1
|
Shivakumar N, Morrison DJ, Hegde SG, Kurpad AV, Kelly P. Is there dietary macronutrient malabsorption in children with environmental enteropathy? Eur J Clin Nutr 2025; 79:181-194. [PMID: 39379550 PMCID: PMC11893463 DOI: 10.1038/s41430-024-01510-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024]
Abstract
Assessing the digestive and absorptive capacity of the gastro-intestinal tract (GIT) using minimally- or non-invasive methods, particularly in children, has been difficult owing to the complex physiology and variability in functional measurements. However, measuring GIT function is increasingly important with the emerging relevance of childhood environmental enteropathy (EE) as a mediating factor in linear growth faltering, severe acute malnutrition, poor oral vaccine uptake and impaired cognition. In EE, sub-optimal nutrient digestion and absorption (malabsorption) forms the critical link to the conditions mentioned above. The present narrative review discusses probable mechanisms that can cause malabsorption of macronutrients, along with mechanistic and experimental evidence, in children (if not, in adults) with EE. The strengths and limitations of the human experimental studies are examined in relation to a battery of existing and potential tests that are used to measure malabsorption. From the available studies conducted in children, lactose and fat malabsorption are more likely to occur in EE. Breath tests (non-invasive) measuring carbohydrate (13C-starch/sucrose/lactose), fat (13C-mixed triglyceride) and dipeptide (benzoyl-L-tyrosyl-L-1-13C-alanine) malabsorption with modifications to the existing protocols seem suitable for use in children with EE. Future research should focus on understanding the degree of macronutrient malabsorption using these tests, in different settings, and link them to functional outcomes (such as growth, muscle strength, cognition).
Collapse
Affiliation(s)
- Nirupama Shivakumar
- Division of Nutrition, St. John's Research Institute, St. John's National Academy of Health Sciences (A Unit of CBCI Society for Medical Education), Bangalore, India
- Center for Doctoral Studies, Manipal Academy of Higher Education, Manipal, India
| | - Douglas J Morrison
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Glasgow, UK
| | - Shalini G Hegde
- Department of Pediatric Surgery, St. John's Medical College Hospital, St. John's National Academy of Health Sciences, Bangalore, India
| | - Anura V Kurpad
- Department of Physiology, St. John's Medical College, St. John's National Academy of Health Sciences, Bangalore, India
| | - Paul Kelly
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
- Tropical Gastroenterology and Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia.
| |
Collapse
|
2
|
Chen D, McKune SL, Yang Y, Usmane IA, Ahmed IA, Amin JK, Ibrahim AM, Seran AJ, Shaik N, Ojeda A, Hassen BM, Deblais L, Ahmedo BU, Hassen KA, Bhrane M, Li X, Singh N, Roba KT, French NP, Rajashekara G, Manary MJ, Hassen JY, Havelaar AH. Campylobacter colonization and undernutrition in infants in rural eastern Ethiopia - a longitudinal community-based birth cohort study. Front Public Health 2025; 12:1467462. [PMID: 39839388 PMCID: PMC11747651 DOI: 10.3389/fpubh.2024.1467462] [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: 07/19/2024] [Accepted: 12/05/2024] [Indexed: 01/23/2025] Open
Abstract
Background Campylobacter is associated with environmental enteric dysfunction (EED) and malnutrition in children. Campylobacter infection could be a linchpin between livestock fecal exposure and health outcomes in low-resource smallholder settings. Methods We followed a birth cohort of 106 infants in rural smallholder households in eastern Ethiopia up to 13 months of age. We measured anthropometry, surveyed sociodemographic determinants, and collected stool and urine samples. A short survey was conducted during monthly visits, infant stool samples were collected, and Campylobacter spp. was quantified using genus-specific qPCR. In month 13, we collected stool and urine samples to assay for EED biomarkers. We employed regression analyses to assess the associations of household determinants with Campylobacter colonization, EED, and growth faltering. Results The Campylobacter load in infant stools increased with age. The mean length-for-age Z-score (LAZ) decreased from -0.45 at 3-4 months of age to -2.06 at 13 months, while the prevalence of stunting increased from 3 to 51%. The prevalence of EED at 13 months of age was 56%. A higher Campylobacter load was associated with more frequent diarrhea. Prelacteal feeding significantly increased Campylobacter load in the first month of life. Over the whole follow-up period, Campylobacter load was increased by keeping chickens unconfined at home and unsanitary disposal of infant stools while decreased by mothers' handwashing with soap. Longitudinally, Campylobacter load was positively associated with food insecurity, introduction of complementary foods, and raw milk consumption. There were no significant associations between Campylobacter load, EED, and LAZ. Conclusion This study found that most determinants associated with increased Campylobacter infection were related to suboptimal feeding practices and hygiene. The findings related to livestock-associated risks were inconclusive. Although stunting, EED, and Campylobacter prevalence rates all increased to high levels by the end of the first year of life, no significant association between them was identified. While additional research is needed to investigate whether findings from this study are replicable in other populations, community efforts to improve infant and young child feeding practices and food hygiene, and water, sanitation, and hygiene (WaSH) at the household level, could reduce (cross-)contamination at the point of exposure.
