation of pathogen-specific pathways enhances malnutrition during enteric co-infection with Giardia lamblia and enteroaggregative Escherichia coli. PLoS Pathog 2017; 13:e1006471. [PMID: 28750066 PMCID: PMC5549954 DOI: 10.1371/journal.ppat.1006471] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/14/2017] [Indexed: 12/17/2022] Open
Abstract
Diverse enteropathogen exposures associate with childhood malnutrition. To
elucidate mechanistic pathways whereby enteric microbes interact during
malnutrition, we used protein deficiency in mice to develop a new model of
co-enteropathogen enteropathy. Focusing on common enteropathogens in
malnourished children, Giardia lamblia and enteroaggregative
Escherichia coli (EAEC), we provide new insights into
intersecting pathogen-specific mechanisms that enhance malnutrition. We show for
the first time that during protein malnutrition, the intestinal microbiota
permits persistent Giardia colonization and simultaneously
contributes to growth impairment. Despite signals of intestinal injury, such as
IL1α, Giardia-infected mice lack pro-inflammatory intestinal
responses, similar to endemic pediatric Giardia infections.
Rather, Giardia perturbs microbial host co-metabolites of
proteolysis during growth impairment, whereas host nicotinamide utilization
adaptations that correspond with growth recovery increase. EAEC promotes
intestinal inflammation and markers of myeloid cell activation. During
co-infection, intestinal inflammatory signaling and cellular recruitment
responses to EAEC are preserved together with a
Giardia-mediated diminishment in myeloid cell activation.
Conversely, EAEC extinguishes markers of host energy expenditure regulatory
responses to Giardia, as host metabolic adaptations appear
exhausted. Integrating immunologic and metabolic profiles during co-pathogen
infection and malnutrition, we develop a working mechanistic model of how
cumulative diet-induced and pathogen-triggered microbial perturbations result in
an increasingly wasted host. Malnourished children are exposed to multiple sequential, and oftentimes,
persistent enteropathogens. Intestinal microbial disruption and inflammation are
known to contribute to the pathogenesis of malnutrition, but how co-pathogens
interact with each other, with the resident microbiota, or with the host to
alter these pathways is unknown. Using a new model of enteric co-infection with
Giardia lamblia and enteroaggregative Escherichia
coli in mice fed a protein deficient diet, we identify host growth
and intestinal immune responses that are differentially mediated by
pathogen-microbe interactions, including parasite-mediated changes in intestinal
microbial host co-metabolism, and altered immune responses during co-infection.
Our data model how early life cumulative enteropathogen exposures progressively
disrupt intestinal immunity and host metabolism during crucial developmental
periods. Furthermore, studies in this co-infection model reveal new insights
into environmental and microbial determinants of pathogenicity for presently
common, but poorly understood enteropathogens like Giardia
lamblia, that may not conform to existing paradigms of microbial
pathogenesis based on single pathogen-designed models.
Collapse
Affiliation(s)
- Luther A. Bartelt
- Division of Infectious Diseases, Department of Medicine, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of
America
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
- * E-mail:
| | - David T. Bolick
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Jordi Mayneris-Perxachs
- Division of Computational and Systems Medicine, Department of Surgery and
Cancer, Imperial College London, United Kingdom
| | - Glynis L. Kolling
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Gregory L. Medlock
- Department of Biomedical Engineering, University of Virginia,
Charlottesville, Virginia, United States of America
| | - Edna I. Zaenker
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| | - Jeffery Donowitz
- Division of Pediatric Infectious Diseases, Children’s Hospital of
Richmond at Virginia Commonwealth University, Richmond, Virginia, United States
of America
| | - Rose Viguna Thomas-Beckett
- Division of Infectious Diseases, Department of Medicine, University of
North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of
America
| | - Allison Rogala
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
| | - Ian M. Carroll
- Center for Gastrointestinal Biology and Disease, Department of Medicine,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United
States of America
| | - Steven M. Singer
- Department of Biology, Georgetown University, Washington, DC, United
States of America
