1
|
Mvalo T, Dhaded SM, Manji KP, Vesel L, Semrau KEA, Kisenge R, Somji S, Chiume M, Saidi F, Hoffman IF, Vernekar S, Bellad R, Koppad B, Tuller DE, Mokhtar R, Lee ACC, North K, Sudfeld CR. Mortality, morbidity and growth among moderately low birthweight infants in India, Malawi, and Tanzania. BMC Pediatr 2025; 25:316. [PMID: 40264138 PMCID: PMC12016151 DOI: 10.1186/s12887-025-05668-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Despite notable global reductions in infant and under-five mortality over the last two decades, about half of the remaining neonatal deaths occur among low birth weight (LBW) infants. We conducted a prospective study to characterize the mortality risk and morbidity of moderately LBW (MLBW; 1500-2499 g birth weight) infants during the first year of life in India, Malawi, and Tanzania. METHODS The multi-site Low Birthweight Infant Feeding Exploration (LIFE) study was conducted from September 2019 to July 2021 and followed a cohort of MLBW infants from India, Malawi, and Tanzania from birth to 52 weeks of age. At follow-up visits conducted at 1, 2, 4, 6, 10, 14, 18, 26, 39, and 52 weeks of age, mothers/caregivers were asked to recall the presence of diarrhea, fever, acute respiratory infections, and convulsions during the past week, and infant weight and length were assessed. Generalized estimating equations (GEE) were used to evaluate study site and sociodemographic risk factors for infant morbidity and mortality, and also to assess the relationship between infant morbidity and anthropometric measures. RESULTS A total of 1,121 MLBW infants were included in the analysis and 47 (4.2%) deaths were recorded by the age of 12 months. Preterm-appropriate-for-gestational age infants had approximately twice the risk of infant death compared to term-small-for-gestational age infants (RR: 2.09; 95% CI: 1.08, 4.05). Period prevalence of diarrhea and fever increased with infant age and differed by study site (p-values < 0.05). In time-varying analyses, reported diarrhea during the past week was associated with lower length-for-age z-score (LAZ) (mean difference (MD): -0.20; 95% CI: -0.31, -0.09), weight-for-age z-score (WAZ) (MD: -0.25; 95% CI: -0.35, -0.16), and weight-for-length z-score (WLZ) (MD: -0.24; 95% CI: -0.36, -0.12), while fever was associated with lower WAZ (MD: -0.14; 95% CI: -0.21, -0.06), and WLZ (MD: -0.17; 95% CI: -0.26, -0.08) but not LAZ at the concurrent study visit. CONCLUSION The risk of death during the first year of life is high for MLBW, but differs by the contribution of prematurity and size-for-gestational age. Interventions that reduce the incidence of diarrhea and fever may improve the growth of MLBW infants. TRIAL REGISTRATION The LIFE study was registered with ClinicalTrials.gov (NCT04002908).
Collapse
Affiliation(s)
- Tisungane Mvalo
- University of North Carolina Project Malawi, Lilongwe, Malawi.
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| | - Sangappa M Dhaded
- Department of Pediatrics, Jawaharlal Nehru Medical College, KLE Academy of Higher Education and Research, Belgaum, Karnataka, India
| | - Karim P Manji
- Department of Pediatrics and Child Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Linda Vesel
- Ariadne Labs, Harvard T.H. Chan School of Public Health / Brigham and Women's Hospital, Boston, MA, USA
| | - Katherine E A Semrau
- Ariadne Labs, Harvard T.H. Chan School of Public Health / Brigham and Women's Hospital, Boston, MA, USA
| | - Rodrick Kisenge
- Department of Pediatrics and Child Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Sarah Somji
- Department of Pediatrics and Child Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Msandeni Chiume
- Department of Pediatrics, Kamuzu Central Hospital (KCH), Ministry of Health, Lilongwe, Malawi
| | - Friday Saidi
- University of North Carolina Project Malawi, Lilongwe, Malawi
- Department of Obstetrics and Gynecology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Irving F Hoffman
- University of North Carolina Project Malawi, Lilongwe, Malawi
- Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Sunil Vernekar
- Department of Physiology, Jawaharlal Nehru Medical College, KLE Academy of Higher Education and Research, Belgaum, Karnataka, India
| | - Roopa Bellad
- Department of Pediatrics, Jawaharlal Nehru Medical College, KLE Academy of Higher Education and Research, Belgaum, Karnataka, India
| | - Bhavana Koppad
- Department of Pediatrics, Jawaharlal Nehru Medical College, KLE Academy of Higher Education and Research, Belgaum, Karnataka, India
| | - Danielle E Tuller
- Ariadne Labs, Harvard T.H. Chan School of Public Health / Brigham and Women's Hospital, Boston, MA, USA
| | - Rana Mokhtar
- Ariadne Labs, Harvard T.H. Chan School of Public Health / Brigham and Women's Hospital, Boston, MA, USA
| | - Anne C C Lee
- Department of Pediatrics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Brown Alliance for Infant and Maternal Health Research, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Krysten North
- Department of Pediatrics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher R Sudfeld
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
2
|
Sthity RA, Islam MZ, Sagar MEK, Gazi MA, Ferdous J, Kabir MM, Mahfuz M, Ahmed T, Mostafa I. Association of Escherichia coli pathotypes with fecal markers of enteropathy and nutritional status among underweight adults in Bangladesh. Front Cell Infect Microbiol 2025; 15:1553688. [PMID: 40276386 PMCID: PMC12018316 DOI: 10.3389/fcimb.2025.1553688] [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: 12/31/2024] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Environmental enteric dysfunction (EED), a subclinical intestinal disorder, is characterized by chronic fecal-oral exposure to entero-pathogens and could be diagnosed by measuring non-invasive biomarkers. Escherichia coli is the one of the key bacterial enteric pathogens that drives EED, but there is a lack of information on the E. coli pathotypes in relation to the biomarkers of EED in malnourished adults. Here, we intended to measure the possible association of these pathotypes with EED biomarkers and nutritional status of adults residing in a slum in Bangladesh. Method Fecal samples were collected from 524 malnourished adults (BMI ≤18.5 kg/m2) living in a slum-setting in Dhaka from March 2016 to September 2019 and analyzed by TaqMan Array Card assays to evaluate the presence of E. coli pathotypes and other entero-pathogens. The multivariable linear regression model was used to assess the association. Results In these malnourished adults, the most prevalent pathotype of E. coli was EAEC (61.7%) and the least prevalent was STEC (6.7%). The prevalence of atypical EPEC, ETEC and Shigella/EIEC were 52%, 48.9% and 45.1% respectively. The infection with atypical EPEC had significant positive association with levels of Myeloperoxidase (b = 0.38; 95% CI = 0.11, 0.65; p-value = 0.006). Similarly, a significantly higher concentration of alpha-1-antitrypsin (b = 0.13; 95% CI = 0.03, 0.22; p-value = 0.011) was found in the STEC-infected adults. However, no notable association was found between the E. coli pathotypes and nutritional status of these adult participants. Moreover, Plesiomonas infected adults were more likely to be infected with EAEC (p-value = 0.017), ETEC (p-value <0.001) and STEC (pvalue = 0.002). Significant coinfection was also detected among the pathotypes and other entero-pathogens such as Giardia, Ascaris, Campylobacter, Salmonella, Enterocytozoon bieneusi, and Adenovirus. Discussion The study results imply that there is an influence of particular E. coli pathotypes (EPEC and STEC) on intestinal inflammation and gut permeability of the malnourished Bangladeshi adults, but no association with nutritional status is found. Potential pathogenicity of the E. coli pathotypes is also observed when co-infection with other pathogens exists in these adults.
Collapse
Affiliation(s)
- Rahvia Alam Sthity
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Zahidul Islam
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Ehsanul Kabir Sagar
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Amran Gazi
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Jafrin Ferdous
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Mamun Kabir
- Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Office of the Executive Director, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| | - Ishita Mostafa
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
3
|
Sinha S, Hackl LS, Huey SL, Lambertini E, Nordhagen S, Bennett AM, Shrestha N, Cole NL, Finkelstein JL, Mehta S. Overview of foodborne hazards associated with inflammation and metabolic health. BMC GLOBAL AND PUBLIC HEALTH 2025; 3:31. [PMID: 40200316 PMCID: PMC11980346 DOI: 10.1186/s44263-025-00150-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Access to safe and nutritious food is key to ensuring health and well-being and is critical to meeting the United Nations' Sustainable Development Goals. However, a synthesis of the associations between foodborne illness and malnutrition, such as metabolic health, remains a gap in the literature base. In this review, we summarized existing evidence on the impacts of biological and chemical hazards on nutrition-related health outcomes, specifically overweight and obesity, inflammation, metabolic disease, thyroid function, cancer development, and adverse birth outcomes, examining physiological mechanisms, epidemiological associations, and animal studies. Mechanisms between some foodborne hazards, such as H. pylori, and adverse pregnancy outcomes, e.g., gestational diabetes mellitus, or between nitrates and impaired thyroid function, are relatively well-studied. However, evidence on the effects of many other chemical hazards on metabolic and human health remains limited: for example, while arsenic exposure is associated with adverse birth outcomes, the limited availability of dose-response studies and other challenges limit ascertaining its causal role. Untangling these associations and physiological mechanisms is of high relevance for both high- as well as low- and middle-income countries. Emerging technologies and novel assessment techniques are needed to improve the detection and understanding of understudied and complex foodborne diseases, particularly those arising from chemical hazards. These evidence gaps are highlighted in this review, as well as the need for establishing surveillance systems for monitoring foodborne diseases and metabolic health outcomes across populations.
Collapse
Affiliation(s)
- Srishti Sinha
- Cornell Joan Klein Jacobs Center for Precision Nutrition and Health, Cornell University, 3101 Martha van Rensselaer Hall, Ithaca, NY, 14853, USA
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Laura S Hackl
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Samantha L Huey
- Cornell Joan Klein Jacobs Center for Precision Nutrition and Health, Cornell University, 3101 Martha van Rensselaer Hall, Ithaca, NY, 14853, USA
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | | | | | - Anna M Bennett
- Cornell Joan Klein Jacobs Center for Precision Nutrition and Health, Cornell University, 3101 Martha van Rensselaer Hall, Ithaca, NY, 14853, USA
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Nidhi Shrestha
- Cornell Joan Klein Jacobs Center for Precision Nutrition and Health, Cornell University, 3101 Martha van Rensselaer Hall, Ithaca, NY, 14853, USA
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Nathaniel L Cole
- Cornell Joan Klein Jacobs Center for Precision Nutrition and Health, Cornell University, 3101 Martha van Rensselaer Hall, Ithaca, NY, 14853, USA
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Julia L Finkelstein
- Cornell Joan Klein Jacobs Center for Precision Nutrition and Health, Cornell University, 3101 Martha van Rensselaer Hall, Ithaca, NY, 14853, USA
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - Saurabh Mehta
- Cornell Joan Klein Jacobs Center for Precision Nutrition and Health, Cornell University, 3101 Martha van Rensselaer Hall, Ithaca, NY, 14853, USA.
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
4
|
Nakiranda R, Malan L, Ricci H, Kruger HS, Nienaber A, Visser M, Ricci C, Faber M, Smuts CM. Daily Complementary Feeding With Eggs Improves Fibroblast Growth Factor 21 in Infants. MATERNAL & CHILD NUTRITION 2025; 21:e13782. [PMID: 39648796 PMCID: PMC11956054 DOI: 10.1111/mcn.13782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 12/10/2024]
Abstract
This secondary analysis of the Eggcel-growth study investigated the effect of daily egg intake for 6 months in infants aged 6-9 months on environmental enteric dysfunction (EED) biomarkers and the association of EED markers with growth faltering. A randomised controlled trial was conducted in Jouberton, South Africa, among 500 infants randomly assigned equally to either an intervention group receiving a daily chicken egg or a control group. Both groups were followed up for 6 months. Data on infant and maternal sociodemographic information and anthropometric status of infants were collected. EED and inflammatory markers were analysed using Q-Plex Human EED (11-Plex) assay. There was a significant reduction in fibroblast growth factor 21 (FGF21) concentration in the intervention group (B = -0.132; 95% CI -0.255, -0.010; p = 0.035). Baseline, insulin-like growth factor 1 (IGF-1) was positively associated with endpoint length-for-age z-score (LAZ), weight-for-age z-score (WAZ) and weight-for-length z-score (WLZ) and there was an inverse relationship between baseline FGF21 and intestinal fatty acid-binding protein (I-FABP) with endpoint growth indicators. Baseline IGF-1 was positively associated with reduced odds of wasting, stunting and being underweight (p < 0.001) and baseline FGF21 was associated with increased odds of stunting (p = 0.002), wasting (p = 0.031) and being underweight (p = 0.035). There was a 20% increased odds of stunting with baseline I-FABP (p = 0.045) and a 30% increased odds of being underweight with baseline soluble CD14 (p = 0.039). Complementary feeding with eggs decreased growth hormone resistance (reduced FGF21 levels); however, FGF21 and I-FABP levels were linked to increased growth faltering. Trial Registration: ClinicalTrials.gov: NCT05168085.
Collapse
Affiliation(s)
- Regina Nakiranda
- Centre of Excellence for NutritionNorth‐West UniversityPotchefstroomSouth Africa
| | - Linda Malan
- Centre of Excellence for NutritionNorth‐West UniversityPotchefstroomSouth Africa
| | - Hannah Ricci
- Centre of Excellence for NutritionNorth‐West UniversityPotchefstroomSouth Africa
- Africa Unit for Transdisciplinary Health Research (AUTHeR)North‐West UniversityPotchefstroomSouth Africa
| | - Herculina S. Kruger
- Centre of Excellence for NutritionNorth‐West UniversityPotchefstroomSouth Africa
| | - Arista Nienaber
- Centre of Excellence for NutritionNorth‐West UniversityPotchefstroomSouth Africa
| | - Marina Visser
- Centre of Excellence for NutritionNorth‐West UniversityPotchefstroomSouth Africa
| | - Cristian Ricci
- Africa Unit for Transdisciplinary Health Research (AUTHeR)North‐West UniversityPotchefstroomSouth Africa
| | - Mieke Faber
- Centre of Excellence for NutritionNorth‐West UniversityPotchefstroomSouth Africa
- South African Medical Research Council, Non‐Communicable Diseases Research UnitTygerbergSouth Africa
| | - Cornelius M. Smuts
- Centre of Excellence for NutritionNorth‐West UniversityPotchefstroomSouth Africa
| |
Collapse
|
5
|
Schwendler TR, Keller KL, Jensen L, Na M, Fofana ML, Daffé M, Sankhon H, Kodish SR. Caregiver Feeding Practices in Guinea: Implications for Infant Dietary Diversity. MATERNAL & CHILD NUTRITION 2025:e70017. [PMID: 40108478 DOI: 10.1111/mcn.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
The aim of this study was to assess the extent to which specific feeding styles may be associated with the diets of infants aged 6-9 months in Guinea. This study was designed to have multiple, iterative phases with methodological triangulation. During Phase 1 of data collection, direct observations (n = 10) were used to develop a tailored tool for Phase 2, during which 72 meal observations were conducted among infants aged 6-9 months to define caregiver feeding styles. Specific behaviours underlying established feeding styles were recorded at the level of the intended bite. Following each observation, infant diet diversity scores (DDS), or the number of food groups consumed in the previous 24 h, were collected. During Phase 3, we interviewed 34 caregivers to understand the drivers of their feeding styles. Caregiver feeding styles were determined using cluster analysis of observed behaviours and a linear regression was used to explore the relationship between feeding style and infant DDS. Textual data from interviews were thematically analysed to explain the drivers of feeding style. Caregivers were characterized as those using forceful (n = 12), responsive (n = 52) or uninvolved (n = 8) feeding styles. Our study found that responsive feeding was not associated with a higher DDS when controlling for child age in months. The most salient factors shaping feeding style in this setting included perception around infant and young child developmental stage, food refusals and trust in infant cues. Overall, food insecurity may need to be addressed in addition to feeding styles to improve DDS.