Collapse
Affiliation(s)
- Dehao Chen
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, United States
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Sarah Lindley McKune
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, United States
- Center for African Studies, University of Florida, Gainesville, FL, United States
| | - Yang Yang
- Department of Statistics, University of Georgia, Athens, GA, United States
| | - Ibsa Aliyi Usmane
- College of Agriculture and Environmental Science, Haramaya University, Haramaya, Ethiopia
| | | | - Jafer Kedir Amin
- College of Veterinary Medicine, Haramaya University, Haramaya, Ethiopia
| | | | | | - Nurmohammad Shaik
- Department of Pediatrics, Washington University, St. Louis, MO, United States
| | - Amanda Ojeda
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | | | - Loic Deblais
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Belisa Usmael Ahmedo
- College of Agriculture and Environmental Science, Haramaya University, Haramaya, Ethiopia
| | - Kedir Abdi Hassen
- College of Agriculture and Environmental Science, Haramaya University, Haramaya, Ethiopia
| | - Mussie Bhrane
- College of Agriculture and Environmental Science, Haramaya University, Haramaya, Ethiopia
| | - Xiaolong Li
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, United States
| | - Nitya Singh
- Department of Animal Sciences, Global Food Systems Institute, and Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Kedir Teji Roba
- College of Health and Medical Sciences, Haramaya University, Haramaya, Ethiopia
| | - Nigel P. French
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Gireesh Rajashekara
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Wooster, OH, United States
| | - Mark J. Manary
- Department of Pediatrics, Washington University, St. Louis, MO, United States
| | | | - Arie Hendrik Havelaar
- Department of Animal Sciences, Global Food Systems Institute, and Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | | |
Collapse
|
3
|
Van Wyk H, Lee GO, Schillinger RJ, Edwards CA, Morrison DJ, Brouwer AF. Performance of empirical and model-based classifiers for detecting sucrase-isomaltase inhibition using the 13C-sucrose breath test. J Breath Res 2024; 18:041003. [PMID: 39197471 PMCID: PMC11385691 DOI: 10.1088/1752-7163/ad748d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/01/2024]
Abstract
The13C-sucrose breath test (13C-SBT) has been proposed to estimate sucrase-isomaltase (SIM) activity and is a promising test for SIM deficiency, which can cause gastrointestinal symptoms, and for intestinal mucosal damage caused by gut dysfunction or chemotherapy. We previously showed how various summary measures of the13C-SBT breath curve reflect SIM inhibition. However, it is uncertain how the performance of these classifiers is affected by test duration. We leveraged13C-SBT data from a cross-over study in 16 adults who received 0, 100, and 750 mg of Reducose, an SIM inhibitor. We evaluated the performance of a pharmacokinetic-model-based classifier,ρ, and three empirical classifiers (cumulative percent dose recovered at 90 min (cPDR90), time to 50% dose recovered, and time to peak dose recovery rate), as a function of test duration using receiver operating characteristic (ROC) curves. We also assessed the sensitivity, specificity, and accuracy of consensus classifiers. Test durations of less than 2 h generally failed to accurately predict later breath curve dynamics. The cPDR90 classifier had the highest ROC area-under-the-curve and, by design, was robust to shorter test durations. For detecting mild SIM inhibition,ρhad a higher sensitivity. We recommend13C-SBT tests run for at least a 2 h duration. Although cPDR90 was the classifier with highest accuracy and robustness to test duration in this application, concerns remain about its sensitivity to misspecification of the CO2production rate. More research is needed to assess these classifiers in target populations.
Collapse
Affiliation(s)
- Hannah Van Wyk
- Department of Epidemiology, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, United States of America
| | - Gwenyth O Lee
- Rutgers Global Health Institute, 112 Paterson St., New Brunswick, NJ 08901, United States of America
| | - Robert J Schillinger
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Rankine Avenue, East Kilbride G750QF, United Kingdom
- School of Medicine, Dentistry and Nursing, University of Glasgow, New Lister Building, Alexandra Parade, Glasgow G31 2ER, United Kingdom
| | - Christine A Edwards
- School of Medicine, Dentistry and Nursing, University of Glasgow, New Lister Building, Alexandra Parade, Glasgow G31 2ER, United Kingdom
| | - Douglas J Morrison
- Scottish Universities Environmental Research Centre (SUERC), University of Glasgow, Rankine Avenue, East Kilbride G750QF, United Kingdom
| | - Andrew F Brouwer
- Department of Epidemiology, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, United States of America
| |
Collapse
|
4
|