| | - Jason Papin
- Department of Biomedical Engineering, University of Virginia,
Charlottesville, Virginia, United States of America
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and
Cancer, Imperial College London, United Kingdom
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, Department of
Medicine, University of Virginia, Charlottesville, Virginia, United States of
America
| |
Collapse
|
29
|
McCormick BJJ, Lee GO, Seidman JC, Haque R, Mondal D, Quetz J, Lima AAM, Babji S, Kang G, Shrestha SK, Mason CJ, Qureshi S, Bhutta ZA, Olortegui MP, Yori PP, Samie A, Bessong P, Amour C, Mduma E, Patil CL, Guerrant RL, Lang DR, Gottlieb M, Caulfield LE, Kosek MN. Dynamics and Trends in Fecal Biomarkers of Gut Function in Children from 1-24 Months in the MAL-ED Study. Am J Trop Med Hyg 2016; 96:465-472. [PMID: 27994110 PMCID: PMC5303054 DOI: 10.4269/ajtmh.16-0496] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/06/2016] [Indexed: 12/28/2022] Open
Abstract
Growth and development shortfalls that are disproportionately prevalent in children living in poor environmental conditions are postulated to result, at least in part, from abnormal gut function. Using data from The Etiology, Risk Factors, and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development (MAL-ED) longitudinal cohort study, we examine biomarkers of gut inflammation and permeability in relation to environmental exposures and feeding practices. Trends in the concentrations of three biomarkers, myeloperoxidase (MPO), neopterin (NEO), and α-1-antitrypsin (AAT), are described from fecal samples collected during the first 2 years of each child's life. A total of 22,846 stool samples were processed during the longitudinal sampling of 2,076 children 0–24 months of age. Linear mixed models were constructed to examine the relationship between biomarker concentrations and recent food intake, symptoms of illness, concurrent enteropathogen infection, and socioeconomic status. Average concentrations of MPO, NEO, and AAT were considerably higher than published references for healthy adults. The concentration of each biomarker tended to decrease over the first 2 years of life and was highly variable between samples from each individual child. Both MPO and AAT were significantly elevated by recent breast milk intake. All three biomarkers were associated with pathogen presence, although the strength and direction varied by pathogen. The interpretation of biomarker concentrations is subject to the context of their collection. Herein, we identify that common factors (age, breast milk, and enteric infection) influence the concentration of these biomarkers. Within the context of low- and middle-income communities, we observe concentrations that indicate gut abnormalities, but more appropriate reference standards are needed.
Collapse
Affiliation(s)
| | - Gwenyth O Lee
- Tulane University, New Orleans, Louisiana.,Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Jessica C Seidman
- Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Dinesh Mondal
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | | | | | | | | | - Sanjaya K Shrestha
- Walter Reed/Armed Forces Research Institute of Medical Science (AFRIMS) Research Unit, Kathmandu, Nepal
| | - Carl J Mason
- Walter Reed/Armed Forces Research Institute of Medical Science (AFRIMS) Research Unit, Kathmandu, Nepal
| | | | | | - Maribel Paredes Olortegui
- Asociacion Benéfica Proyectos en Informatica, Salud, Medicina, y Agricultura (A. B. PRISMA), Iquitos, Peru
| | - Pablo Peñataro Yori
- Asociacion Benéfica Proyectos en Informatica, Salud, Medicina, y Agricultura (A. B. PRISMA), Iquitos, Peru
| | | | | | | | | | | | | | - Dennis R Lang
- Foundation for the National Institutes of Health, Bethesda, Maryland.,Fogarty International Center/National Institutes of Health, Bethesda, Maryland
| | - Michael Gottlieb
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | - Laura E Caulfield
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Margaret N Kosek
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
30
|
Guerrant RL, Leite AM, Pinkerton R, Medeiros PHQS, Cavalcante PA, DeBoer M, Kosek M, Duggan C, Gewirtz A, Kagan JC, Gauthier AE, Swann J, Mayneris-Perxachs J, Bolick DT, Maier EA, Guedes MM, Moore SR, Petri WA, Havt A, Lima IF, Prata MDMG, Michaleckyj JC, Scharf RJ, Sturgeon C, Fasano A, Lima AAM. Biomarkers of Environmental Enteropathy, Inflammation, Stunting, and Impaired Growth in Children in Northeast Brazil. PLoS One 2016; 11:e0158772. [PMID: 27690129 PMCID: PMC5045163 DOI: 10.1371/journal.pone.0158772] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/21/2016] [Indexed: 01/27/2023] Open
Abstract