Collapse
Affiliation(s)
- Teresa R Schwendler
- Department of Nutritional Sciences, The Pennsylvania State University, State College, Pennsylvania, USA
| | - Kathleen L Keller
- Department of Nutritional Sciences, The Pennsylvania State University, State College, Pennsylvania, USA
- Department of Food Science, The Pennsylvania State University, State College, Pennsylvania, USA
| | - Leif Jensen
- Department of Agricultural Economics, Sociology, and Education, The Pennsylvania State University, State College, Pennsylvania, USA
| | - Muzi Na
- Department of Nutritional Sciences, The Pennsylvania State University, State College, Pennsylvania, USA
| | | | - Mamady Daffé
- Ministère de la Santé et de l'Hygiène Publique, Conakry, Guinée
| | - Hermine Sankhon
- Department of Nutritional Sciences, The Pennsylvania State University, State College, Pennsylvania, USA
| | - Stephen R Kodish
- Department of Nutritional Sciences, The Pennsylvania State University, State College, Pennsylvania, USA
- Department of Biobehavioral Health, The Pennsylvania State University, State College, Pennsylvania, USA
| |
Collapse
|
6
|
Butzin-Dozier Z, Ji Y, Coyle J, Malenica I, Rogawski McQuade ET, Grembi JA, Platts-Mills JA, Houpt ER, Graham JP, Ali S, Rahman MZ, Alauddin M, Famida SL, Akther S, Hossen MS, Mutsuddi P, Shoab AK, Rahman M, Islam MO, Miah R, Taniuchi M, Liu J, Alauddin ST, Stewart CP, Luby SP, Colford Jr. JM, Hubbard AE, Mertens AN, Lin A. Treatment heterogeneity of water, sanitation, hygiene, and nutrition interventions on child growth by environmental enteric dysfunction and pathogen status for young children in Bangladesh. PLoS Negl Trop Dis 2025; 19:e0012881. [PMID: 39965021 PMCID: PMC11882089 DOI: 10.1371/journal.pntd.0012881] [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: 06/12/2024] [Revised: 03/05/2025] [Accepted: 01/30/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Water, sanitation, hygiene (WSH), nutrition (N), and combined (N+WSH) interventions are often implemented by global health organizations, but WSH interventions may insufficiently reduce pathogen exposure, and nutrition interventions may be modified by environmental enteric dysfunction (EED), a condition of increased intestinal permeability and inflammation. This study investigated the heterogeneity of these treatments' effects based on individual pathogen and EED biomarker status with respect to child linear growth. METHODS We applied cross-validated targeted maximum likelihood estimation and super learner ensemble machine learning to assess the conditional treatment effects in subgroups defined by biomarker and pathogen status. We analyzed treatment (N+WSH, WSH, N, or control) randomly assigned in-utero, child pathogen and EED data at 14 months of age, and child HAZ at 28 months of age. We estimated the difference in mean child height for age Z-score (HAZ) under the treatment rule and the difference in stratified treatment effect (treatment effect difference) comparing children with high versus low pathogen/biomarker status while controlling for baseline covariates. RESULTS We analyzed data from 1,522 children who had a median HAZ of -1.56. We found that fecal myeloperoxidase (N+WSH treatment effect difference 0.0007 HAZ, WSH treatment effect difference 0.1032 HAZ, N treatment effect difference 0.0037 HAZ) and Campylobacter infection (N+WSH treatment effect difference 0.0011 HAZ, WSH difference 0.0119 HAZ, N difference 0.0255 HAZ) were associated with greater effect of all interventions on anthropometry. In other words, children with high myeloperoxidase or Campylobacter infection experienced a greater impact of the interventions on anthropometry. We found that a treatment rule that assigned the N+WSH (HAZ difference 0.23, 95% CI (0.05, 0.41)) and WSH (HAZ difference 0.17, 95% CI (0.04, 0.30)) interventions based on EED biomarkers and pathogens increased predicted child growth compared to the randomly allocated intervention. CONCLUSIONS These findings indicate that EED biomarkers and pathogen status, particularly Campylobacter and myeloperoxidase (a measure of gut inflammation), may be related to the impact of N+WSH, WSH, and N interventions on child linear growth.
Collapse
Affiliation(s)
- Zachary Butzin-Dozier
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Yunwen Ji
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Jeremy Coyle
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Ivana Malenica
- School of Public Health, University of California, Berkeley, California, United States of America
| | | | - Jessica Anne Grembi
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - James A. Platts-Mills
- School of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Eric R. Houpt
- School of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jay P. Graham
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Shahjahan Ali
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Md Ziaur Rahman
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, California, United States of America
| | - Mohammad Alauddin
- Wagner College, Staten Island, New York, New York, United States of America
| | - Syeda L. Famida
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Salma Akther
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Md. Saheen Hossen
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Palash Mutsuddi
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Abul K. Shoab
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Mahbubur Rahman
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Md. Ohedul Islam
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Rana Miah
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Mami Taniuchi
- School of Medicine, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jie Liu
- School of Public Health, Qingdao University, Qingdao, China
| | - Sarah T. Alauddin
- Wagner College, Staten Island, New York, New York, United States of America
| | - Christine P. Stewart
- Institute for Global Nutrition, University of California, Davis, California, United States of America
| | - Stephen P. Luby
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, United States of America
| | - John M. Colford Jr.
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Alan E. Hubbard
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Andrew N. Mertens
- School of Public Health, University of California, Berkeley, California, United States of America
| | - Audrie Lin
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, California, United States of America
| |
Collapse
|
7
|
Otiti MI, Were FA, Zaim S, Nabwera H, Kariuki S, Allen S. Probiotics and Synbiotics Administered to Young Infants: Perceptions and Acceptability Amongst Carers and Healthcare Workers in Western Kenya. Nutrients 2025; 17:495. [PMID: 39940358 PMCID: PMC11820114 DOI: 10.3390/nu17030495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES A contributory factor to childhood undernutrition is poor gut health occurring within the first 6-12 weeks of life despite exclusive breastfeeding. Pro/synbiotic administration may protect gut health. A qualitative study was conducted amongst mothers/carers and healthcare workers (HCWs) to explore their perceptions and the acceptability of pro/synbiotics administration in early life. METHODS This study was nested within a randomised, open, clinical trial of pro/synbiotics with 32 doses administered under supervision to infants between ages 0 and 5 months in western Kenya. Semi-structured interviews were conducted with 14 mothers/carers, 12 Peer Mothers and 7 healthcare workers (HCWs) selected by purposive critical and key informant sampling. Interviews were transcribed and analysed using a thematic coding framework. RESULTS The satisfaction with the pro/synbiotic administration was very high amongst all three groups. Commonly perceived benefits included protection from diseases, healthy growth of the infant and improved appetite. The main barriers were working mothers and other commitments making it difficult to stick to scheduled administration visits, adverse judgement and opinions in the community, and a lack of engagement from fathers. Insights were gained into different means of administering pro/synbiotics to young infants. Triangulation of the findings of the mothers/carers with HCWs showed that most identified motivations and challenges were similar. CONCLUSIONS Pro/synbiotic administration was well accepted by the mothers/carers and HCWs and generally perceived to have health benefits. The administration of pro/synbiotics by the mothers/carers themselves to their infants may be feasible and overcome logistical challenges. Greater efforts to sensitise and engage fathers and communities would likely be critical for a community-based program.
Collapse
Affiliation(s)
- Mary Iwaret Otiti
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK; (M.I.O.); (H.N.)
- Medical Research Institute (KEMRI) Centre for Global Health Research, Kisumu P.O. Box 1578 – 40100, Kenya; (F.A.W.); (S.K.)
| | - Florence Achieng Were
- Medical Research Institute (KEMRI) Centre for Global Health Research, Kisumu P.O. Box 1578 – 40100, Kenya; (F.A.W.); (S.K.)
| | - Sevim Zaim
- Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK;
| | - Helen Nabwera
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK; (M.I.O.); (H.N.)
| | - Simon Kariuki
- Medical Research Institute (KEMRI) Centre for Global Health Research, Kisumu P.O. Box 1578 – 40100, Kenya; (F.A.W.); (S.K.)
| | - Stephen Allen
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK; (M.I.O.); (H.N.)
| |
Collapse
|
8
|
Profir M, Enache RM, Roşu OA, Pavelescu LA, Creţoiu SM, Gaspar BS. Malnutrition and Its Influence on Gut sIgA-Microbiota Dynamics. Biomedicines 2025; 13:179. [PMID: 39857762 PMCID: PMC11762760 DOI: 10.3390/biomedicines13010179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
In the current era, malnutrition is seen as both undernutrition and overweight and obesity; both conditions are caused by nutrient deficiency or excess and improper use or imbalance in the intake of macro and micronutrients. Recent evidence suggests that malnutrition alters the intestinal microbiota, known as dysbiosis. Secretory immunoglobulin A (sIgA) plays an important role in maintaining and increasing beneficial intestinal microbiota populations and protecting against pathogenic species. Depletion of beneficial bacterial populations throughout life is also conditioned by malnutrition. This review aims to synthesize the evidence that establishes an interrelationship between diet, malnutrition, changes in the intestinal flora, and sIgA levels. Targeted nutritional therapies combined with prebiotic, probiotic, and postbiotic administration can restore the immune response in the intestine and the host's homeostasis.
Collapse
Affiliation(s)
- Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.); (L.A.P.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Robert Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania;
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.); (L.A.P.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Luciana Alexandra Pavelescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.); (L.A.P.)
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (M.P.); (O.A.R.); (L.A.P.)
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
9
|
Vresk L, Flanagan M, Daniel AI, Potani I, Bourdon C, Spiegel-Feld C, Thind MK, Farooqui A, Ling C, Miraglia E, Hu G, Wen B, Zlotkin S, James P, McGrath M, Bandsma RHJ. Micronutrient status in children aged 6-59 months with severe wasting and/or nutritional edema: implications for nutritional rehabilitation formulations. Nutr Rev 2025; 83:112-145. [PMID: 38350491 PMCID: PMC11632376 DOI: 10.1093/nutrit/nuad165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024] Open
Abstract
Undernutrition remains a global struggle and is associated with almost 45% of deaths in children younger than 5 years. Despite advances in management of severe wasting (though less so for nutritional edema), full and sustained recovery remains elusive. Children with severe wasting and/or nutritional edema (also commonly referred to as severe acute malnutrition and part of the umbrella term "severe malnutrition") continue to have a high mortality rate. This suggests a likely multifactorial etiology that may include micronutrient deficiency. Micronutrients are currently provided in therapeutic foods at levels based on expert opinion, with few supportive studies of high quality having been conducted. This narrative review looks at the knowledge base on micronutrient deficiencies in children aged 6-59 months who have severe wasting and/or nutritional edema, in addition to highlighting areas where further research is warranted (See "Future Directions" section).
Collapse
Affiliation(s)
- Laura Vresk
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mary Flanagan
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Allison I Daniel
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Isabel Potani
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Celine Bourdon
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Carolyn Spiegel-Feld
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mehakpreet K Thind
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amber Farooqui
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Catriona Ling
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Emiliano Miraglia
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Guanlan Hu
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Bijun Wen
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stanley Zlotkin
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Philip James
- Emergency Nutrition Network, Oxford, United Kingdom
| | | | - Robert H J Bandsma
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Mareș CR, Săsăran MO, Mărginean CO. Small Intestinal Bacterial Overgrowth and Childhood Malnutrition: A Comprehensive Review of Available Evidence. Nutrients 2024; 16:4319. [PMID: 39770940 PMCID: PMC11679674 DOI: 10.3390/nu16244319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The gut microbiome is essential for children's normal growth and development, with its formation aligning closely with key stages of growth. Factors like birth method, feeding practices, and antibiotic exposure significantly shape the composition and functionality of the infant gut microbiome. Small intestinal bacterial overgrowth (SIBO) involves an abnormal increase in bacteria within the small intestine. This overgrowth can interfere with digestion, impair nutrient absorption, and lead to both local and systemic inflammation, potentially contributing to malnutrition. In this review, we provide a comprehensive overview of the current understanding of the relationship between SIBO and malnutrition, with a particular focus on the pediatric population. SIBO seems to play an important role in nutrient malabsorption through the gut microbiome imbalance, local inflammation, and disruption of the mucosal intestinal barrier. Additionally, SIBO is more prevalent in digestive disorders linked to malabsorption and malnutrition. Different therapeutic strategies for addressing malnutrition-related SIBO have been proposed. While antibiotics are the primary treatment for SIBO, their effectiveness in promoting weight gain among malnourished children remains uncertain. Hence, future research directed at the impact of microbiome imbalance on nutrient intake and absorption could bring to light new strategies for the effective prevention and treatment of malnutrition.
Collapse
Affiliation(s)
- Cristina Roxana Mareș
- Department of Pediatrics 1, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania; (C.R.M.); (C.O.M.)
| | - Maria Oana Săsăran
- Department of Pediatrics 3, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania
| | - Cristina Oana Mărginean
- Department of Pediatrics 1, “George Emil Palade” University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mureș, Romania; (C.R.M.); (C.O.M.)
| |
Collapse
|
11
|
Bildstein T, Charbit-Henrion F, Azabdaftari A, Cerf-Bensussan N, Uhlig HH. Cellular and molecular basis of proximal small intestine disorders. Nat Rev Gastroenterol Hepatol 2024; 21:687-709. [PMID: 39117867 DOI: 10.1038/s41575-024-00962-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
The proximal part of the small intestine, including duodenum and jejunum, is not only dedicated to nutrient digestion and absorption but is also a highly regulated immune site exposed to environmental factors. Host-protective responses against pathogens and tolerance to food antigens are essential functions in the small intestine. The cellular ecology and molecular pathways to maintain those functions are complex. Maladaptation is highlighted by common immune-mediated diseases such as coeliac disease, environmental enteric dysfunction or duodenal Crohn's disease. An expanding spectrum of more than 100 rare monogenic disorders inform on causative molecular mechanisms of nutrient absorption, epithelial homeostasis and barrier function, as well as inflammatory immune responses and immune regulation. Here, after summarizing the architectural and cellular traits that underlie the functions of the proximal intestine, we discuss how the integration of tissue immunopathology and molecular mechanisms can contribute towards our understanding of disease and guide diagnosis. We propose an integrated mechanism-based taxonomy and discuss the latest experimental approaches to gain new mechanistic insight into these disorders with large disease burden worldwide as well as implications for therapeutic interventions.
Collapse
Affiliation(s)
- Tania Bildstein
- Great Ormond Street Hospital for Children, Department of Paediatric Gastroenterology, London, UK
| | - Fabienne Charbit-Henrion
- Department of Genomic Medicine for Rare Diseases, Necker-Enfants Malades Hospital, APHP, University of Paris-Cité, Paris, France
- INSERM UMR1163, Intestinal Immunity, Institut Imagine, Paris, France
| | - Aline Azabdaftari
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Oxford, UK
| | | | - Holm H Uhlig
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
- National Institute for Health and Care Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
12
|
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
|
13
|
Jamil Z, VanBuskirk K, Mweetwa M, Mouksassi S, Smith G, Ahmed T, Chandwe K, Denno DM, Fahim SM, Kelly P, Mahfuz M, Mallawaarachchi I, Marie C, Moore SR, Petri WA, Ali SA. Anthropometry relationship with duodenal histologic features of children with environmental enteric dysfunction: a multicenter cross-sectional study. Am J Clin Nutr 2024; 120 Suppl 1:S65-S72. [PMID: 39300664 DOI: 10.1016/j.ajcnut.2024.02.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/11/2024] [Accepted: 02/22/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is a precursor of growth faltering in children living in impoverished conditions who are frequently exposed to environmental toxins and enteropathogens, leading to small bowel inflammatory, malabsorptive, and permeability derangements and low-grade chronic systemic inflammation. OBJECTIVES We explored the association between anthropometrics and duodenal histologic features of EED among children from 3 lower middle-income country centers. METHODS In this cross-sectional study, Pakistani children (n = 63) with wasting, Bangladesh children (n = 116) with stunting or at risk for stunting (height-for-age Z score [HAZ] <-1 but ≥-2), and Zambian children (n = 108) with wasting or stunting received nutritional intervention. Children with anthropometric status refractory to intervention underwent endoscopy. Linear regression models included anthropometric around endoscopy, scores of histology parameters, and a global index score of EED-the total score percent-5 (TSP-5). Multivariable models were adjusted for center, age, sex, and histology slide quality. RESULTS Intersite variation was observed while exploring the association between anthropometrics and the TSP-5; for example, Pakistani children had the worst HAZ, yet their median TSP-5 score was lower than that of the other 2 centers. Even within each site, no overall pattern of higher TSP-5 score was observed with worsening HAZ. During univariate analysis, TSP-5 (coefficient: 0.01; 95% confidence interval [CI]: 0, 0.02), goblet cell depletion (coefficient: 0.22; 95% CI: 0.06, 0.37), and Paneth cell depletion (coefficient: 0.14; 95% CI: 0.01, 0.27) were associated with HAZ scores; however, they lost statistical significance in the multivariable models, with study center most strongly confounding the relationships seen in univariate models between anthropometry and histology. CONCLUSIONS This study contributes a crucial negative finding that duodenal morphological features did not associate with anthropometric phenotypes; hence, anthropometric measurements may not be a suitable outcome measure for use in EED trials. Trial outcomes may need to be defined by combining the functional and structural elements of the gut to monitor EED.