Rachmadi RA, Ariani Y, Alatas FS. IMPACT OF NUTRITIONAL SUPPLEMENTATION ON ENVIRONMENTAL ENTERIC DYSFUNCTION (EED) IN CHILDREN LIVING IN RURAL AREAS: A SYSTEMATIC REVIEW. ARQUIVOS DE GASTROENTEROLOGIA 2024; 61:e23159. [PMID: 39230089 DOI: 10.1590/s0004-2803.24612023-159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/07/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND A staggering 99% of infant undernutrition mortality comes from Sub-Saharan Africa and South Asia. Despite multiple interventions focusing on nutrition adequacy, 2.7 million children worldwide remain associated with undernutrition-related mortality. The lack of impact from multiple interventions toward undernutrition reflects a strong reason to believe that EED is the missing link that sustains undernutrition in low-to-middle-income countries (LMICs). EED is a sub-clinical condition caused by repeated oral enteropathogenic and non-pathogenic fecal microbes exposure that causes intestinal villous malformation, multi-omics changes, chronic intestinal and systemic inflammation, and gut dysbiosis. EED impacts the absorptive capacity and the integrity of the gut, causing a cycle of undernutrition in children. There is currently no protocol for the diagnosis and treatment of EED, hence EED is widely believed to be highly prevalent and underdiagnosed in LMICs. OBJECTIVE To our knowledge, this is the first systematic review to study the impact of nutritional interventions on EED. Previous studies yielded inconsistent results, hence the synthesis of this information is essential in attaining a deeper understanding of EED to formulate new targets of intervention against child undernutrition. METHODS This systematic review is registered to PROSPERO (CRD42022363157) in accordance to PRISMA, using keywords referring to nutrient supplementation, EED, and child growth failure. RESULTS Eleven articles were eligible for review, comprising randomized controlled trials performed mainly in the African continent, with a total of 5689 healthy children eligible for analysis. CONCLUSION The systematic review illustrates that nutritional interventions have a minimal impact on EED biomarkers and linear growth and reflects the importance of understanding better the mechanisms causing EED and its consequences. It appears that the anabolic contribution of nutrition intervention to child growth is negated by EED.
Collapse
Affiliation(s)
| | - Yulia Ariani
- Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Medical Biology, Faculty of Medicine, Universitas Indonesia. Indonesia
| | - Fatima Safira Alatas
- Gastrohepatology Division, Department of Child Health, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| |
Collapse
|
5
|
Fasano A. Environmental enteropathy: an old clinical conundrum with new diagnostic and therapeutic opportunities. Am J Clin Nutr 2024; 120 Suppl 1:S1-S3. [PMID: 39300659 DOI: 10.1016/j.ajcnut.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 05/14/2024] [Indexed: 09/22/2024] Open
Affiliation(s)
- Alessio Fasano
- Mucosal Immunology and Biology Research Center, Mass General Brigham - Harvard Medical School, Boston MA (USA); Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston MA (USA); European Bimedical Research Institute of Salerno (Italy).
| |
Collapse
|
6
|
Kelly P, VanBuskirk K, Coomes D, Mouksassi S, Smith G, Jamil Z, Hossain MS, Syed S, Marie C, Tarr PI, Sullivan PB, Petri WA, Denno DM, Ahmed T, Mahfuz M, Ali SA, Moore SR, Ndao IM, Tearney GJ, Ömer H Yilmaz, Raghavan SS, Moskaluk CA, Liu TC. Histopathology underlying environmental enteric dysfunction in a cohort study of undernourished children in Bangladesh, Pakistan, and Zambia compared with United States children. Am J Clin Nutr 2024; 120 Suppl 1:S15-S30. [PMID: 39300660 DOI: 10.1016/j.ajcnut.2024.02.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is an asymptomatic intestinal disorder associated with growth impairment, delayed neurocognitive development, and impaired oral vaccine responses. OBJECTIVES We set out to develop and validate a histopathologic scoring system on duodenal biopsies from a cohort study of children with growth failure in Bangladesh, Pakistan, and Zambia ("EED") with reference to biopsies from United States children with no clinically reported histologic pathology (referred to hereafter as "normal") or celiac disease. METHODS Five gastrointestinal pathologists evaluated 745 hematoxylin and eosin slide images from 291 children with EED (mean age: 1.6 y) and 66 United States children (mean age: 6.8 y). Histomorphologic features (i.e., villus/crypt architecture, goblet cells, epithelial and lamina propria acute/chronic inflammation, Brunner's glands, Paneth cells, epithelial detachment, enterocyte injury, and foveolar metaplasia) were used to score each histopathologic slide. Generalized estimating equations were used to determine differences between EED, normal, and celiac disease, and receiver operating characteristic curves were used to assess predictive value. RESULTS Biopsies from the duodenal bulb showed higher intramucosal Brunner's gland scores and lower intraepithelial lymphocyte scores than from the second or third parts of the duodenum (D2/3), so only D2/3 were included in the final analysis. Although 7 parameters differed significantly between EED and normal biopsies in regression models, only 5 (blunted villus architecture, increased intraepithelial lymphocytosis, goblet cell depletion, Paneth cell depletion, and reduced intramucosal Brunner's glands) were required to create a total score percentage (TSP-5) that correctly identified EED against normal biopsies (AUC: 0.992; 95% CI: 0.983, 0.998). Geographic comparisons showed more severe goblet cell depletion in Bangladesh and more marked intraepithelial lymphocytosis in Pakistan. CONCLUSIONS This scoring system involving 5 histologic parameters demonstrates very high discrimination between EED and normal biopsies, indicating that this scoring system can be applied with confidence to studies of intestinal biopsies in EED.