Critical to the design and assessment of interventions for enteropathy and its developmental consequences in children living in impoverished conditions are non-invasive biomarkers that can detect intestinal damage and predict its effects on growth and development. We therefore assessed fecal, urinary and systemic biomarkers of enteropathy and growth predictors in 375 6–26 month-old children with varying degrees of malnutrition (stunting or wasting) in Northeast Brazil. 301 of these children returned for followup anthropometry after 2-6m. Biomarkers that correlated with stunting included plasma IgA anti-LPS and anti-FliC, zonulin (if >12m old), and intestinal FABP (I-FABP, suggesting prior barrier disruption); and with citrulline, tryptophan and with lower serum amyloid A (SAA) (suggesting impaired defenses). In contrast, subsequent growth was predicted in those with higher fecal MPO or A1AT and also by higher L/M, plasma LPS, I-FABP and SAA (showing intestinal barrier disruption and inflammation). Better growth was predicted in girls with higher plasma citrulline and in boys with higher plasma tryptophan. Interactions were also seen with fecal MPO and neopterin in predicting subsequent growth impairment. Biomarkers clustered into markers of 1) functional intestinal barrier disruption and translocation, 2) structural intestinal barrier disruption and inflammation and 3) systemic inflammation. Principle components pathway analyses also showed that L/M with %L, I-FABP and MPO associate with impaired growth, while also (like MPO) associating with a systemic inflammation cluster of kynurenine, LBP, sCD14, SAA and K/T. Systemic evidence of LPS translocation associated with stunting, while markers of barrier disruption or repair (A1AT and Reg1 with low zonulin) associated with fecal MPO and neopterin. We conclude that key noninvasive biomarkers of intestinal barrier disruption, LPS translocation and of intestinal and systemic inflammation can help elucidate how we recognize, understand, and assess effective interventions for enteropathy and its growth and developmental consequences in children in impoverished settings.
Collapse
Affiliation(s)
- Richard L. Guerrant
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
- * E-mail:
| | - Alvaro M. Leite
- Clinical Research Unit, Federal University of Ceara, Fortaleza, Brazil
| | - Relana Pinkerton
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | | | | | - Mark DeBoer
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Margaret Kosek
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Christopher Duggan
- Division of Gastroenterology at Boston Children’s Hospital, Harvard University, Boston, MA, United States of America
| | - Andrew Gewirtz
- Institute for Biomedical Sciences in the Center for Inflammation, Immunity and Infection at Georgia State University, Atlanta, GA, United States of America
| | - Jonathan C. Kagan
- Division of Gastroenterology at Boston Children’s Hospital, Harvard University, Boston, MA, United States of America
| | - Anna E. Gauthier
- Division of Gastroenterology at Boston Children’s Hospital, Harvard University, Boston, MA, United States of America
| | | | | | - David T. Bolick
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Elizabeth A. Maier
- Cincinnati Children’s Hospital, Cincinnati, OH, United States of America
| | - Marjorie M. Guedes
- Cincinnati Children’s Hospital, Cincinnati, OH, United States of America
| | - Sean R. Moore
- Cincinnati Children’s Hospital, Cincinnati, OH, United States of America
| | - William A. Petri
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Alexandre Havt
- Clinical Research Unit, Federal University of Ceara, Fortaleza, Brazil
| | - Ila F. Lima
- Clinical Research Unit, Federal University of Ceara, Fortaleza, Brazil
| | | | - Josyf C. Michaleckyj
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Rebecca J. Scharf
- University of Virginia School of Medicine (Division of Infectious Diseases and International Health, Department of Medicine, Department of Pediatrics and Center for Global Health), Charlottesville, VA, United States of America
| | - Craig Sturgeon
- Mucosal Immunology and Biology Research Center and Division of Pediatric Gastroenterology and Nutrition at Massachusetts General Hospital for Children, Harvard University, Boston, MA, United States of America
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center and Division of Pediatric Gastroenterology and Nutrition at Massachusetts General Hospital for Children, Harvard University, Boston, MA, United States of America
| | - Aldo A. M. Lima
- Clinical Research Unit, Federal University of Ceara, Fortaleza, Brazil
| |
Collapse
|