Collapse
Affiliation(s)
- Zehra Jamil
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Kelley VanBuskirk
- Department of Global Health, University of Washington School of Public Health, Seattle, WA, United States
| | - Monica Mweetwa
- Tropical Gastroenterology and Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | | | | | - Tahmeed Ahmed
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Kanta Chandwe
- Tropical Gastroenterology and Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Donna M Denno
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - S Mohammad Fahim
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Paul Kelly
- Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Indika Mallawaarachchi
- Department of Public Health Sciences, 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
| | - Sean R Moore
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - William A Petri
- Department University of Virginia, Charlottesville, VA, United States
| | - S Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan.
| |
Collapse
|
14
|
VanBuskirk K, Mweetwa M, Kolterman T, Raghavan S, Ahmed T, Ali SA, Nahar Begum SK, Besa E, Denno DM, Jamil Z, Kelly P, Mahfuz M, Moore SR, Mouksassi S, Petri WA, Tarr PI, Sullivan PB, Moskaluk CA. Multiplexed immunohistochemical evaluation of small bowel inflammatory and epithelial parameters in environmental enteric dysfunction. Am J Clin Nutr 2024; 120 Suppl 1:S31-S40. [PMID: 39300661 DOI: 10.1016/j.ajcnut.2024.02.033] [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: 09/09/2023] [Revised: 01/15/2024] [Accepted: 02/22/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is characterized by reduced absorptive capacity and barrier function of the small intestine, leading to poor ponderal and linear childhood growth. OBJECTIVES To further define gene expression patterns that are associated with EED to uncover new pathophysiology of this disorder. METHODS Duodenal biopsies from cohorts of children with EED from Bangladesh, Pakistan and Zambia were analyzed by immunohistochemistry (IHC) to interrogate gene products that distinguished differentiation and various biochemical pathways in immune and epithelial cells, some identified by prior bulk RNA sequence analyses. Immunohistochemical staining was digitally quantified from scanned images and compared to cohorts of North American children with celiac disease (gluten-sensitive enteropathy) or with no known enteric disease and no pathologic abnormality (NPA) detected in their clinical biopsies. RESULTS After multivariable statistical analysis, we identified statistically significant (P < 0.05, 2-tailed t-test) elevated signals representing cluster of differentiation 45 (80%; 95% confidence interval [CI]: 24%, 127%), lipocalin 2 (659%; 95% CI: 198%, 1838%), and regenerating family 1 beta (221%; 95% CI: 47%, 600%) and lower signals corresponding to granzyme B (-74%; 95% CI: -82%, -62%), and sucrase isomaltase (-58%; 95% CI: -75%, -29%) in EED biopsies compared with NPA biopsies. Computerized algorithms also detected statistically significant elevation in intraepithelial lymphocytes (49%; 95% CI: 9%, 105%) and proliferation of leukocytes (267%; 95% CI: 92%, 601%) in EED biopsies compared with NPA biopsies. CONCLUSIONS Our results support a model of chronic epithelial stress that decreases epithelial differentiation and absorptive function. The close association of several IHC parameters with manual histologic scoring suggests that automated digital quantification of IHC panels complements traditional histomorphologic assessment in EED.
Collapse
Affiliation(s)
- Kelley VanBuskirk
- Department of Global Health, University of Washington School of Public Health, Seattle, WA, United States
| | - Monica Mweetwa
- Tropical Gastroenterology and Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Tad Kolterman
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Shyam Raghavan
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Tahmeed Ahmed
- 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
| | | | - Ellen Besa
- Tropical Gastroenterology and Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Donna M Denno
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Zehra Jamil
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Paul Kelly
- Blizard Institute, Barts & the London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Sean R Moore
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | | - William A Petri
- 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
| | - Christopher A Moskaluk
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States.
| |
Collapse
|
15
|
Wasan Y, Baxter JAB, Spiegel-Feld C, Begum K, Rizvi A, Iqbal J, Hulst J, Bandsma R, Suleman S, Soofi S, Parkinson J, Bhutta ZA. Elucidating the dynamics and impact of the gut microbiome on maternal nutritional status during pregnancy, effect on pregnancy outcomes and infant health in rural Pakistan: study protocol for a prospective, longitudinal observational study. BMJ Open 2024; 14:e081629. [PMID: 39134435 PMCID: PMC11331926 DOI: 10.1136/bmjopen-2023-081629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 07/19/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Undernutrition during pregnancy is linked to adverse pregnancy and birth outcomes and has downstream effects on the growth and development of children. The gut microbiome has a profound influence on the nutritional status of the host. This phenomenon is understudied in settings with a high prevalence of undernutrition, and further investigation is warranted to better understand such interactions. METHODS AND ANALYSIS This is a prospective, longitudinal observational study to investigate the relationship between prokaryotic and eukaryotic microbes in the gut and their association with maternal body mass index (BMI), gestational weight gain, and birth and infant outcomes among young mothers (17-24 years) in Matiari District, Pakistan. We aim to enrol 400 pregnant women with low and normal BMIs at the time of recruitment (<16 weeks of gestation). To determine the weight gain during pregnancy, maternal weight is measured in the first and third trimesters. Gut microbiome dynamics (bacterial and eukaryotic) will be assessed using 16S and 18S rDNA surveys applied to the maternal stool samples. Birth outcomes include birth weight, small for gestational age, large for gestational age, preterm birth and mortality. Infant growth and nutritional parameters include WHO z-scores for weight, length and head circumference at birth through infancy. To determine the impact of the maternal microbiome, including exposure to pathogens and parasites on the development of the infant microbiome, we will analyse maternal and infant microbiome composition, micronutrients in serum using metallomics (eg, zinc, magnesium and selenium) and macronutrients in the stool. Metatranscriptomics metabolomics and markers of inflammation will be selectively deployed on stool samples to see the variations in dietary intake and maternal nutritional status. We will also use animal models to explore the bacterial and eukaryotic components of the microbiome. ETHICS AND DISSEMINATION The study is approved by the National Bioethics Committee (NBC) in Pakistan, the Ethics Review Committee (ERC) at Aga Khan University and the Research Ethics Board (REB) at the Hospital for Sick Children, and findings will be published in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT05108675.
Collapse
Affiliation(s)
- Yaqub Wasan
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Sindh, Pakistan
| | - Jo-Anna B Baxter
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Carolyn Spiegel-Feld
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kehkashan Begum
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Sindh, Pakistan
| | - Arjumand Rizvi
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Sindh, Pakistan
| | - Junaid Iqbal
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Sindh, Pakistan
| | - Jessie Hulst
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences and Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Robert Bandsma
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Nutritional Sciences and Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shazeen Suleman
- Department of Pediatrics, and Global Health Faculty Fellow, Centre for Innovation in Global Health, Stanford University, Stanford, California, USA
| | - Sajid Soofi
- Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Sindh, Pakistan
| | - John Parkinson
- Department of Pediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Zulfiqar Ahmed Bhutta
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Institute for Global Health and Development and Centre of Excellence in Women and Child Health, The Aga Khan University, Karachi, Sindh, Pakistan
- Department of Nutritional Sciences and Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Lui JC, Palmer AC, Christian P. Nutrition, Other Environmental Influences, and Genetics in the Determination of Human Stature. Annu Rev Nutr 2024; 44:205-229. [PMID: 38759081 DOI: 10.1146/annurev-nutr-061121-091112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
Linear growth during three distinct stages of life determines attained stature in adulthood: namely, in utero, early postnatal life, and puberty and the adolescent period. Individual host factors, genetics, and the environment, including nutrition, influence attained human stature. Each period of physical growth has its specific biological and environmental considerations. Recent epidemiologic investigations reveal a strong influence of prenatal factors on linear size at birth that in turn influence the postnatal growth trajectory. Although average population height changes have been documented in high-income regions, stature as a complex human trait is not well understood or easily modified. This review summarizes the biology of linear growth and its major drivers, including nutrition from a life-course perspective, the genetics of programmed growth patterns or height, and gene-environment interactions that determine human stature in toto over the life span. Implications for public health interventions and knowledge gaps are discussed.
Collapse
Affiliation(s)
- Julian C Lui
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Amanda C Palmer
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| | - Parul Christian
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| |
Collapse
|
17
|
Vray M, Tondeur L, Hedible BG, Randremanana RV, Manirakiza A, Lazoumar RH, Platen CV, Vargas A, Briend A, Jambou R. Three-arm clinical trial of improved flour targeting intestinal microbiota (MALINEA). MATERNAL & CHILD NUTRITION 2024; 20:e13649. [PMID: 38599819 PMCID: PMC11168351 DOI: 10.1111/mcn.13649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
The main objective of this project was to compare in the field conditions two strategies of re-nutrition of children with moderate acute malnutrition (MAM) aged from 6 to 24 months, targeting the microbiota in comparison with a standard regimen. A three-arm, open-label, pragmatic randomised trial was conducted in four countries (Niger, CAR, Senegal and Madagascar). Children received for 12 weeks either fortified blended flour (FBF control) = arm 1, or FBF + azithromycin (oral suspension of 20 mg/kg/day daily given with a syringe) for the first 3 days at inclusion = arm 2 or mix FBF with inulin/fructo-oligosaccharides (6 g/day if age ≥12 months and 4 g if age <12 months) = arm 3. For each arm, children aged from 6 to 11 months received 100 g x 2 per day of flours and those aged from 12 to 24 months received 100 g × 3 per day of FBF. The primary endpoint was nutritional recovery, defined by reaching a weight-for-height z-score (WHZ) ≥ -1.5 within 12 weeks. Overall, 881 children were randomised (297, 290 and 294 in arm 1, arm 2 and arm 3, respectively). Three hundred and forty-four children were males (39%) and median/mean age were 14.6/14.4 months (SD = 4.9, IQR = 10.5-18.4). At inclusion, the three arms were comparable for all criteria, but differences were observed between countries. Overall, 44% (390/881) of the children recovered at week 12 from MAM, with no significant difference between the three arms (41.4%, 45.5% and 45.9%, in arm 1, arm 2 and arm 3, respectively, p = 0.47). This study did not support the true advantages of adding a prebiotic or antibiotic to flour. When using a threshold of WHZ ≥ -2 as an exploratory endpoint, significant differences were observed between the three arms, with higher success rates in arms with antibiotics or prebiotics compared to the control arm (66.9%, 66.0% and 55.2%, respectively, p = 0.005).
Collapse
Affiliation(s)
- Muriel Vray
- Emerging Diseases Epidemiology Unit, Institut de PasteurUniversité Paris‐CitéParisFrance
| | - Laura Tondeur
- Emerging Diseases Epidemiology Unit, Institut de PasteurUniversité Paris‐CitéParisFrance
| | | | | | - Alexandre Manirakiza
- Epidemiology and Clinical Research UnitInstitut Pasteur de BanguiBanguiCentral African Republic
| | | | | | - Antonio Vargas
- Nutrition and Health Unit, Action Against HungerMadridSpain
| | - André Briend
- Department of Nutrition, Exercise and Sports, Faculty of ScienceUniversity of CopenhagenFrederiksbergDenmark
- Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health TechnologyTampere University and Tampere University HospitalTampereFinland
| | - Ronan Jambou
- Direction Scientifique, Centre de Recherche Médicale et Sanitaire (CERMES)NiameyNiger
| |
Collapse
|
18
|
Sahiledengle B, Petrucka P, Desta F, Sintayehu Y, Mesfin T, Mwanri L. Childhood undernutrition mediates the relationship between open defecation with anemia among Ethiopian children: a nationally representative cross-sectional study. BMC Public Health 2024; 24:1484. [PMID: 38831296 PMCID: PMC11145842 DOI: 10.1186/s12889-024-18931-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Poor sanitation and/or open defecation are a significant public health problem in Ethiopia, where access to improved sanitation facilities is still limited. There is a growing body of literature about the effect of open defecation on children's linear growth failure. However, very few studies about the effects of open defecation on child anemia exist. In this study, we examine whether childhood undernutrition (i.e. stunting, wasting, and underweight) mediates the relationship between open defecation and childhood anemia in children aged 6-59 months in Ethiopia. METHODS We used pooled Ethiopia Demographic and Health Survey data (2005-2016) comprising 21,918 (weighted data) children aged 6-59 months. Anemia was defined as an altitude-adjusted hemoglobin (Hb) level of less than 11 g/deciliter (g/dl) for children under 5 years. Childhood undernutrition was assessed using height-for-age Z-scores (HAZ), weight-for-age Z-scores (WAZ), and weight-for-height Z-scores (WHZ) for stunting, wasting, and underweight respectively. Mediation effects were calculated using the bootstrap and the indirect effect was considered significant when the 95% bootstrap confidence intervals (95% CI) did not contain zero. Moreover, separate multilevel regression analyses were used to explore the statistical association between open defecation and child anemia, after adjusting for potential confounders. RESULTS Our analysis revealed that nearly half (49.6%) of children aged 6 to 59 months were anemic, 46.8% were stunted, 9.9% were wasted, and 29.5% were underweight. Additionally, 45.1% of children belonged to households that practiced open defecation (OD). Open defecation was associated with anemia (AOR: 1.28; 95% CI: 1.18-1.39) and it positively predicted anemia with direct effect of β = 0.233, p < 0.001. Childhood undernutrition showed a partial mediating role in the relationship between OD and anemia. Analyzing the indirect effects, results revealed that child undernutrition significantly mediated the relationship between open defecation and anemia (stunting (βindirect = 0.014, p < 0.001), wasting (βindirect = 0.009, p = 0.002), and underweight (βindirect = 0.012, p < 0.001)). When the mediating role of child undernutrition was accounted for, open defecation had a positive impact on anemia with a total effect of βtotal = 0.285, p < 0.001. CONCLUSION Open defecation showed a significant direct effect on anemia. Child undernutrition remarkably mediated the relationship between OD and anemia that further magnified the effect. This finding has an important programmatic implication calling for strengthened, accelerated and large-scale implementation of strategies to end open defecation and achieve universal access to sanitation in Ethiopia.