Collapse
Affiliation(s)
- Paul Kelly
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, United Kingdom.
| | - Kelley VanBuskirk
- Department of Global Health, University of Washington School of Public Health, Seattle, WA, United States
| | - David Coomes
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, United States
| | | | | | - Zehra Jamil
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Md Shabab Hossain
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Sana Syed
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Chelsea Marie
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Peter B Sullivan
- Department of Paediatrics, Children's Hospital, University of Oxford, Oxford, United Kingdom
| | - William A Petri
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Donna M Denno
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Tahmeed Ahmed
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - S Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sean R Moore
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - I Malick Ndao
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | | | - Ömer H Yilmaz
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Shyam S Raghavan
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Christopher A Moskaluk
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Ta-Chiang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
7
|
Zulqarnain F, Zhao X, Setchell KD, Sharma Y, Fernandes P, Srivastava S, Shrivastava A, Ehsan L, Jain V, Raghavan S, Moskaluk C, Haberman Y, Denson LA, Mehta K, Iqbal NT, Rahman N, Sadiq K, Ahmad Z, Idress R, Iqbal J, Ahmed S, Hotwani A, Umrani F, Amadi B, Kelly P, Brown DE, Moore SR, Ali SA, Syed S. Machine-learning-based integrative -'omics analyses reveal immunologic and metabolic dysregulation in environmental enteric dysfunction. iScience 2024; 27:110013. [PMID: 38868190 PMCID: PMC11167436 DOI: 10.1016/j.isci.2024.110013] [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: 10/12/2023] [Revised: 02/18/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
Environmental enteric dysfunction (EED) is a subclinical enteropathy challenging to diagnose due to an overlap of tissue features with other inflammatory enteropathies. EED subjects (n = 52) from Pakistan, controls (n = 25), and a validation EED cohort (n = 30) from Zambia were used to develop a machine-learning-based image analysis classification model. We extracted histologic feature representations from the Pakistan EED model and correlated them to transcriptomics and clinical biomarkers. In-silico metabolic network modeling was used to characterize alterations in metabolic flux between EED and controls and validated using untargeted lipidomics. Genes encoding beta-ureidopropionase, CYP4F3, and epoxide hydrolase 1 correlated to numerous tissue feature representations. Fatty acid and glycerophospholipid metabolism-related reactions showed altered flux. Increased phosphatidylcholine, lysophosphatidylcholine (LPC), and ether-linked LPCs, and decreased ester-linked LPCs were observed in the duodenal lipidome of Pakistan EED subjects, while plasma levels of glycine-conjugated bile acids were significantly increased. Together, these findings elucidate a multi-omic signature of EED.
Collapse
Affiliation(s)
| | - Xueheng Zhao
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Kenneth D.R. Setchell
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Yash Sharma
- University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | - Varun Jain
- University of Virginia, Charlottesville, VA, USA
| | | | | | - Yael Haberman
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Lee A. Denson
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Khyati Mehta
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | | | | | | | | | | | | | | | | | | | | | - Paul Kelly
- University Teaching Hospital, Lusaka, Zambia
- Queen Mary University of London, London, UK
| | | | | | | | - Sana Syed
- University of Virginia, Charlottesville, VA, USA
- Aga Khan University, Karachi, Pakistan
| |
Collapse
|
8
|
Urugo MM, Teka TA, Lema TB, Lusweti JN, Djedjibegovíc J, Lachat C, Tesfamariam K, Mesfin A, Astatkie T, Abdel-Wahhab MA. Dietary aflatoxins exposure, environmental enteropathy, and their relation with childhood stunting. Int J Food Sci Nutr 2024; 75:241-254. [PMID: 38404064 DOI: 10.1080/09637486.2024.2314676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/16/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Childhood stunting is a global phenomenon affecting more than 149 million children under the age of 5 worldwide. Exposure to aflatoxins (AFs) in utero, during breastfeeding, and consumption of contaminated food affect the gut microbiome, resulting in intestinal dysfunction and potentially contributing to stunting. This review explores the potential relationship between AF exposure, environmental enteropathy and childhood stunting. AFs bind to DNA, disrupt protein synthesis and elicit environmental enteropathy (EE). An EE alters the structure of intestinal epithelial cells, impairs nutrient uptake and leads to malabsorption. This article proposes possible intervention strategies for researchers and policymakers to reduce AF exposure, EE and childhood stunting, such as exposure reduction, the implementation of good agricultural practices, dietary diversification and improving environmental water sanitation and hygiene.
Collapse
Affiliation(s)
- Markos Makiso Urugo
- Department of Food Science and Postharvest Technology, College of Agricultural Sciences, Wachemo University, Hosaina, Ethiopia
- Department of Postharvest Management, College of Agriculture and Veterinary Medicine, Jimma University, Jimma, Ethiopia
| | - Tilahun A Teka
- Department of Postharvest Management, College of Agriculture and Veterinary Medicine, Jimma University, Jimma, Ethiopia
| | - Tefera Belachew Lema
- Department of Nutrition and Dietetics, Faculty of Public Health, Institute of Health, Jimma University, Jimma, Ethiopia
| | | | | | - Carl Lachat
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kokeb Tesfamariam
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
- Department of Public Health, College of Medicine and Health Sciences, Ambo University, Ambo, Ethiopia
| | - Addisalem Mesfin
- Center of Excellence in Mycotoxicology and Public Health, MYTOX-SOUTH, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Department of Human Nutrition, College of Agriculture, Hawassa University, Hawassa, Ethiopia
| | - Tess Astatkie
- Faculty of Agriculture, Dalhousie University, Truro, NS, Canada
| | - Mosaad A Abdel-Wahhab
- Food Toxicology & Contaminants Department, National Research Center, Dokki, Cairo, Egypt
| |
Collapse
|
9
|
Sutter RW, Eisenhawer M, Molodecky NA, Verma H, Okayasu H. Inactivated Poliovirus Vaccine: Recent Developments and the Tortuous Path to Global Acceptance. Pathogens 2024; 13:224. [PMID: 38535567 PMCID: PMC10974833 DOI: 10.3390/pathogens13030224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/21/2024] Open
Abstract
Inactivated poliovirus vaccine (IPV), available since 1955, became the first vaccine to be used to protect against poliomyelitis. While the immunogenicity of IPV to prevent paralytic poliomyelitis continues to be irrefutable, its requirement for strong containment (due to large quantities of live virus used in the manufacturing process), perceived lack of ability to induce intestinal mucosal immunity, high cost and increased complexity to administer compared to oral polio vaccine (OPV), have limited its use in the global efforts to eradicate poliomyelitis. In order to harvest the full potential of IPV, a program of work has been carried out by the Global Polio Eradication Initiative (GPEI) over the past two decades that has focused on: (1) increasing the scientific knowledge base of IPV; (2) translating new insights and evidence into programmatic action; (3) expanding the IPV manufacturing infrastructure for global demand; and (4) continuing to pursue an ambitious research program to develop more immunogenic and safer-to-produce vaccines. While the knowledge base of IPV continues to expand, further research and product development are necessary to ensure that the program priorities are met (e.g., non-infectious production through virus-like particles, non-transmissible vaccine inducing humoral and intestinal mucosal immunity and new methods for house-to-house administration through micro-needle patches and jet injectors), the discussions have largely moved from whether to how to use this vaccine most effectively. In this review, we summarize recent developments on expanding the science base of IPV and provide insight into policy development and the expansion of IPV manufacturing and production, and finally we provide an update on the current priorities.
Collapse
Affiliation(s)
| | - Martin Eisenhawer
- Polio Eradication Department, World Health Organization, 1211 Geneva, Switzerland; (M.E.); (H.V.)
| | - Natalia A. Molodecky
- Polio Surge Capacity Support Program, The Task Force for Global Health, Inc., Decatur, GE 30030, USA;
| | - Harish Verma
- Polio Eradication Department, World Health Organization, 1211 Geneva, Switzerland; (M.E.); (H.V.)
| | - Hiromasa Okayasu
- Division of Healthy Environments and Population, Regional Office for the Western Pacific, World Health Organization, Manila 1000, Philippines
| |
Collapse
|
10
|
Fahim SM, Donowitz JR, Smirnova E, Jan NJ, Das S, Mahfuz M, Gaffar SMA, Petri WA, Marie C, Ahmed T. Small Intestine Bacterial Overgrowth is associated with increased Campylobacter and epithelial injury in duodenal biopsies of Bangladeshi children. PLoS Negl Trop Dis 2024; 18:e0012023. [PMID: 38536881 PMCID: PMC11020352 DOI: 10.1371/journal.pntd.0012023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 04/16/2024] [Accepted: 02/26/2024] [Indexed: 04/18/2024] Open
Abstract
Small intestine bacterial overgrowth (SIBO) has been associated with enteric inflammation, linear growth stunting, and neurodevelopmental delays in children from low-income countries. Little is known about the histologic changes or epithelial adherent microbiota associated with SIBO. We sought to describe these relationships in a cohort of impoverished Bangladeshi children. Undernourished 12-18-month-old children underwent both glucose hydrogen breath testing for SIBO and duodenoscopy with biopsy. Biopsy samples were subject to both histological scoring and 16s rRNA sequencing. 118 children were enrolled with 16s sequencing data available on 53. Of 11 histological features, we found that SIBO was associated with one, enterocyte injury in the second part of the duodenum (R = 0.21, p = 0.02). SIBO was also associated with a significant increase in Campylobacter by 16s rRNA analysis (Log 2-fold change of 4.43; adjusted p = 1.9 x 10-6). These findings support the growing body of literature showing an association between SIBO and enteric inflammation and enterocyte injury and further delineate the subgroup of children with environmental enteric dysfunction who have SIBO. Further, they show a novel association between SIBO and Campylobacter. Mechanistic work is needed to understand the relationship between SIBO, enterocyte injury, and Campylobacter.