Collapse
Affiliation(s)
- Biniyam Sahiledengle
- Department of Public Health, Madda Walabu University Goba Referral Hospital, Bale-Goba, Ethiopia.
| | - Pammla Petrucka
- College of Nursing, University of Saskatchewan, Saskatoon, Canada
| | - Fikreab Desta
- Department of Public Health, Madda Walabu University Goba Referral Hospital, Bale-Goba, Ethiopia
| | - Yordanos Sintayehu
- Department of Public Health, Madda Walabu University Goba Referral Hospital, Bale-Goba, Ethiopia
| | - Telila Mesfin
- Department of Medicine, Madda Walabu University Goba Referral Hospital, Bale-Goba, Ethiopia
| | - Lillian Mwanri
- Equity and Human Flourishing, Research Centre for Public Health Research, Torrens University Australia, Adelaide Campus, Adelaide, SA, 5000, Australia
| |
Collapse
|
19
|
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 13 C-sucrose breath test. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.01.24306704. [PMID: 38746107 PMCID: PMC11092706 DOI: 10.1101/2024.05.01.24306704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Environmental enteric dysfunction (EED) is a syndrome characterized by epithelial damage including blunting of the small intestinal villi and altered digestive and absorptive capacity which may negatively impact linear growth in children. The 13 C-sucrose breath test ( 13 C-SBT) has been proposed to estimate sucrase-isomaltase (SIM) activity, which is thought to be reduced in EED. We previously showed how various summary measures of the 13 C-SBT breath curve reflect SIM inhibition. However, it is uncertain how the performance of these classifiers is affected by test duration. Methods We leveraged SBT data from a cross-over study in 16 adults who received 0, 100, and 750 mg of Reducose, a natural SIM inhibitor. We evaluated the performance of a pharmacokinetic-model-based classifier, ρ , and three empirical classifiers (cumulative percent dose recovered at 90 minutes (cPDR90), time to 50% dose recovered, and time to peak dose recovery rate), as a function of test duration using receiver operating characteristic curves. We also assessed the sensitivity, specificity, and accuracy of consensus classifiers. Results Test durations of less than 2 hours generally failed to accurately predict later breath curve dynamics. The cPDR90 classifier had the highest area-under-the-curve and, by design, was robust to shorter test durations. For detecting mild SIM inhibition, ρ had a higher sensitivity. Conclusions We recommend SBT tests run for at least a 2-hour 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 CO 2 production rate. More research is needed to assess these classifiers in target populations.
Collapse
|
20
|
Lowe C, Sarma H, Gray D, Kelly M. Perspective: Connecting the dots between domestic livestock ownership and child linear growth in low- and middle-income countries. MATERNAL & CHILD NUTRITION 2024; 20:e13618. [PMID: 38192051 PMCID: PMC10981488 DOI: 10.1111/mcn.13618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 01/10/2024]
Abstract
Child stunting due to linear growth faltering remains a pervasive issue in low- and middle-income countries. Two schools of thought have existed pertaining to the role of domestic livestock ownership (DLO) in child linear growth. On one hand, it is argued that DLO leads to greater income and financial security, resulting in better child-raising conditions, including greater animal-source food (ASF) consumption, having protective effects towards child stunting. On the other hand, researchers argue that DLO contributes to faecal contamination and transmission of zoonotic enteric infections from animals to children, thus having destructive effects on child growth. Reviews of this association have revealed ambiguous findings. In this perspective, we argue that measuring the association between exposures to domesticated animals and child stunting is difficult and the ambiguous associations revealed are a result of confounding and differences in the management of DLO. We also argue that the increasingly prominent area of research of environmental enteric dysfunction, a sub-clinical condition of the small intestine thought to be due to frequent faecal pathogen exposure and associated with stunting, will be a useful tool to measure the potential destructive effects of DLO on child growth. We present our argument and identify challenges and considerations and directions for future research.
Collapse
Affiliation(s)
- Callum Lowe
- Department of Applied Epidemiology, National Centre for Epidemiology and Population Health, College of Health and MedicineAustralian National UniversityActonAustralian Capital TerritoryAustralia
| | - Haribondhu Sarma
- Department of Applied Epidemiology, National Centre for Epidemiology and Population Health, College of Health and MedicineAustralian National UniversityActonAustralian Capital TerritoryAustralia
| | - Darren Gray
- Population Health ProgramQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Matthew Kelly
- Department of Applied Epidemiology, National Centre for Epidemiology and Population Health, College of Health and MedicineAustralian National UniversityActonAustralian Capital TerritoryAustralia
| |
Collapse
|
21
|
Butzin-Dozier Z, Ji Y, Coyle J, Malenica I, McQuade ETR, Grembi JA, Platts-Mills JA, Houpt ER, Graham JP, Ali S, Rahman MZ, Alauddin M, Famida SL, Akther S, Hossen MS, Mutsuddi P, Shoab AK, Rahman M, Islam MO, Miah R, Taniuchi M, Liu J, Alauddin S, Stewart CP, Luby SP, Colford JM, Hubbard AE, Mertens AN, Lin A. Treatment Heterogeneity of Water, Sanitation, Hygiene, and Nutrition Interventions on Child Growth by Environmental Enteric Dysfunction and Pathogen Status for Young Children in Bangladesh. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.21.24304684. [PMID: 38585931 PMCID: PMC10996736 DOI: 10.1101/2024.03.21.24304684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Background Water, sanitation, hygiene (WSH), nutrition (N), and combined (N+WSH) interventions are often implemented by global health organizations, but WSH interventions may insufficiently reduce pathogen exposure, and nutrition interventions may be modified by environmental enteric dysfunction (EED), a condition of increased intestinal permeability and inflammation. This study investigated the heterogeneity of these treatments' effects based on individual pathogen and EED biomarker status with respect to child linear growth. Methods We applied cross-validated targeted maximum likelihood estimation and super learner ensemble machine learning to assess the conditional treatment effects in subgroups defined by biomarker and pathogen status. We analyzed treatment (N+WSH, WSH, N, or control) randomly assigned in-utero, child pathogen and EED data at 14 months of age, and child LAZ at 28 months of age. We estimated the difference in mean child length for age Z-score (LAZ) under the treatment rule and the difference in stratified treatment effect (treatment effect difference) comparing children with high versus low pathogen/biomarker status while controlling for baseline covariates. Results We analyzed data from 1,522 children, who had median LAZ of -1.56. We found that myeloperoxidase (N+WSH treatment effect difference 0.0007 LAZ, WSH treatment effect difference 0.1032 LAZ, N treatment effect difference 0.0037 LAZ) and Campylobacter infection (N+WSH treatment effect difference 0.0011 LAZ, WSH difference 0.0119 LAZ, N difference 0.0255 LAZ) were associated with greater effect of all interventions on growth. In other words, children with high myeloperoxidase or Campylobacter infection experienced a greater impact of the interventions on growth. We found that a treatment rule that assigned the N+WSH (LAZ difference 0.23, 95% CI (0.05, 0.41)) and WSH (LAZ difference 0.17, 95% CI (0.04, 0.30)) interventions based on EED biomarkers and pathogens increased predicted child growth compared to the randomly allocated intervention. Conclusions These findings indicate that EED biomarker and pathogen status, particularly Campylobacter and myeloperoxidase (a measure of gut inflammation), may be related to impact of N+WSH, WSH, and N interventions on child linear growth.
Collapse
Affiliation(s)
| | - Yunwen Ji
- School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | - Jeremy Coyle
- School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | - Ivana Malenica
- School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | | | - Jessica Anne Grembi
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA USA
| | | | - Eric R. Houpt
- School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jay P. Graham
- School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | - Shahjahan Ali
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md Ziaur Rahman
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mohammad Alauddin
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Syeda L. Famida
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Salma Akther
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md. Saheen Hossen
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Palash Mutsuddi
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Abul K. Shoab
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mahbubur Rahman
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md. Ohedul Islam
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Rana Miah
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mami Taniuchi
- School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jie Liu
- School of Public Health, Qingdao University, Qingdao, China
| | | | | | - Stephen P. Luby
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA USA
| | - John M. Colford
- School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | - Alan E. Hubbard
- School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | - Andrew N. Mertens
- School of Public Health, University of California, Berkeley, Berkeley, CA USA
| | - Audrie Lin
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, CA USA
| |
Collapse
|
22
|
Mutumba R, Pesu H, Mbabazi J, Greibe E, Nexo E, Olsen MF, Briend A, Mølgaard C, Michaelsen KF, Ritz C, Filteau S, Mupere E, Friis H, Grenov B. Effect of lipid-based nutrient supplements on micronutrient status and hemoglobin among children with stunting: secondary analysis of a randomized controlled trial in Uganda. Am J Clin Nutr 2024; 119:829-837. [PMID: 38278366 DOI: 10.1016/j.ajcnut.2024.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/08/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Micronutrient deficiencies and anemia are widespread among children with stunting. OBJECTIVES We assessed the effects of lipid-based nutrient supplements (LNS) containing milk protein (MP) and/or whey permeate (WP) on micronutrient status and hemoglobin (Hb) among children with stunting. METHODS This was a secondary analysis of a randomized controlled trial. Children aged 12-59 mo with stunting were randomly assigned to LNS (100 g/d) with milk or soy protein and WP or maltodextrin for 12 wk, or no supplement. Hb, serum ferritin (S-FE), serum soluble transferrin receptor (S-TfR), plasma cobalamin (P-Cob), plasma methylmalonic acid (P-MMA), plasma folate (P-Fol), and serum retinol-binding protein (S-RBP) were measured at inclusion and at 12 wk. Data were analyzed using linear and logistic mixed-effects models. RESULTS Among 750 children, with mean age ± SD of 32 ± 11.7 mo, 45% (n = 338) were female and 98% (n = 736) completed follow-up. LNS, compared with no supplementation, resulted in 43% [95% confidence interval (CI): 28, 60] greater increase in S-FE corrected for inflammation (S-FEci), 2.4 (95% CI: 1.2, 3.5) mg/L greater decline in S-TfR, 138 (95% CI: 111, 164) pmol/L greater increase in P-Cob, 33% (95% CI: 27, 39) reduction in P-MMA, and 8.5 (95% CI: 6.6, 10.3) nmol/L greater increase in P-Fol. There was no effect of LNS on S-RBP. Lactation modified the effect of LNS on markers of cobalamin status, reflecting improved status among nonbreastfed and no effects among breastfed children. LNS increased Hb by 3.8 (95% CI: 1.7, 6.0) g/L and reduced the odds of anemia by 55% (odds ratio: 0.45, 95% CI: 0.29, 0.70). MP compared with soy protein increased S-FEci by 14% (95% CI: 3, 26). CONCLUSIONS LNS supplementation increases Hb and improves iron, cobalamin, and folate status, but not vitamin A status among children with stunting. LNS should be considered for children with stunting. This trial was registered at ISRCTN as 13093195.
Collapse
Affiliation(s)
- Rolland Mutumba
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Hannah Pesu
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Joseph Mbabazi
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Eva Greibe
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Ebba Nexo
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Mette F Olsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - André Briend
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark; Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Christian Mølgaard
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kim F Michaelsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Ritz
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Suzanne Filteau
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ezekiel Mupere
- Department of Pediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Henrik Friis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Benedikte Grenov
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
23
|
Brouwer AF, Lee GO, Van Wyk H, Schillinger RJ, Edwards CA, Morrison DJ. A Model-Based 13C-Sucrose Breath Test Diagnostic for Gut Function Disorders Characterized by a Loss of Sucrase-Isomaltase Enzymatic Activity. J Nutr 2024; 154:815-825. [PMID: 37995914 PMCID: PMC10942859 DOI: 10.1016/j.tjnut.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) causes malnutrition in children in low-resource settings. Stable-isotope breath tests have been proposed as noninvasive tests of altered nutrient metabolism and absorption in EED, but uncertainty over interpreting the breath curves has limited their use. The activity of sucrose-isomaltase, the glucosidase enzyme responsible for sucrose hydrolysis, may be reduced in EED. We previously developed a mechanistic model describing the dynamics of the 13C-sucrose breath test (13C-SBT) as a function of underlying metabolic processes. OBJECTIVES This study aimed to determine which breath test curve dynamics are associated with sucrose hydrolysis and with the transport and metabolism of the fructose and glucose moieties and to propose and evaluate a model-based diagnostic for the loss of activity of sucrase-isomaltase. METHODS We applied the mechanistic model to 2 sets of exploratory 13C-SBT experiments in healthy adult participants. First, 19 participants received differently labeled sucrose tracers (U-13C fructose, U-13C glucose, and U-13C sucrose) in a crossover study. Second, 16 participants received a sucrose tracer accompanied by 0, 100, and 750 mg of Reducose, a sucrase-isomaltase inhibitor. We evaluated a model-based diagnostic distinguishing between inhibitor concentrations using receiver operator curves, comparing with conventional statistics. RESULTS Sucrose hydrolysis and the transport and metabolism of the fructose and glucose moieties were reflected in the same mechanistic process. The model distinguishes these processes from the fraction of tracer exhaled and an exponential metabolic process. The model-based diagnostic performed as well as the conventional summary statistics in distinguishing between no and low inhibition [area under the curve (AUC): 0.77 vs. 0.66-0.79] and for low vs. high inhibition (AUC 0.92 vs. 0.91-0.99). CONCLUSIONS Current summary approaches to interpreting 13C breath test curves may be limited to identifying only gross gut dysfunction. A mechanistic model-based approach improved interpretation of breath test curves characterizing sucrose metabolism.
Collapse
Affiliation(s)
- Andrew F Brouwer
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA.
| | - Gwenyth O Lee
- Rutgers Global Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Hannah Van Wyk
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Robert J Schillinger
- Scottish Universities Environmental Research Centre, University of Glasgow, East Kilbride, United Kingdom; School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom
| | - Christine A Edwards
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom
| | - Douglas J Morrison
- Scottish Universities Environmental Research Centre, University of Glasgow, East Kilbride, United Kingdom
| |
Collapse
|
24
|
Pesu H, Mbabazi J, Mutumba R, Savolainen O, Olsen MF, Mølgaard C, Michaelsen KF, Ritz C, Filteau S, Briend A, Mupere E, Friis H, Grenov B. Correlates of Plasma Citrulline, a Potential Marker of Enterocyte Mass, among Children with Stunting: A Cross-Sectional Study in Uganda. J Nutr 2024; 154:765-776. [PMID: 38135004 DOI: 10.1016/j.tjnut.2023.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is associated with stunting. Citrulline, produced in mature enterocytes, may be a valuable biomarker of small intestinal enterocyte mass in the context of EED. OBJECTIVES We aimed to explore the correlates of plasma citrulline (p-cit) in children with stunting. METHODS In a cross-sectional study using baseline data from the community-based MAGNUS (milk affecting growth, cognition and the gut in child stunting) trial (ISRCTN13093195), we explored potential correlates of p-cit in Ugandan children with stunting aged 12-59 mo. Using linear regression in univariate and multivariate models, we explored associations with socioeconomics, diet, micronutrient status, and water, sanitation, and hygiene characteristics. The influence of covariates age, fasting, and systemic inflammation were also explored. RESULTS In 750 children, the mean ± standard deviation age was 32.0 ± 11.7 mo, and height-for-age z-score was -3.02 ± 0.74. P-cit, available for 730 children, differed according to time fasted and was 20.7 ± 8.9, 22.3 ± 10.6 and 24.2 ± 13.1 μmol/L if fasted <2, 2-5 and >5 h, respectively. Positive correlates of p-cit were age [0.07; 95% confidence interval (CI): 0.001, 0.15 μmol/L] and log10 serum insulin-like growth factor-1 (8.88; 95% CI: 5.09, 12.67 μmol/L). With adjustment for systemic inflammation, the association with serum insulin-like growth factor-1 reduced (4.98; 95% CI: 0.94, 9.03 μmol/L). Negative correlates of p-cit included food insecurity, wet season (-3.12; 95% CI: -4.97, -1.26 μmol/L), serum C-reactive protein (-0.15; 95% CI: -0.20, -0.10 μmol/L), serum α1-acid glycoprotein (-5.34; 95% CI: -6.98, -3.70 μmol/L) and anemia (-1.95; 95% CI: -3.72, -0.18 μmol/L). Among the negatively correlated water, sanitation, and hygiene characteristics was lack of soap for handwashing (-2.53; 95% CI: -4.82, -0.25 μmol/L). Many associations attenuated with adjustment for inflammation. CONCLUSIONS Many of the correlates of p-cit are characteristic of populations with a high EED prevalence. Systemic inflammation is strongly associated with p-cit and is implicated in EED and stunting. Adjustment for systemic inflammation attenuates many associations, reflecting either confounding, mediation, or both. This study highlights the complex interplay between p-cit and systemic inflammation.