Collapse
Affiliation(s)
- Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States of America
| | - Jeffrey R. Donowitz
- Division of Pediatric Infectious Diseases, Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, Virginia, United States of America
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
- Division of Pediatric Infectious Diseases, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ekaterina Smirnova
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Ning-Juin Jan
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - S. M. Abdul Gaffar
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - William A. Petri
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Chelsea Marie
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Office of the Executive Director, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| |
Collapse
|
11
|
Cowardin CA, Syed S, Iqbal N, Jamil Z, Sadiq K, Iqbal J, Ali SA, Moore SR. Environmental enteric dysfunction: gut and microbiota adaptation in pregnancy and infancy. Nat Rev Gastroenterol Hepatol 2023; 20:223-237. [PMID: 36526906 PMCID: PMC10065936 DOI: 10.1038/s41575-022-00714-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 03/31/2023]
Abstract
Environmental enteric dysfunction (EED) is a subclinical syndrome of intestinal inflammation, malabsorption and barrier disruption that is highly prevalent in low- and middle-income countries in which poverty, food insecurity and frequent exposure to enteric pathogens impair growth, immunity and neurodevelopment in children. In this Review, we discuss advances in our understanding of EED, intestinal adaptation and the gut microbiome over the 'first 1,000 days' of life, spanning pregnancy and early childhood. Data on maternal EED are emerging, and they mirror earlier findings of increased risks for preterm birth and fetal growth restriction in mothers with either active inflammatory bowel disease or coeliac disease. The intense metabolic demands of pregnancy and lactation drive gut adaptation, including dramatic changes in the composition, function and mother-to-child transmission of the gut microbiota. We urgently need to elucidate the mechanisms by which EED undermines these critical processes so that we can improve global strategies to prevent and reverse intergenerational cycles of undernutrition.
Collapse
Affiliation(s)
- Carrie A Cowardin
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA
| | - Sana Syed
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeha Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Zehra Jamil
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Junaid Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Syed Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sean R Moore
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
12
|
Besa E, Tembo MJ, Mulenga C, Mweetwa M, Choudhry N, Chandwe K, Storer C, Head R, Amadi B, Haritunians T, McGovern D, Kwenda G, Peiris M, Kelly P. Potential determinants of low circulating glucagon-like peptide 2 concentrations in Zambian children with non-responsive stunting. Exp Physiol 2023; 108:568-580. [PMID: 36744850 PMCID: PMC10103869 DOI: 10.1113/ep090492] [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: 01/23/2023] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
NEW FINDINGS What is the central question of this study? Non-responsive stunting is characterised by a progressive decline of circulating glucagon-like peptide 2: what are the possible causes of this decline? What is the main finding and its importance? In contrast with the established loss of Paneth and goblet cells in environmental enteropathy, there was no evidence of a parallel loss of enteroendocrine cells as seen by positive tissue staining for chromogranin A. Transcriptomic and genomic analyses showed evidence of genetic transcripts that could account for some of the variability seen in circulating glucagon-like peptide 2 values. ABSTRACT Nutrient sensing determines digestive and hormonal responses following nutrient ingestion. We have previously reported decreased levels of glucagon-like peptide 2 (GLP-2) in children with stunting. Here we demonstrate the presence of enteroendocrine cells in stunted children and explore potential pathways that may be involved in reduced circulating levels of GLP-2. At the time of performing diagnostic endoscopies for non-responsive stunted children, intestinal biopsies were collected for immunofluorescence staining of enteroendocrine cells and transcriptomic analysis. Circulating levels of GLP-2 were also measured and correlated with transcriptomic data. An exploratory genome-wide association study (GWAS) was conducted on DNA samples (n = 158) to assess genetic contribution to GLP-2 variability. Intestinal tissue sections collected from non-responsive stunted children stained positive for chromogranin A (88/89), alongside G-protein-coupled receptors G-protein receptor 119 (75/87), free fatty acid receptor 3 (76/89) and taste 1 receptor 1 (39/45). Transcriptomic analysis found three pathways correlated with circulating GLP-2: sugar metabolism, epithelial transport, and barrier function, which likely reflect downstream events following receptor-ligand interaction. GWAS analysis revealed potential genetic contributions to GLP-2 half-life and receptor binding. Enteroendocrine cell loss was not identified in stunted Zambian children as has been observed for goblet and Paneth cells. Transcriptomic analysis suggests that GLP-2 has pleiotrophic actions on the intestinal mucosa in malnutrition, but further work is needed to dissect pathways leading to perturbations in nutrient sensing.