Collapse
Affiliation(s)
- Hannah Pesu
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Joseph Mbabazi
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Rolland Mutumba
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Otto Savolainen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden; Faculty of Health Sciences, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Mette F Olsen
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kim F Michaelsen
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Ritz
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Suzanne Filteau
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - André Briend
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark; Tampere Centre for Child Health Research, Tampere University, Tampere, Finland
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Henrik Friis
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Benedikte Grenov
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
25
|
Sahiledengle B, Mwanri L, Agho KE. Household environment associated with anaemia among children aged 6-59 months in Ethiopia: a multilevel analysis of Ethiopia demographic and health survey (2005-2016). BMC Public Health 2024; 24:315. [PMID: 38287295 PMCID: PMC10823679 DOI: 10.1186/s12889-024-17780-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 01/15/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Anaemia continues to be a major public health challenge globally, including in Ethiopia. Previous studies have suggested that improved household environmental conditions may reduce anaemia prevalence; however, population-level evidence of this link is lacking in low-income countries. Therefore, this study aimed to examine the association between environmental factors and childhood anaemia in Ethiopia. METHODS In this study, we conducted an analysis of the data from the Ethiopian Demographic and Health Survey (EDHS), a nationally representative population-based survey conducted in Ethiopia between 2005 and 2016. The study included a total of 21,918 children aged 6-59 months. Children were considered anemic if their hemoglobin (Hb) concentration was less than 11.0 g/dl. To examine the association between environmental factors and anemia, we used multilevel mixed-effect models. These models allowed us to control for various confounding factors including: child, maternal, household and community-level variables. The study findings have been reported as adjusted odds ratios (AORs) along with 95% confidence intervals (CIs) at a significance level of p < 0.05. RESULTS The study found the overall prevalence of childhood anaemia to be 49.3% (95%CI: 48.7-49.9) between 2005 and 2016 in Ethiopia. The prevalence was 47.6% (95%CI: 46.1-49.1) in 2005, 42.8% (95%CI: 41.8-43.8) in 2011, and increased to 57.4% (95%CI: 56.3-58.4) in 2016. The pooled data showed that children from households practising open defecation were more likely to be anaemic (AOR: 1.19, 95% CI: 1.05-1.36). In our survey specify analysis, the odds of anaemia were higher among children from households practising open defecation (AOR: 1.33, 95% CI: 1.12-1.58) in the EDHS-2011 and EDHS-2016 (AOR: 1.49, 95% CI: 1.13-1.90). In contrast, neither household water sources nor the time to obtain water was associated with anaemia after controlling for potential confounders. The other variables significantly associated with childhood anaemia include: the child's age (6-35 months), not fully vaccinated (AOR: 1.14, 95%CI: 1.05-1.24), children not dewormed in the last 6 months (AOR: 1.11, 95%CI: 1.01-1.24), children born to mothers not working (AOR: 1.10, 95%CI: 1.02-1.19), children from poor households (AOR: 1.18: 95%CI: 1.06-1.31), and rural residence (AOR: 1.23, 95%CI: 1.06-1.42). CONCLUSION In Ethiopia, about fifty percent of children suffer from childhood anemia, making it a serious public health issue. Open defecation is a major contributing factor to this scourge. To address this issue effectively, it is recommended to strengthen initiatives aimed at eliminating open defecation that involve various approaches, including sanitation infrastructure development, behavior change campaigns, and policy interventions. In addition, to reduce the burden of anemia in children, a multi-faceted approach is necessary, involving both prevention and treatment strategies.
Collapse
Affiliation(s)
- Biniyam Sahiledengle
- Department of Public Health, Madda Walabu University Goba Referral Hospital, Bale-Goba, Ethiopia.
| | - Lillian Mwanri
- Research Centre for Public Health Research, Equity and Human Flourishing, Torrens University Australia, Adelaide Campus, Adelaide, SA, 5000, Australia
| | - Kingsley Emwinyore Agho
- School of Health Sciences, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia
- School of Medicine, Translational Health Research Institute, Western Sydney University, Campbelltown Campus, Penrith, NSW, 2571, Australia
- African Vision Research Institute, University of KwaZulu-Natal, Durban, 4041, South Africa
| |
Collapse
|
26
|
Mostafa I, Hasan SMT, Gazi MA, Alam MA, Fahim SM, Saqeeb KN, Ahmed T. Alteration of stool pH and its association with biomarkers of gut enteropathy among slum-dwelling women of reproductive age in Bangladesh. BMC Womens Health 2023; 23:661. [PMID: 38071298 PMCID: PMC10710701 DOI: 10.1186/s12905-023-02758-6] [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: 05/07/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Recent evidence suggests that measures of maternal gut enteropathy are associated with unfavorable fetal outcomes. It is, therefore, crucial to identify and treat the features of intestinal enteropathy among reproductive-age women living in areas where enteropathy is highly prevalent. However, there is a lack of non-invasive diagnostic tests to determine EED, making it difficult to identify the disease in field settings. In this study, we tested the potential of fecal pH as a biomarker of gut enteropathy and investigated its relationship with fecal biomarkers of intestinal enteropathy in reproductive-age women living in resource-limited environments. METHODS Data on socio-demographic information, anthropometry, and biological samples were collected from 78 apparently healthy women aged between 20 and 27 years from November 2018 to December 2019. The association of stool pH with two fecal biomarkers of gut enteropathy (i.e., intestinal alkaline phosphatase [IAP] and fecal lipocalin-2 [LCN-2] was investigated using multiple linear regression models after adjusting for relevant covariates. RESULTS In the adjusted models, alkaline stool pH (pH > 7.2) was found to be significantly associated with a decrease in the fecal IAP level by 1.05 unit (95% CI: -1.68, -0.42; p < 0.001) in the log scale, and acidic stool pH (pH < 6) was found to be significantly associated with an increase in the fecal LCN-2 level by 0.89 units (95% CI: 0.12, 1.67; p < 0.025) in the log scale. CONCLUSIONS The study findings demonstrated an association of fecal pH with biomarkers of gut enteropathy indicating its applicability as a simple tool for understanding intestinal enteropathy among reproductive-age women living in resource-limited settings.
Collapse
Affiliation(s)
- Ishita Mostafa
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh.
| | - S M Tafsir Hasan
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Amran Gazi
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Ashraful Alam
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Shah Mohammad Fahim
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Kazi Nazmus Saqeeb
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
- Office of the Executive Director, icddr,b, Dhaka, 1212, Bangladesh
- Department of Global Health, University of Washington, Seattle, WA, 98195, USA
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka, 1212, Bangladesh
| |
Collapse
|
27
|
Sturgeon JP, Njunge JM, Bourke CD, Gonzales GB, Robertson RC, Bwakura-Dangarembizi M, Berkley JA, Kelly P, Prendergast AJ. Inflammation: the driver of poor outcomes among children with severe acute malnutrition? Nutr Rev 2023; 81:1636-1652. [PMID: 36977352 PMCID: PMC10639108 DOI: 10.1093/nutrit/nuad030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Severe acute malnutrition (SAM) is the most life-threatening form of undernutrition and underlies at least 10% of all deaths among children younger than 5 years in low-income countries. SAM is a complex, multisystem disease, with physiological perturbations observed in conjunction with the loss of lean mass, including structural and functional changes in many organ systems. Despite the high mortality burden, predominantly due to infections, the underlying pathogenic pathways remain poorly understood. Intestinal and systemic inflammation is heightened in children with SAM. Chronic inflammation and its consequent immunomodulation may explain the increased morbidity and mortality from infections in children with SAM, both during hospitalization and in the longer term after discharge. Recognition of the role of inflammation in SAM is critical in considering new therapeutic targets in this disease, which has not seen a transformational approach to treatment for several decades. This review highlights the central role of inflammation in the wide-ranging pathophysiology of SAM, as well as identifying potential interventions that have biological plausibility based on evidence from other inflammatory syndromes.
Collapse
Affiliation(s)
- Jonathan P Sturgeon
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - James M Njunge
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Claire D Bourke
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - Gerard Bryan Gonzales
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, Netherlands
| | - Ruairi C Robertson
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | | | - James A Berkley
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Paul Kelly
- is with the Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
28
|
Mweetwa MN, Haritunians T, Dube S, Chandwe K, Amadi B, Zyambo K, Liu TC, McGovern D, Kelly P. Genetic variation in environmental enteropathy and stunting in Zambian children: A pilot genome wide association study using the H3Africa chip. PLoS One 2023; 18:e0291311. [PMID: 37756315 PMCID: PMC10529557 DOI: 10.1371/journal.pone.0291311] [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: 10/12/2022] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
PURPOSE Stunting is known to be heavily influenced by environmental factors, so the genetic contribution has received little attention. Here we report an exploration of genetic influences in stunted Zambian children with environmental enteropathy. METHOD Children with stunting (LAZ < -2) were enrolled and given nutritional therapy. Those that were non-responsive to therapy were designated as cases, and children with good growth (LAZ > -1) from the same community as controls. Blood and stool samples were taken to measure biomarkers of intestinal inflammation, epithelial damage, and microbial translocation. Single nucleotide polymorphism array genotyping was carried out on saliva samples using the H3Africa consortium array. RESULTS Genome wide associations were analysed in 117 cases and 41 controls. While no significant associations with stunting were observed at P<5x10-8, likely due to the small sample size, interesting associations were observed at lower thresholds. SNPs associated with stunting were in genomic regions known to modulate neuronal differentiation and fatty acid biosynthesis. SNPs associated with increased microbial translocation were associated with non-integrin membrane ECM interactions, tight junctions, hemostasis, and G-alpha signalling events. SNPs associated with increased inflammation were associated with, ECM interactions, purine metabolism, axon guidance, and cell motility. SNPs negatively associated with inflammation overlapped genes involved in semaphoring interactions. We explored the existing coeliac disease risk HLA genotypes and found present: DQ2.5 (7.5%), DQ8 (3.5%) and DQ2.2 (3.8%); however, no children were positive for coeliac antibodies. We detected HLA-DRB:1301 and HLA-C:1802 with high odds ratios and P<0.05 in stunted children compared to controls. CONCLUSION Genetic variations associated with stunting and the enteropathy underlying it, include variants associated with multiple pathways relating to gene expression, glycosylation, nerve signalling, and sensing of the nutritional and microbiological milieu.
Collapse
Affiliation(s)
- Monica N. Mweetwa
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Department of Physiology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Shishir Dube
- F. Widjaja Foundation Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Kanta Chandwe
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Department of Paediatrics, University of Zambia School of Medicine, Lusaka, Zambia
| | - Beatrice Amadi
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Department of Paediatrics, University of Zambia School of Medicine, Lusaka, Zambia
| | - Kanekwa Zyambo
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Ta-Chiang Liu
- Washington University in St. Louis (WUSTL), St. Louis, Missouri, United States of America
| | - Dermot McGovern
- F. Widjaja Foundation Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Paul Kelly
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Washington University in St. Louis (WUSTL), St. Louis, Missouri, United States of America
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
29
|
Sigh S, Lauritzen L, Wieringa FT, Laillou A, Chamnan C, Stark KD, Roos N. Changes in polyunsaturated fatty acids during treatment of malnourished children may be insufficient to reach required essential fatty acid levels - A randomised controlled trial. Clin Nutr 2023; 42:1778-1787. [PMID: 37572581 DOI: 10.1016/j.clnu.2023.08.003] [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: 03/21/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND & AIMS Severe acute malnutrition (SAM) is a global concern. Studies on the impact of ready-to-use therapeutic foods (RUTFs) on polyunsaturated fatty acids (PUFA) are almost non-existent. The aim was to investigate the change in whole-blood PUFA and nutrition and health markers among Cambodian children with SAM after treatment with RUTFs. METHODS The trial was an 8-week randomised clinical trial of the effectiveness of locally produced fish-based RUTF (L-RUTF) vs standard milk-based RUFT (S-RUTF). Whole-blood fatty acids were analysed using dried blood spots. Nutrition and health markers were assessed using anthropometric assessment and blood samples for markers of inflammation. The trial was conducted at the National Pediatric Hospital, Phnom Penh, Cambodia, with one hundred and twenty-one 6-59-month-old children in treatment for SAM. RESULTS L-RUTF had a higher content of n-3 PUFA and a higher content of arachidonic acid (AA) and docosahexaenoic acid (DHA), while S-RUTF had the highest content of n-6 PUFA. At baseline, the children presented with a Mead acid level in whole-blood of around 0.08% of total fatty acids (FA%) and an omega-3 index of ∼0.91 ± 0.44. After eight weeks of S-RUTF treatment, linoleic acid (LA), AA, n-6/n-3 PUFA ratio, and Mead acid levels were increased. The L-RUTF intervention did not change the whole-blood PUFAs from baseline. At discharge, the children in the L-RUTF group had a lower n-6/n-3 PUFA ratio than the children in the S-RUTF group, driven by a lower alpha-linolenic acid (ALA) (0.20 vs 0.27 FA%, p = 0.004) and lower LA (15.77 vs 14.21 FA%, p = 0.018) with no significant differences in AA or DHA levels. Weight-for-height z-score at discharge was negatively associated with total PUFA (β -1.4 FA%, 95%CI. -2.7; -0.1), n-6 LCPUFA (β -1.3 FA%, 95%CI. -1.3; -0.3), and AA (β -0.6 FA%, 95%CI. -1.0; -0.2). Age-adjusted height was negatively associated with the Mead acid:AA ratio (β -1.2 FA%, 95%CI. -2.2; -0.2). No significant change was seen in inflammation markers within groups or between groups during treatment, and n-3 and n-6 PUFAs were not associated with markers of inflammation or haemoglobin status at discharge. CONCLUSION The trial found that whole-blood markers of PUFA status were low in children at admission and discharge from SAM treatment, indicating that the currently recommended composition of RUTFs are not able to correct their compromised essential fatty acid status. The higher content of DHA and AA in L-RUTF did not give rise to any improvement in PUFA status. No changes in health markers or associations between PUFA and health markers were found. TRIAL REGISTRATION ClinicalTrials.gov: NCT02907424.
Collapse
Affiliation(s)
- Sanne Sigh
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark; Department of Fisheries Post-Harvest Technologies and Quality Control, Fisheries Administration, #186 Preah Norodom Boulevard, 12101 Phnom Penh, Cambodia.
| | - Lotte Lauritzen
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark.
| | - Frank T Wieringa
- UMR QualiSud, Institut de Recherche Pour le Développement (IRD), 34394 Montpellier, France; Qualisud, University of Montpellier, Avignon University, CIRAD, Institut Agro, IRD, Université de la Réunion, 34394 Montpellier, France.
| | - Arnaud Laillou
- Nutrition Section, UNICEF West and Central Africa Region, Dakar 29720, Senegal.
| | - Chhoun Chamnan
- Department of Fisheries Post-Harvest Technologies and Quality Control, Fisheries Administration, #186 Preah Norodom Boulevard, 12101 Phnom Penh, Cambodia.
| | - Ken D Stark
- University of Waterloo, Department of Kinesiology and Health Sciences, 200 University Avenue, Waterloo, ON N2L3G1, Canada.
| | - Nanna Roos
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark.
| |
Collapse
|
30
|
Park HJ, Choi J, Kim H, Yang DY, An TH, Lee EW, Han BS, Lee SC, Kim WK, Bae KH, Oh KJ. Cellular heterogeneity and plasticity during NAFLD progression. Front Mol Biosci 2023; 10:1221669. [PMID: 37635938 PMCID: PMC10450943 DOI: 10.3389/fmolb.2023.1221669] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/18/2023] [Indexed: 08/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a progressive liver disease that can progress to nonalcoholic steatohepatitis (NASH), NASH-related cirrhosis, and hepatocellular carcinoma (HCC). NAFLD ranges from simple steatosis (or nonalcoholic fatty liver [NAFL]) to NASH as a progressive form of NAFL, which is characterized by steatosis, lobular inflammation, and hepatocellular ballooning with or without fibrosis. Because of the complex pathophysiological mechanism and the heterogeneity of NAFLD, including its wide spectrum of clinical and histological characteristics, no specific therapeutic drugs have been approved for NAFLD. The heterogeneity of NAFLD is closely associated with cellular plasticity, which describes the ability of cells to acquire new identities or change their phenotypes in response to environmental stimuli. The liver consists of parenchymal cells including hepatocytes and cholangiocytes and nonparenchymal cells including Kupffer cells, hepatic stellate cells, and endothelial cells, all of which have specialized functions. This heterogeneous cell population has cellular plasticity to adapt to environmental changes. During NAFLD progression, these cells can exert diverse and complex responses at multiple levels following exposure to a variety of stimuli, including fatty acids, inflammation, and oxidative stress. Therefore, this review provides insights into NAFLD heterogeneity by addressing the cellular plasticity and metabolic adaptation of hepatocytes, cholangiocytes, hepatic stellate cells, and Kupffer cells during NAFLD progression.
Collapse
Affiliation(s)
- Hyun-Ju Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Juyong Choi
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hyunmi Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Da-Yeon Yang
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Tae Hyeon An
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Baek-Soo Han
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
31
|
Razanajatovo IM, Andrianomiadana L, Habib A, Randrianarisoa MM, Razafimanjato H, Rakotondrainipiana M, Andriantsalama P, Randriamparany R, Andriamandimby SF, Vonaesch P, Sansonetti PJ, Lacoste V, Randremanana RV, Collard JM, Heraud JM. Factors Associated with Carriage of Enteropathogenic and Non-Enteropathogenic Viruses: A Reanalysis of Matched Case-Control Data from the AFRIBIOTA Site in Antananarivo, Madagascar. Pathogens 2023; 12:1009. [PMID: 37623969 PMCID: PMC10459613 DOI: 10.3390/pathogens12081009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 08/26/2023] Open
Abstract
Environmental Enteric Dysfunction (EED) is an associate driver of stunting in poor settings, and intestinal infections indirectly contribute to the pathophysiology underlying EED. Our work aimed at assessing whether enteric viral carriage is determinant to stunting. A total of 464 healthy and asymptomatic children, aged 2 to 5 years, were recruited, and classified as non-stunted, moderately stunted, or severely stunted. Among the recruited children, 329 stool samples were obtained and screened for enteric and non-enteric viruses by real-time polymerase chain reaction. We statistically tested for the associations between enteric viral and potential risk factors. Approximately 51.7% of the stool samples were positive for at least one virus and 40.7% were positive for non-enteric adenoviruses. No statistical difference was observed between virus prevalence and the growth status of the children. We did not find any statistically significant association between viral infection and most of the socio-demographic risk factors studied, except for having an inadequate food quality score or an over-nourished mother. In addition, being positive for Ascaris lumbricoides was identified as a protective factor against viral infection. In conclusion, we did not find evidence of a direct link between stunting and enteropathogenic viral carriage in our population.
Collapse
Affiliation(s)
- Iony Manitra Razanajatovo
- Virology Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (I.M.R.); (H.R.); (S.F.A.); (V.L.)
| | - Lova Andrianomiadana
- Experimental Bacteriology Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar (A.H.); (J.-M.C.)
| | - Azimdine Habib
- Experimental Bacteriology Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar (A.H.); (J.-M.C.)
| | - Mirella Malala Randrianarisoa
- Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (M.M.R.); (M.R.); (P.A.); (R.R.); (R.V.R.)
| | - Helisoa Razafimanjato
- Virology Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (I.M.R.); (H.R.); (S.F.A.); (V.L.)
| | - Maheninasy Rakotondrainipiana
- Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (M.M.R.); (M.R.); (P.A.); (R.R.); (R.V.R.)
| | - Prisca Andriantsalama
- Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (M.M.R.); (M.R.); (P.A.); (R.R.); (R.V.R.)
| | - Ravaka Randriamparany
- Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (M.M.R.); (M.R.); (P.A.); (R.R.); (R.V.R.)
| | - Soa Fy Andriamandimby
- Virology Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (I.M.R.); (H.R.); (S.F.A.); (V.L.)
| | - Pascale Vonaesch
- Department of Fundamental Microbiology, University of Lausanne, 1002 Lausanne, Switzerland;
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 75015 Paris, France
| | | | - Vincent Lacoste
- Virology Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (I.M.R.); (H.R.); (S.F.A.); (V.L.)
| | - Rindra Vatosoa Randremanana
- Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (M.M.R.); (M.R.); (P.A.); (R.R.); (R.V.R.)
| | - Jean-Marc Collard
- Experimental Bacteriology Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar (A.H.); (J.-M.C.)
| | - Jean-Michel Heraud
- Virology Unit, Institut Pasteur de Madagascar, Antananarivo 101, Madagascar; (I.M.R.); (H.R.); (S.F.A.); (V.L.)
| | | |
Collapse
|
32
|
Sahiledengle B, Mwanri L, Petrucka P, Agho KE. Coexistence of Anaemia and Stunting among Children Aged 6-59 Months in Ethiopia: Findings from the Nationally Representative Cross-Sectional Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6251. [PMID: 37444099 PMCID: PMC10341109 DOI: 10.3390/ijerph20136251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023]
Abstract
INTRODUCTION Stunting and anaemia, two severe public health problems, affect a significant number of children under the age of five. To date, the burden of and predictive factors for coexisting forms of stunting and anaemia in childhood have not been well documented in Ethiopia, where both the conditions are endemic. The primary aims of the present study were to: (i) determine the prevalence of co-morbid anaemia and stunting (CAS); (ii) and identify factors associated with these co-morbid conditions among children aged 6-59 months in Ethiopia. METHODS The study was based on data from the Ethiopian Demographic and Health Survey (EDHS 2005-2016). The EDHS was a cross-sectional study that used a two-stage stratified cluster sampling technique to select households. A total weighted sample of 21,172 children aged 6-59 months was included in the current study (EDHS-2005 (n = 3898), EDHS-2011 (n = 8943), and EDHS-2016 (n = 8332)). Children with height-for-age z-scores (HAZ) less than -2 SD were classified as stunted. Anaemia status was measured by haemoglobin level with readings below 11.0 g/deciliter (g/dL) categorized as anaemic. A multilevel mixed-effects logistic regression model was used to identify the factors associated with CAS. The findings from the models were reported as adjusted odds ratios (AOR) with 95% confidence intervals (CIs). RESULTS Almost half of the children were males (51.1%) and the majority were from rural areas (89.2%). The prevalence of CAS was 24.4% [95% CI: (23.8-24.9)]. Multivariate analyses revealed that children aged 12-23 months, 24-35 months, and 36-59 months, and children perceived by their mothers to be smaller than normal at birth had higher odds of CAS. The odds of CAS were significantly higher among children born to anaemic mothers [AOR: 1.25, 95% CI: (1.11-1.41)], mothers with very short stature [AOR: 2.04, 95% CI: (1.44-2.91)], children from households which practiced open defecation [AOR: 1.57, 95% CI: (1.27-1.92)], children born to mothers without education [AOR: 3.66, 95% CI: (1.85-7.22)], and those who reside in rural areas [AOR: 1.41, 95% CI: (1.10, 1.82)]. Male children had 19% lower odds of having CAS compared to female children [AOR: 0.81, 95% CI: (0.73-0.91)]. Children born to mothers who had normal body mass index (BMI) [AOR: 0.82, 95%CI: (0.73-0.92)] reported lower odds of CAS. CONCLUSIONS One in four preschool-age children in Ethiopia had co-morbid anaemia and stunting, which is a significant public health problem. Future interventions to reduce CAS in Ethiopia should target those children perceived to be small at birth, anaemic mothers, and mothers with short stature.
Collapse
Affiliation(s)
- Biniyam Sahiledengle
- Department of Public Health, Madda Walabu University Goba Referral Hospital, Bale-Goba P.O. Box 302, Ethiopia
| | - Lillian Mwanri
- Research Centre for Public Health, Equity and Human Flourishing, Torrens University Australia, Adelaide Campus, Adelaide 5000, Australia
| | - Pammla Petrucka
- College of Nursing, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Kingsley Emwinyore Agho
- School of Health Sciences, Western Sydney University, Locked Bag 1797, Penrith 2751, Australia
| |
Collapse
|
33
|
Hendrickson SM, Thomas A, Raué HP, Prongay K, Haertel AJ, Rhoades NS, Slifka JF, Gao L, Quintel BK, Amanna IJ, Messaoudi I, Slifka MK. Campylobacter vaccination reduces diarrheal disease and infant growth stunting among rhesus macaques. Nat Commun 2023; 14:3806. [PMID: 37365162 PMCID: PMC10293212 DOI: 10.1038/s41467-023-39433-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Campylobacter-associated enteric disease is estimated to be responsible for more than 160 million cases of gastroenteritis each year and is linked to growth stunting of infants living under conditions of poor sanitation and hygiene. Here, we examine naturally occurring Campylobacter-associated diarrhea among rhesus macaques as a model to determine if vaccination could reduce severe diarrheal disease and infant growth stunting. Compared to unvaccinated controls, there are no Campylobacter diarrhea-associated deaths observed among vaccinated infant macaques and all-cause diarrhea-associated infant mortality is decreased by 76% (P = 0.03). By 9 months of age, there is a 1.3 cm increase in dorsal length that equaled a significant 1.28 LAZ (Length-for-Age Z score) improvement in linear growth among vaccinated infants compared to their unvaccinated counterparts (P = 0.001). In this work, we show that Campylobacter vaccination not only reduces diarrheal disease but also potentially serves as an effective intervention that improves infant growth trajectories.
Collapse
Affiliation(s)
- Sara M Hendrickson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Archana Thomas
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Hans-Peter Raué
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Kamm Prongay
- Division of Animal Resources and Research Support, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Andrew J Haertel
- Division of Animal Resources and Research Support, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Nicholas S Rhoades
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY, 40506, USA
| | - Jacob F Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Lina Gao
- Biostatistics and Bioinformatics Core, Oregon National Primate Research Center, Biostatistics Shared Resource, Knight Cancer Institute, Portland, OR, 97239, USA
| | | | - Ian J Amanna
- Najít Technologies, Inc., Beaverton, OR, 97006, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY, 40506, USA
| | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA.
| |
Collapse
|
34
|
Brouwer AF, Lee GO, Schillinger RJ, Edwards CA, Van Wyk H, Yazbeck R, Morrison DJ. Mechanistic inference of the metabolic rates underlying
13
C breath test curves. J Pharmacokinet Pharmacodyn 2023; 50:203-214. [PMID: 36790613 PMCID: PMC10544773 DOI: 10.1007/s10928-023-09847-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
Carbon stable isotope breath tests offer new opportunities to better understand gastrointestinal function in health and disease. However, it is often not clear how to isolate information about a gastrointestinal or metabolic process of interest from a breath test curve, and it is generally unknown how well summary statistics from empirical curve fitting correlate with underlying biological rates. We developed a framework that can be used to make mechanistic inference about the metabolic rates underlying a 13C breath test curve, and we applied it to a pilot study of 13C-sucrose breath test in 20 healthy adults. Starting from a standard conceptual model of sucrose metabolism, we determined the structural and practical identifiability of the model, using algebra and profile likelihoods, respectively, and we used these results to develop a reduced, identifiable model as a function of a gamma-distributed process; a slower, rate-limiting process; and a scaling term related to the fraction of the substrate that is exhaled as opposed to sequestered or excreted through urine. We demonstrated how the identifiable model parameters impacted curve dynamics and how these parameters correlated with commonly used breath test summary measures. Our work develops a better understanding of how the underlying biological processes impact different aspect of 13C breath test curves, enhancing the clinical and research potential of these 13C breath tests.
Collapse
Affiliation(s)
- Andrew F. Brouwer
- Department of Epidemiology, University of Michigan, 1415 Washington Heights, Ann Arbor, 48109, MI, United States
| | - Gwenyth O. Lee
- Department of Epidemiology, University of Michigan, 1415 Washington Heights, Ann Arbor, 48109, MI, United States
| | - Robert J. Schillinger
- Scottish Universities Environmental Research Centre, University of Glasgow, Rankine Avenue, East Kilbride, G75 0QF, United Kingdom
- School of Medicine, Dentistry and Nursing, University of Glasgow, University Ave, Glasgow, G12 8QQ, United Kingdom
| | - Christine A. Edwards
- School of Medicine, Dentistry and Nursing, University of Glasgow, University Ave, Glasgow, G12 8QQ, United Kingdom
| | - Hannah Van Wyk
- Department of Epidemiology, University of Michigan, 1415 Washington Heights, Ann Arbor, 48109, MI, United States
| | - Roger Yazbeck
- College of Medicine and Public Health, Flinders University, University Ave, Adelaide, 5001, South Australia
| | - Douglas J. Morrison
- Scottish Universities Environmental Research Centre, University of Glasgow, Rankine Avenue, East Kilbride, G75 0QF, United Kingdom
| |
Collapse
|
35
|
Mbabazi J, Pesu H, Mutumba R, Filteau S, Lewis JI, Wells JC, Olsen MF, Briend A, Michaelsen KF, Mølgaard C, Ritz C, Nabukeera-Barungi N, Mupere E, Friis H, Grenov B. Effect of milk protein and whey permeate in large quantity lipid-based nutrient supplement on linear growth and body composition among stunted children: A randomized 2 × 2 factorial trial in Uganda. PLoS Med 2023; 20:e1004227. [PMID: 37220111 DOI: 10.1371/journal.pmed.1004227] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 03/29/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Despite possible benefits for growth, milk is costly to include in foods for undernourished children. Furthermore, the relative effects of different milk components, milk protein (MP), and whey permeate (WP) are unclear. We aimed to assess the effects of MP and WP in lipid-based nutrient supplement (LNS), and of LNS itself, on linear growth and body composition among stunted children. METHODS AND FINDINGS We performed a randomized, double-blind, 2 × 2 factorial trial among 12 to 59 months old stunted children in Uganda. Children were randomized to 4 formulations of LNS with MP or soy protein isolate and WP or maltodextrin (100 g/day for 12 weeks) or no supplementation. Investigators and outcome assessors were blinded; however, participants were only blinded to the ingredients in LNS. Data were analyzed based on intention-to-treat (ITT) using linear mixed-effects models adjusted for age, sex, season, and site. Primary outcomes were change in height and knee-heel length, and secondary outcomes included body composition by bioimpedance analysis (ISRCTN13093195). Between February and September 2020, we enrolled 750 children with a median age of 30 (interquartile range 23 to 41) months, with mean (± standard deviation) height-for-age z-score (HAZ) -3.02 ± 0.74 and 12.7% (95) were breastfed. The 750 children were randomized to LNS (n = 600) with or without MP (n = 299 versus n = 301) and WP (n = 301 versus n = 299), or no supplementation (n = 150); 736 (98.1%), evenly distributed between groups, completed 12-week follow-up. Eleven serious adverse events occurred in 10 (1.3%) children, mainly hospitalization with malaria and anemia, all deemed unrelated to the intervention. Unsupplemented children had 0.06 (95% confidence interval, CI [0.02, 0.10]; p = 0.015) decline in HAZ, accompanied by 0.29 (95% CI [0.20, 0.39]; p < 0.001) kg/m2 increase in fat mass index (FMI), but 0.06 (95% CI [-0.002; 0.12]; p = 0.057) kg/m2 decline in fat-free mass index (FFMI). There were no interactions between MP and WP. The main effects of MP were 0.03 (95% CI [-0.10, 0.16]; p = 0.662) cm in height and 0.2 (95% CI [-0.3, 0.7]; p = 0.389) mm in knee-heel length. The main effects of WP were -0.08 (95% CI [-0.21, 0.05]; p = 220) cm and -0.2 (95% CI [-0.7; 0.3]; p = 403) mm, respectively. Interactions were found between WP and breastfeeding with respect to linear growth (p < 0.02), due to positive effects among breastfed and negative effects among non-breastfed children. Overall, LNS resulted in 0.56 (95% CI [0.42, 0.70]; p < 0.001) cm height increase, corresponding to 0.17 (95% CI [0.13, 0.21]; p < 0.001) HAZ increase, and 0.21 (95% CI [0.14, 0.28]; p < 0.001) kg weight increase, of which 76.5% (95% CI [61.9; 91.1]) was fat-free mass. Using height-adjusted indicators, LNS increased FFMI (0.07 kg/m2, 95% CI [0.0001; 0.13]; p = 0.049), but not FMI (0.01 kg/m2, 95% CI [-0.10, 0.12]; p = 0.800). Main limitations were lack of blinding of caregivers and short study duration. CONCLUSIONS Adding dairy to LNS has no additional effects on linear growth or body composition in stunted children aged 12 to 59 months. However, supplementation with LNS, irrespective of milk, supports linear catch-up growth and accretion of fat-free mass, but not fat mass. If left untreated, children already on a stunting trajectory gain fat at the expense of fat-free mass, thus nutrition programs to treat such children should be considered. TRIAL REGISTRATION ISRCTN13093195.