Collapse
Affiliation(s)
- Ellen Besa
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Mizinga Jacqueline Tembo
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Chola Mulenga
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Monica Mweetwa
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Naheed Choudhry
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Kanta Chandwe
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Chad Storer
- Genome Technology Access Center at McDonnell Genome InstituteWashington University in St LouisSt LouisMOUSA
| | - Richard Head
- Genome Technology Access Center at McDonnell Genome InstituteWashington University in St LouisSt LouisMOUSA
| | - Beatrice Amadi
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
| | - Talin Haritunians
- Cedars‐Sinai Medical CenterInflammatory Bowel and Immunobiology Research InstituteLos AngelesCAUSA
| | - Dermot McGovern
- Cedars‐Sinai Medical CenterInflammatory Bowel and Immunobiology Research InstituteLos AngelesCAUSA
| | - Geoffrey Kwenda
- Department of Biomedical Sciences, School of Health SciencesUniversity of ZambiaLusakaZambia
| | - Madusha Peiris
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Paul Kelly
- Tropical Gastroenterology and Nutrition Group, School of MedicineUniversity of ZambiaLusakaZambia
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
13
|
Oriá RB, Freitas RS, Roque CR, Nascimento JCR, Silva AP, Malva JO, Guerrant RL, Vitek MP. ApoE Mimetic Peptides to Improve the Vicious Cycle of Malnutrition and Enteric Infections by Targeting the Intestinal and Blood-Brain Barriers. Pharmaceutics 2023; 15:pharmaceutics15041086. [PMID: 37111572 PMCID: PMC10141726 DOI: 10.3390/pharmaceutics15041086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Apolipoprotein E (apoE) mimetic peptides are engineered fragments of the native apoE protein’s LDL-receptor binding site that improve the outcomes following a brain injury and intestinal inflammation in a variety of models. The vicious cycle of enteric infections and malnutrition is closely related to environmental-driven enteric dysfunction early in life, and such chronic inflammatory conditions may blunt the developmental trajectories of children with worrisome and often irreversible physical and cognitive faltering. This window of time for microbiota maturation and brain plasticity is key to protecting cognitive domains, brain health, and achieving optimal/full developmental potential. This review summarizes the potential role of promising apoE mimetic peptides to improve the function of the gut-brain axis, including targeting the blood-brain barrier in children afflicted with malnutrition and enteric infections.
Collapse
Affiliation(s)
- Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
- Correspondence: ; Tel.: +55-85-3366-8239
| | - Raul S. Freitas
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
| | - Cássia R. Roque
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
| | - José Carlos R. Nascimento
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
- Institute of Health Sciences, Medicine, University of International Integration of Afro-Brazilian Lusofonia, Redenção 62790-970, Brazil
| | - Ana Paula Silva
- Institute of Pharmacology and Experimental Therapeutics and Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - João O. Malva
- Institute of Pharmacology and Experimental Therapeutics and Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Michael P. Vitek
- Division of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
14
|
Liu Z, Fan YM, Ashorn P, Chingwanda C, Maleta K, Hallamaa L, Hyöty H, Chaima D, Ashorn U. Lack of Associations between Environmental Exposures and Environmental Enteric Dysfunction among 18-Month-Old Children in Rural Malawi. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10891. [PMID: 36078607 PMCID: PMC9517768 DOI: 10.3390/ijerph191710891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
Environmental enteric dysfunction (EED) is common and contributes to linear growth faltering (stunting) and mortality among children in low-resource settings. A few studies on the environmental causes of EED have been conducted but the exact exposures that cause or predispose children to EED are context-specific and not clear. This study aimed to assess associations between selected environmental exposures and EED markers among 620 18-month-old children. This was a secondary analysis of data from Malawian children who participated in a randomized controlled trial (iLiNS-DYAD, registered at clinicaltrials.gov as NCT01239693) from birth to 18 months of age. Data on environmental exposures, including drinking water source, sanitation, exposure to animals, housing materials, season, residential area, and food insecurity were collected at enrolment. Biomarkers of EED included concentrations of calprotectin, regenerating 1B protein (REG1B), and alpha-1-antitrypsin from stool samples to assess intestinal inflammation, repair, and permeability, respectively. We performed bivariate and multivariable analyses to assess associations between environmental exposures and EED biomarkers. Adjusting for possible confounders, we did not find associations between the selected environmental exposures and the three biomarkers. These results do not provide support for our hypothesis that the studied adverse environmental exposures are associated with increased concentrations of children's EED markers in rural Malawi.
Collapse
Affiliation(s)
- Zhifei Liu
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Yue-Mei Fan
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Per Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
- Department of Paediatrics, Tampere University Hospital, 33521 Tampere, Finland
| | - Chilungamo Chingwanda
- School of Public Health & Family Medicine, Kamuzu University of Health Sciences, Private Bag 360, Chichiri, Blantyre 3, Malawi
| | - Kenneth Maleta
- School of Public Health & Family Medicine, Kamuzu University of Health Sciences, Private Bag 360, Chichiri, Blantyre 3, Malawi
| | - Lotta Hallamaa
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| | - Heikki Hyöty
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, 33521 Tampere, Finland
| | - David Chaima
- School of Public Health & Family Medicine, Kamuzu University of Health Sciences, Private Bag 360, Chichiri, Blantyre 3, Malawi
| | - Ulla Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland
| |
Collapse
|
15
|
Vonaesch P, Winkel M, Kapel N, Nestoret A, Barbot-Trystram L, Pontoizeau C, Barouki R, Rakotondrainipiana M, Kandou K, Andriamanantena Z, Andrianonimiadana L, Habib A, Rodriguez-Pozo A, Hasan M, Vigan-Womas I, Collard JM, Gody JC, Djorie S, Sansonetti PJ, Randremanana RV. Putative Biomarkers of Environmental Enteric Disease Fail to Correlate in a Cross-Sectional Study in Two Study Sites in Sub-Saharan Africa. Nutrients 2022; 14:nu14163312. [PMID: 36014817 PMCID: PMC9412633 DOI: 10.3390/nu14163312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Environmental enteric dysfunction (EED) is an elusive, inflammatory syndrome of the small intestine thought to be associated with enterocyte loss and gut leakiness and lead to stunted child growth. To date, the gold standard for diagnosis is small intestine biopsy followed by histology. Several putative biomarkers for EED have been proposed and are widely used in the field. Here, we assessed in a cross-sectional study of children aged 2–5 years for a large set of biomarkers including markers of protein exudation (duodenal and fecal alpha-1-antitrypsin (AAT)), inflammation (duodenal and fecal calprotectin, duodenal, fecal and blood immunoglobulins, blood cytokines, C-reactive protein (CRP)), gut permeability (endocab, lactulose-mannitol ratio), enterocyte mass (citrulline) and general nutritional status (branched-chain amino acids (BCAA), insulin-like growth factor) in a group of 804 children in two Sub-Saharan countries. We correlated these markers with each other and with anemia in stunted and non-stunted children. AAT and calprotectin, CRP and citrulline and citrulline and BCAA correlated with each other. Furthermore, BCAA, citrulline, ferritin, fecal calprotectin and CRP levels were correlated with hemoglobin levels. Our results show that while several of the biomarkers are associated with anemia, there is little correlation between the different biomarkers. Better biomarkers and a better definition of EED are thus urgently needed.