Collapse
Affiliation(s)
- Joseph Mbabazi
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Hannah Pesu
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Rolland Mutumba
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Suzanne Filteau
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Jack I Lewis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan C Wells
- Childhood Nutrition Research Centre, Population Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Mette F Olsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - André Briend
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Kim F Michaelsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Ritz
- The National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | | | - Ezekiel Mupere
- Department of Paediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Henrik Friis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Benedikte Grenov
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Urlacher SS. The energetics of childhood: Current knowledge and insights into human variation, evolution, and health. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2023. [PMID: 36866969 DOI: 10.1002/ajpa.24719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/22/2022] [Accepted: 02/10/2023] [Indexed: 03/04/2023]
Abstract
How organisms capture and ultimately use metabolic energy-a limiting resource of life-has profound implications for understanding evolutionary legacies and current patterns of phenotypic variation, adaptation, and health. Energetics research among humans has a rich history in biological anthropology and beyond. The energetics of childhood, however, remains relatively underexplored. This shortcoming is notable given the accepted importance of childhood in the evolution of the unique human life history pattern as well as the known sensitivity of childhood development to local environments and lived experiences. In this review, I have three objectives: (1) To overview current knowledge regarding how children acquire and use energy, highlighting work among diverse human populations and pointing to recent advances and remaining areas of uncertainty; (2) To discuss key applications of this knowledge for understanding human variation, evolution, and health; (3) To recommend future avenues for research. A growing body of evidence supports a model of trade-offs and constraint in childhood energy expenditure. This model, combined with advancements on topics such as the energetics of immune activity, the brain, and the gut, provides insights into the evolution of extended human subadulthood and the nature of variation in childhood development, lifetime phenotype, and health.
Collapse
Affiliation(s)
- Samuel S Urlacher
- Department of Anthropology, Baylor University, Waco, Texas, USA
- Child and Brain Development Program, CIFAR, Toronto, Canada
| |
Collapse
|
37
|
Gabain IL, Ramsteijn AS, Webster JP. Parasites and childhood stunting - a mechanistic interplay with nutrition, anaemia, gut health, microbiota, and epigenetics. Trends Parasitol 2023; 39:167-180. [PMID: 36707340 DOI: 10.1016/j.pt.2022.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 01/26/2023]
Abstract
Globally, stunting affects approximately 149.2 million children under 5 years of age. The underlying aetiology and pathophysiological mechanisms leading to stunting remain elusive, and therefore few effective treatment and prevention strategies exist. Crucial evidence directly linking parasites to stunting is often lacking - in part due to the complex nature of stunting, as well as a lack of critical multidisciplinary research amongst key age groups. Here, based on available studies, we present potential mechanistic pathways by which parasitic infection of mother and/or infant may lead to childhood stunting. We highlight the need for future multidisciplinary longitudinal studies and clinical trials aimed at elucidating the most influential factors, and synergies therein, that can lead to stunting, and ultimately towards finding solutions to successfully mitigate against it.
Collapse
Affiliation(s)
- Isobel L Gabain
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, Herts, AL9 7TA, UK; London Centre for Neglected Tropical Diseases Research, Imperial College London Faculty of Medicine, St Mary's Hospital Campus, London, W2 1NY, UK.
| | | | - Joanne P Webster
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, Herts, AL9 7TA, UK; London Centre for Neglected Tropical Diseases Research, Imperial College London Faculty of Medicine, St Mary's Hospital Campus, London, W2 1NY, UK
| |
Collapse
|
38
|
Robertson RC, Edens TJ, Carr L, Mutasa K, Gough EK, Evans C, Geum HM, Baharmand I, Gill SK, Ntozini R, Smith LE, Chasekwa B, Majo FD, Tavengwa NV, Mutasa B, Francis F, Tome J, Stoltzfus RJ, Humphrey JH, Prendergast AJ, Manges AR. The gut microbiome and early-life growth in a population with high prevalence of stunting. Nat Commun 2023; 14:654. [PMID: 36788215 PMCID: PMC9929340 DOI: 10.1038/s41467-023-36135-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/12/2023] [Indexed: 02/16/2023] Open
Abstract
Stunting affects one-in-five children globally and is associated with greater infectious morbidity, mortality and neurodevelopmental deficits. Recent evidence suggests that the early-life gut microbiome affects child growth through immune, metabolic and endocrine pathways. Using whole metagenomic sequencing, we map the assembly of the gut microbiome in 335 children from rural Zimbabwe from 1-18 months of age who were enrolled in the Sanitation, Hygiene, Infant Nutrition Efficacy Trial (SHINE; NCT01824940), a randomized trial of improved water, sanitation and hygiene (WASH) and infant and young child feeding (IYCF). Here, we show that the early-life gut microbiome undergoes programmed assembly that is unresponsive to the randomized interventions intended to improve linear growth. However, maternal HIV infection is associated with over-diversification and over-maturity of the early-life gut microbiome in their uninfected children, in addition to reduced abundance of Bifidobacterium species. Using machine learning models (XGBoost), we show that taxonomic microbiome features are poorly predictive of child growth, however functional metagenomic features, particularly B-vitamin and nucleotide biosynthesis pathways, moderately predict both attained linear and ponderal growth and growth velocity. New approaches targeting the gut microbiome in early childhood may complement efforts to combat child undernutrition.
Collapse
Affiliation(s)
- Ruairi C Robertson
- Blizard Institute, Queen Mary University of London, London, UK
- Microenvironment & Immunity Unit, INSERM U1224, Institut Pasteur, 75015, Paris, France
| | | | - Lynnea Carr
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ethan K Gough
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ceri Evans
- Blizard Institute, Queen Mary University of London, London, UK
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Hyun Min Geum
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Iman Baharmand
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Sandeep K Gill
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Robert Ntozini
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Laura E Smith
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of Public and Ecosystem Health, Cornell University, Ithaca, NY, USA
| | - Bernard Chasekwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Florence D Majo
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Naume V Tavengwa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Batsirai Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Freddy Francis
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Joice Tome
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Jean H Humphrey
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, UK
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Amee R Manges
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada.
- British Columbia Centre for Disease Control, Vancouver, BC, Canada.
| |
Collapse
|
39
|
Sigh S, Roos N, Chhoun C, Laillou A, Wieringa FT. Ready-to-Use Therapeutic Foods Fail to Improve Vitamin A and Iron Status Meaningfully during Treatment for Severe Acute Malnutrition in 6-59-Month-old Cambodian Children. Nutrients 2023; 15:nu15040905. [PMID: 36839263 PMCID: PMC9961841 DOI: 10.3390/nu15040905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Severe acute malnutrition (SAM) remains a global health concern. Studies on the impact of ready-to-use therapeutic foods (RUTFs) on micronutrient status during SAM treatment are almost nonexistent. The objective was to investigate the impact of RUTFs on the iron and vitamin A status of 6-59-month-old children receiving SAM treatment. Biomarkers of vitamin A status (retinol-binding protein, RBP), iron status (ferritin and soluble transferrin receptor, sTfR), and inflammation (C-reactive protein, CRP, and alpha-1 acid glycoprotein, AGP) were collected at admission and discharge (week 8) during an RUTF effectiveness trial. Vitamin A deficiency was defined as RBP <0.70 µmol/L, low body iron as body iron (BI) <0 mg/kg and deficient iron stores as ferritin <12 µg/L. Data were available for 110 and 75 children at admission and discharge, respectively. There was no significant difference in haemoglobin, ferritin, sTfR, BI or RBP concentrations between admission and discharge. At discharge, BI was 0.2 mg/kg lower, and there was a tendency towards a slightly lower RBP concentration, but the prevalence of vitamin A deficiency was low at admission and discharge (6% and 3%, respectively). The small impact of both RUTFs on improving vitamin A and iron status during SAM treatment calls for further research on the bioavailability of micronutrients to enhance the effectiveness of SAM treatment on micronutrient status.
Collapse
Affiliation(s)
- Sanne Sigh
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark
- Department of Fisheries Post-Harvest Technologies and Quality Control, Fisheries Administration, 186 Preah Norodom Boulevard, Phnom Penh 12101, Cambodia
- Correspondence: ; Tel.: +855-(0)95-201-308
| | - Nanna Roos
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 26, 1958 Frederiksberg C, Denmark
| | - Chamnan Chhoun
- Department of Fisheries Post-Harvest Technologies and Quality Control, Fisheries Administration, 186 Preah Norodom Boulevard, Phnom Penh 12101, Cambodia
| | - Arnaud Laillou
- Nutrition Section, UNICEF West and Central Africa Region, Dakar 29720, Senegal
| | - Frank T. Wieringa
- UMR QualiSud, Institut de Recherche Pour le Développement (IRD), 34394 Montpellier, France
- Qualisud, University of Montpellier, Avignon University, CIRAD, Institut Agro, IRD, Université de la Réunion, 34394 Montpellier, France
| |
Collapse
|
40
|
Espira LM, Lee GO, Baye K, Jones AD, Love NG, Eisenberg JNS. Stool biomarkers as measures of enteric pathogen infection in infants from Addis Ababa informal settlements. PLoS Negl Trop Dis 2023; 17:e0011112. [PMID: 36809375 PMCID: PMC9983878 DOI: 10.1371/journal.pntd.0011112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 03/03/2023] [Accepted: 01/19/2023] [Indexed: 02/23/2023] Open
Abstract
Frequent enteric infections in children may be an important cause of growth faltering; however, we do not fully understand the mechanisms by which pathogen infections and the physiological responses to these infections result in poorer growth. Commonly used protein fecal biomarkers (anti-alpha trypsin, neopterin, and myeloperoxidase) provide broad immunological information on an inflammatory response; however, they do not provide information on non-immune processes (e.g., gut integrity) that may be important indicators of chronic end states such as environmental enteric dysfunction (EED). To explore how additional biomarkers will better inform which physiological pathways (both immune and non-immune) are impacted by pathogen exposure we added to the traditional panel of 3 protein fecal biomarkers 4 novel fecal mRNA transcript biomarkers (sucrase isomaltase, caudal homeobox 1, S100A8, and mucin 12) and analyzed stool samples from infants living in informal settlements in Addis Ababa, Ethiopia. To test how this expanded biomarker panel captures distinct pathogen exposure processes we used two different scoring systems. First, we used a theory-based approach to assign each biomarker to specific physiological attributes based on prior understanding of each biomarker. Second, we used data reduction methods to categorize biomarkers and then assign physiological attributes to those categories. We used linear models to examine the association between the derived biomarker scores (based on mRNA and protein levels) and stool pathogen gene counts to determine pathogen specific effects on gut physiology and immune responses. Inflammation scores were positively associated with Shigella and enteropathogenic E.Coli (EPEC) infection, while gut integrity scores were negatively associated with Shigella, EPEC and, shigatoxigenic E.coli (STEC) infection. Our expanded panel of biomarkers hold promise as tools to measure systemic outcomes of enteric pathogen infection. mRNA biomarkers complement established protein biomarkers by providing important cell-specific physiological and immunological consequences of pathogen carriage that can lead to chronic end states such as EED.
Collapse
Affiliation(s)
- Leon M. Espira
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Gwenyth O. Lee
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
- Rutgers Global Health Institute & Department of Biostatistics and Epidemiology School of Public Health Rutgers, The State University of New Jersey, New Brunswick, New Jersey, United States of America
| | - Kaleab Baye
- Center for Food Science and Nutrition, College of Natural and Computational Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Andrew D. Jones
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
| | - Nancy G. Love
- Department of Civil and Environmental Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Joseph N. S. Eisenberg
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
41
|
Citrulline and kynurenine to tryptophan ratio: potential EED (environmental enteric dysfunction) biomarkers in acute watery diarrhea among children in Bangladesh. Sci Rep 2023; 13:1416. [PMID: 36697429 PMCID: PMC9876903 DOI: 10.1038/s41598-023-28114-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023] Open
Abstract
Two emerging biomarkers of environmental enteric dysfunction (EED) include plasma citrulline (CIT), and the kynurenine (KYN): tryptophan (TRP)/ (KT) ratio. We sought to investigate the plasma concentration of CIT and KT ratio among the children having dehydrating diarrhea and examine associations between concentrations of CIT and KT ratio with concurrent factors. For this analysis, we used cross-sectional data from a total of 102, 6-36 months old male children who suffered from non-cholera acute watery diarrhea and had some dehydration admitted to an urban diarrheal hospital, in Bangladesh. CIT, TRP, and KYN concentrations were determined at enrollment from plasma samples using ELIZA. At enrollment, the mean plasma CIT concentration was 864.48 ± 388.55 µmol/L. The mean plasma kynurenine, tryptophan concentrations, and the KT ratio (× 1000) were 6.93 ± 3.08 µmol/L, 33.44 ± 16.39 µmol/L, and 12.12 ± 18.10, respectively. With increasing child age, KYN concentration decreased (coefficient: - 0.26; 95%CI: - 0.49, - 0.04; p = 0.021); with increasing lymphocyte count, CIT concentration decreased (coef.: - 0.01; 95% CI: - 0.02,0.001, p = 0.004); the wasted child had decreased KT ratio (coef.: - 0.6; 95% CI: - 1.18, - 0.02; p = 0.042) after adjusting for potential covariates. The CIT concentration was associated with blood neutrophils (coef.: 0.02; 95% CI: 0.01, 0.03; p < 0.001), lymphocytes (coef.: - 0.02; 95% CI: - 0.03, - 0.02; p < 0.001) and monocyte (coef.: 0.06; 95% CI: 0.01, 0.11; p = 0.021); KYN concentration was negatively associated with basophil (coef.: - 0.62; 95% CI: - 1.23, - 0.01; p = 0.048) after adjusting for age. In addition, total stool output (gm) increased (coef.: 793.84; 95% CI: 187.16, 1400.52; p = 0.011) and also increased duration of hospital stay (hour) (coef.: 22.89; 95% CI: 10.24, 35.54; p = 0.001) with increasing CIT concentration. The morphological changes associated with EED may increase the risk of enteric infection and diarrheal disease among children. Further research is critically needed to better understand the complex mechanisms by which EED biomarkers may impact susceptibility to dehydrating diarrhea in children.
Collapse
|
42
|
Chen B, Jin F, Zhu Y. The impact of access to sanitary toilets on rural adult residents' health: Evidence from the China family panel survey. Front Public Health 2022; 10:1026714. [PMID: 36568759 PMCID: PMC9780489 DOI: 10.3389/fpubh.2022.1026714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Toilet sanitation is related to public health and environmental protection. In the context of the toilet revolution in rural China, an ordered probit regression analysis was conducted to evaluate the impact of access to sanitary toilets on rural residents' health. Using data from the China Family Panel Study (CFPS) in 2014, we found that access to sanitary toilets in rural households significantly improved residents' health, and this finding remained robust across a series of checks. Meanwhile, results of the mechanism analysis showed that preventing feces from contaminating water sources was an important mechanism behind the positive effects of sanitary toilet use on health. We also found that the impact of access to sanitary toilets was more pronounced among female, middle-aged, and low-income people. Toilet revolution plays an important role in ensuring residents' health and protecting water sources, thereby underscoring the need for governments in developing countries to invest in sanitary toilets. In addition, the existing policies and sanitation programs in China need to be improved to promote public health.