Collapse
Affiliation(s)
- Pascale Vonaesch
- Unité de Pathogénie Microbienne, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
- Department of Fundamental Microbiology, University of Lausanne, Campus UNIL-Sorge, 1015 Lausanne, Switzerland
- Human and Animal Health Unit, Swiss Tropical and Public Health Institute & University of Basel, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- Correspondence:
| | - Munir Winkel
- Department of Fundamental Microbiology, University of Lausanne, Campus UNIL-Sorge, 1015 Lausanne, Switzerland
| | - Nathalie Kapel
- Service de Coprologie Fonctionnelle, Hôpital Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, 47-83 boulevard de l’Hôpital, 75013 Paris, France
| | - Alison Nestoret
- Service de Coprologie Fonctionnelle, Hôpital Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, 47-83 boulevard de l’Hôpital, 75013 Paris, France
| | - Laurence Barbot-Trystram
- Service de Coprologie Fonctionnelle, Hôpital Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, 47-83 boulevard de l’Hôpital, 75013 Paris, France
| | - Clément Pontoizeau
- Laboratoire de biochimie métabolique, Hôpital Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris, 149 Rue de Sèvres, 75015 Paris, France
| | - Robert Barouki
- Laboratoire de biochimie métabolique, Hôpital Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris, 149 Rue de Sèvres, 75015 Paris, France
| | - Maheninasy Rakotondrainipiana
- Unité d’Epidémiologie et de Recherche Clinique, Institut Pasteur de Madagascar, BP 1274, Antananarivo 101, Madagascar
| | - Kaleb Kandou
- Unité d’Epidémiologie, Institut Pasteur de Bangui, Bangui BP 923, Central African Republic
| | - Zo Andriamanantena
- Unité d’Immunologie des Maladies Infectieuses, Institut Pasteur de Madagascar, BP 1274, Antananarivo 101, Madagascar
| | - Lova Andrianonimiadana
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, BP 1274, Antananarivo 101, Madagascar
| | - Azimdine Habib
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, BP 1274, Antananarivo 101, Madagascar
| | - Andre Rodriguez-Pozo
- Unité de Pathogénie Microbienne, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
- Cytometry and Biomarkers Unit of Technology and Service, Institut Pasteur and Université Paris Cité, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Milena Hasan
- Cytometry and Biomarkers Unit of Technology and Service, Institut Pasteur and Université Paris Cité, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Inès Vigan-Womas
- Unité d’Immunologie des Maladies Infectieuses, Institut Pasteur de Madagascar, BP 1274, Antananarivo 101, Madagascar
| | - Jean-Marc Collard
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, BP 1274, Antananarivo 101, Madagascar
| | | | - Serge Djorie
- Unité d’Epidémiologie, Institut Pasteur de Bangui, Bangui BP 923, Central African Republic
| | - Philippe J. Sansonetti
- Unité de Pathogénie Microbienne, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Rindra Vatosoa Randremanana
- Unité d’Epidémiologie et de Recherche Clinique, Institut Pasteur de Madagascar, BP 1274, Antananarivo 101, Madagascar
| | | |
Collapse
|
16
|
Engineering immunity via skin-directed drug delivery devices. J Control Release 2022; 345:385-404. [DOI: 10.1016/j.jconrel.2022.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 12/18/2022]
|
17
|
Denson LA. Application of mucosal functional genomics to childhood undernutrition and stunting: Insights into mechanisms and targeted interventions. EBioMedicine 2021; 71:103553. [PMID: 34482071 PMCID: PMC8426530 DOI: 10.1016/j.ebiom.2021.103553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 11/03/2022] Open
Affiliation(s)
- Lee A Denson
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Cincinnati College of Medicine and the Cincinnati Children's Hospital Medical Center, MLC 2010, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|