Collapse
|
43
|
Fahim SM, Gazi MA, Alam MA, Hasan MM, Das S, Mahfuz M, Ahmed T. Association between Circulating Retinol Binding Protein 4, Body Mass Index, and Biomarkers of Environmental Enteric Dysfunction among Slum-Dwelling Lean Adults in Bangladesh. Am J Trop Med Hyg 2022; 107:1315-1322. [PMID: 36216318 PMCID: PMC9768260 DOI: 10.4269/ajtmh.21-0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/01/2022] [Indexed: 12/30/2022] Open
Abstract
The relationship of retinol binding protein 4 (RBP4) with biomarkers of intestinal health and gut integrity in adults is unknown. We sought to determine the correlation between plasma RBP4 level and BMI and investigate the relationship of circulating RBP4 concentration with biomarkers of environmental enteric dysfunction among lean adults (body mass index [BMI] < 25.0 kg/m2) in Bangladesh. Overall, 270 adults (135 undernourished with a BMI < 18.5 kg/m2 and 135 healthy controls with a BMI between 18.5 and 24.9 kg/m2) aged 18 to 45 years were evaluated. Multivariable linear regression was performed to test the association between RBP4 and fecal biomarkers of impaired gut health. RBP4 concentration was positively correlated (rho = 0.27, P < 0.001) with BMI and was significantly higher in healthy controls than undernourished adults (P < 0.001), in male than female (P < 0.001), and also in employed (P < 0.001), smokers (P = 0.048) and participants with low Self-Reporting Questionnaire (SRQ)-20 scores (an instrument to screen mental health disorders) (P = 0.049). Statistically significant negative correlations were observed between RBP4 and fecal biomarkers of gut enteropathy including myeloperoxidase (rho = -0.23, P < 0.001), neopterin (rho = -0.30, P < 0.001), and alpha-1 anti-trypsin (rho = -0.21, P < 0.001). Multivariable linear regression analysis showed that increased RBP4 concentration was associated with a significant reduction in fecal neopterin (coefficient = -0.95; 95% confidence interval: -1.44 to -0.45]; P < 0.001) after adjustment for age, sex, nutritional status at enrollment, education, dietary diversity score, SRQ-20 score, improved sanitation, household animal exposure, and alpha-1-acid glycoprotein. The study findings revealed an inverse relationship of plasma RBP4 concentration with fecal biomarkers of altered gut health among slum-dwelling lean adults in Bangladesh.
Collapse
Affiliation(s)
- Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh;,Address correspondence to Shah Mohammad Fahim, Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka-1212. E-mail:
| | - Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md. Ashraful Alam
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Md. Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh;,Faculty of Medicine and Life Sciences, University of Tampere, Finland
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh;,Department of Global Health, University of Washington, Seattle, Washington;,James P Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| |
Collapse
|
44
|
Njunge JM, Tickell K, Diallo AH, Sayeem Bin Shahid ASM, Gazi MA, Saleem A, Kazi Z, Ali S, Tigoi C, Mupere E, Lancioni CL, Yoshioka E, Chisti MJ, Mburu M, Ngari M, Ngao N, Gichuki B, Omer E, Gumbi W, Singa B, Bandsma R, Ahmed T, Voskuijl W, Williams TN, Macharia A, Makale J, Mitchel A, Williams J, Gogain J, Janjic N, Mandal R, Wishart DS, Wu H, Xia L, Routledge M, Gong YY, Espinosa C, Aghaeepour N, Liu J, Houpt E, Lawley TD, Browne H, Shao Y, Rwigi D, Kariuki K, Kaburu T, Uhlig HH, Gartner L, Jones K, Koulman A, Walson J, Berkley J. The Childhood Acute Illness and Nutrition (CHAIN) network nested case-cohort study protocol: a multi-omics approach to understanding mortality among children in sub-Saharan Africa and South Asia. Gates Open Res 2022; 6:77. [PMID: 36415883 PMCID: PMC9646488 DOI: 10.12688/gatesopenres.13635.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2022] [Indexed: 08/10/2023] Open
Abstract
Introduction: Many acutely ill children in low- and middle-income settings have a high risk of mortality both during and after hospitalisation despite guideline-based care. Understanding the biological mechanisms underpinning mortality may suggest optimal pathways to target for interventions to further reduce mortality. The Childhood Acute Illness and Nutrition (CHAIN) Network ( www.chainnnetwork.org) Nested Case-Cohort Study (CNCC) aims to investigate biological mechanisms leading to inpatient and post-discharge mortality through an integrated multi-omic approach. Methods and analysis; The CNCC comprises a subset of participants from the CHAIN cohort (1278/3101 hospitalised participants, including 350 children who died and 658 survivors, and 270/1140 well community children of similar age and household location) from nine sites in six countries across sub-Saharan Africa and South Asia. Systemic proteome, metabolome, lipidome, lipopolysaccharides, haemoglobin variants, toxins, pathogens, intestinal microbiome and biomarkers of enteropathy will be determined. Computational systems biology analysis will include machine learning and multivariate predictive modelling with stacked generalization approaches accounting for the different characteristics of each biological modality. This systems approach is anticipated to yield mechanistic insights, show interactions and behaviours of the components of biological entities, and help develop interventions to reduce mortality among acutely ill children. Ethics and dissemination. The CHAIN Network cohort and CNCC was approved by institutional review boards of all partner sites. Results will be published in open access, peer reviewed scientific journals and presented to academic and policy stakeholders. Data will be made publicly available, including uploading to recognised omics databases. Trial registration NCT03208725.
Collapse
Affiliation(s)
- James M. Njunge
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kirkby Tickell
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - Abdoulaye Hama Diallo
- Department of Public Health, Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | | | - Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ali Saleem
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Zaubina Kazi
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Syed Ali
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Caroline Tigoi
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Emily Yoshioka
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - Mohammod Jobayer Chisti
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Moses Mburu
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Moses Ngari
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Narshion Ngao
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Bonface Gichuki
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Elisha Omer
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Wilson Gumbi
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Benson Singa
- Kenya Medical Research Institute, Nairobi, Kenya
| | - Robert Bandsma
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biomedical Sciences, University of Malawi College of Medicine, Blantyre, Malawi
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Wieger Voskuijl
- Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Centre for Global Child Health & Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Thomas N. Williams
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Institute of Global Health Innovation, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alex Macharia
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | | | | | | | - Rupasri Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - David S. Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Hang Wu
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Lei Xia
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Michael Routledge
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yun Yun Gong
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Camilo Espinosa
- Departments of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nima Aghaeepour
- Departments of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | | | | | - Yan Shao
- Wellcome Sanger Institute, Hinxton, UK
| | - Doreen Rwigi
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kevin Kariuki
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Timothy Kaburu
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Holm H. Uhlig
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics and Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Lisa Gartner
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kelsey Jones
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Gastroenterology Department, Great Ormond Street Hospital for Children, London, UK
| | - Albert Koulman
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- NIHR BRC Nutritional Biomarker Laboratory, University of Cambridge, Cambridge, UK
| | - Judd Walson
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - James Berkley
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| |
Collapse
|
45
|
Stunted children display ectopic small intestinal colonization by oral bacteria, which cause lipid malabsorption in experimental models. Proc Natl Acad Sci U S A 2022; 119:e2209589119. [PMID: 36197997 PMCID: PMC9573096 DOI: 10.1073/pnas.2209589119] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Environmental enteric dysfunction (EED) is an inflammatory syndrome postulated to contribute to stunted child growth and to be associated with intestinal dysbiosis and nutrient malabsorption. However, the small intestinal contributions to EED remain poorly understood. This study aimed to assess changes in the proximal and distal intestinal microbiota in the context of stunting and EED and to test for a causal role of these bacterial isolates in the underlying pathophysiology. We performed a cross-sectional study in two African countries recruiting roughly 1,000 children aged 2 to 5 years and assessed the microbiota in the stomach, duodenum, and feces. Upper gastrointestinal samples were obtained from stunted children and stratified according to stunting severity. Fecal samples were collected. We then investigated the role of clinical isolates in EED pathophysiology using tissue culture and animal models. We find that small intestinal bacterial overgrowth (SIBO) is extremely common (>80%) in stunted children. SIBO is frequently characterized by an overgrowth of oral bacteria, leading to increased permeability and inflammation and to replacement of classical small intestinal strains. These duodenal bacterial isolates decrease lipid absorption in both cultured enterocytes and mice, providing a mechanism by which they may exacerbate EED and stunting. Further, we find a specific fecal signature associated with the EED markers fecal calprotectin and alpha-antitrypsin. Our study shows a causal implication of ectopic colonization of oral bacterial isolated from the small intestine in nutrient malabsorption and gut leakiness in vitro. These findings have important therapeutic implications for modulating the microbiota through microbiota-targeted interventions.
Collapse
|
46
|
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
|
47
|
Njunge JM, Tickell K, Diallo AH, Sayeem Bin Shahid ASM, Gazi MA, Saleem A, Kazi Z, Ali S, Tigoi C, Mupere E, Lancioni CL, Yoshioka E, Chisti MJ, Mburu M, Ngari M, Ngao N, Gichuki B, Omer E, Gumbi W, Singa B, Bandsma R, Ahmed T, Voskuijl W, Williams TN, Macharia A, Makale J, Mitchel A, Williams J, Gogain J, Janjic N, Mandal R, Wishart DS, Wu H, Xia L, Routledge M, Gong YY, Espinosa C, Aghaeepour N, Liu J, Houpt E, Lawley TD, Browne H, Shao Y, Rwigi D, Kariuki K, Kaburu T, Uhlig HH, Gartner L, Jones K, Koulman A, Walson J, Berkley J. The Childhood Acute Illness and Nutrition (CHAIN) network nested case-cohort study protocol: a multi-omics approach to understanding mortality among children in sub-Saharan Africa and South Asia. Gates Open Res 2022; 6:77. [PMID: 36415883 PMCID: PMC9646488 DOI: 10.12688/gatesopenres.13635.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 02/15/2024] Open
Abstract
Introduction: Many acutely ill children in low- and middle-income settings have a high risk of mortality both during and after hospitalisation despite guideline-based care. Understanding the biological mechanisms underpinning mortality may suggest optimal pathways to target for interventions to further reduce mortality. The Childhood Acute Illness and Nutrition (CHAIN) Network ( www.chainnnetwork.org) Nested Case-Cohort Study (CNCC) aims to investigate biological mechanisms leading to inpatient and post-discharge mortality through an integrated multi-omic approach. Methods and analysis; The CNCC comprises a subset of participants from the CHAIN cohort (1278/3101 hospitalised participants, including 350 children who died and 658 survivors, and 270/1140 well community children of similar age and household location) from nine sites in six countries across sub-Saharan Africa and South Asia. Systemic proteome, metabolome, lipidome, lipopolysaccharides, haemoglobin variants, toxins, pathogens, intestinal microbiome and biomarkers of enteropathy will be determined. Computational systems biology analysis will include machine learning and multivariate predictive modelling with stacked generalization approaches accounting for the different characteristics of each biological modality. This systems approach is anticipated to yield mechanistic insights, show interactions and behaviours of the components of biological entities, and help develop interventions to reduce mortality among acutely ill children. Ethics and dissemination. The CHAIN Network cohort and CNCC was approved by institutional review boards of all partner sites. Results will be published in open access, peer reviewed scientific journals and presented to academic and policy stakeholders. Data will be made publicly available, including uploading to recognised omics databases. Trial registration NCT03208725.
Collapse
Affiliation(s)
- James M. Njunge
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kirkby Tickell
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - Abdoulaye Hama Diallo
- Department of Public Health, Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | | | - Md. Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Ali Saleem
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Zaubina Kazi
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Syed Ali
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi, Karachi, Pakistan
| | - Caroline Tigoi
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | | | - Emily Yoshioka
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - Mohammod Jobayer Chisti
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Moses Mburu
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Moses Ngari
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Narshion Ngao
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Bonface Gichuki
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Elisha Omer
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Wilson Gumbi
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Benson Singa
- Kenya Medical Research Institute, Nairobi, Kenya
| | - Robert Bandsma
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biomedical Sciences, University of Malawi College of Medicine, Blantyre, Malawi
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Wieger Voskuijl
- Amsterdam UMC location, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Centre for Global Child Health & Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Thomas N. Williams
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Institute of Global Health Innovation, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alex Macharia
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | | | | | | | - Rupasri Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - David S. Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Hang Wu
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Lei Xia
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Michael Routledge
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yun Yun Gong
- School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Camilo Espinosa
- Departments of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nima Aghaeepour
- Departments of Anesthesiology, Pain, and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | | | | | - Yan Shao
- Wellcome Sanger Institute, Hinxton, UK
| | - Doreen Rwigi
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kevin Kariuki
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Timothy Kaburu
- The Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Holm H. Uhlig
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Paediatrics and Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Lisa Gartner
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kelsey Jones
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Gastroenterology Department, Great Ormond Street Hospital for Children, London, UK
| | - Albert Koulman
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- NIHR BRC Nutritional Biomarker Laboratory, University of Cambridge, Cambridge, UK
| | - Judd Walson
- Global Health and Epidemiology, University of Washington, Seattle, Seattle, USA
| | - James Berkley
- The Childhood Acute Illness and Nutrition Network, Nairobi, Kenya
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Center for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| |
Collapse
|
48
|
Bein A, Fadel CW, Swenor B, Cao W, Powers RK, Camacho DM, Naziripour A, Parsons A, LoGrande N, Sharma S, Kim S, Jalili-Firoozinezhad S, Grant J, Breault DT, Iqbal J, Ali A, Denson LA, Moore SR, Prantil-Baun R, Goyal G, Ingber DE. Nutritional deficiency in an intestine-on-a-chip recapitulates injury hallmarks associated with environmental enteric dysfunction. Nat Biomed Eng 2022; 6:1236-1247. [PMID: 35739419 DOI: 10.1038/s41551-022-00899-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/12/2022] [Indexed: 01/03/2023]
Abstract
Environmental enteric dysfunction (EED)-a chronic inflammatory condition of the intestine-is characterized by villus blunting, compromised intestinal barrier function and reduced nutrient absorption. Here we show that essential genotypic and phenotypic features of EED-associated intestinal injury can be reconstituted in a human intestine-on-a-chip lined by organoid-derived intestinal epithelial cells from patients with EED and cultured in nutrient-deficient medium lacking niacinamide and tryptophan. Exposure of the organ chip to such nutritional deficiencies resulted in congruent changes in six of the top ten upregulated genes that were comparable to changes seen in samples from patients with EED. Chips lined with healthy epithelium or with EED epithelium exposed to nutritional deficiencies resulted in severe villus blunting and barrier dysfunction, and in the impairment of fatty acid uptake and amino acid transport; and the chips with EED epithelium exhibited heightened secretion of inflammatory cytokines. The organ-chip model of EED-associated intestinal injury may facilitate the analysis of the molecular, genetic and nutritional bases of the disease and the testing of candidate therapeutics for it.
Collapse
Affiliation(s)
- Amir Bein
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Quris Technologies, Boston, MA, USA
| | - Cicely W Fadel
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA.,Division of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ben Swenor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Wuji Cao
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Rani K Powers
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Pluto Biosciences, Inc., Golden, CO, USA
| | - Diogo M Camacho
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Rheos Medicines, Cambridge, MA, USA
| | - Arash Naziripour
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Andrew Parsons
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Nina LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sanjay Sharma
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Seongmin Kim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sasan Jalili-Firoozinezhad
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Jennifer Grant
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.,Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA.,Harvard Stem Cell Institute, Harvard University, Boston, MA, USA
| | - Junaid Iqbal
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Asad Ali
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sean R Moore
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Virginia, Charlottesville, VA, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA. .,Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA. .,Vascular Biology Program and Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
49
|
Bauer KC, Littlejohn PT, Ayala V, Creus-Cuadros A, Finlay BB. Nonalcoholic Fatty Liver Disease and the Gut-Liver Axis: Exploring an Undernutrition Perspective. Gastroenterology 2022; 162:1858-1875.e2. [PMID: 35248539 DOI: 10.1053/j.gastro.2022.01.058] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/31/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic condition affecting one quarter of the global population. Although primarily linked to obesity and metabolic syndrome, undernutrition and the altered (dysbiotic) gut microbiome influence NAFLD progression. Both undernutrition and NAFLD prevalence are predicted to considerably increase, but how the undernourished gut microbiome contributes to hepatic pathophysiology remains far less studied. Here, we present undernutrition conditions with fatty liver features, including kwashiorkor and micronutrient deficiency. We then review the gut microbiota-liver axis, highlighting key pathways linked to NAFLD progression within both overnutrition and undernutrition. To conclude, we identify challenges and collaborative possibilities of emerging multiomic research addressing the pathology and treatment of undernourished NAFLD.
Collapse
Affiliation(s)
- Kylynda C Bauer
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada; Thoracic and Gastrointestinal Malignancies Branch, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland
| | - Paula T Littlejohn
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Victoria Ayala
- Institut de Recerca Biomèdica de Lleida (IRB-Lleida), Lleida, Spain; Department of Experimental Medicine, Universitat de Lleida, Lleida, Spain
| | - Anna Creus-Cuadros
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada; Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
50
|
Collard JM, Andrianonimiadana L, Habib A, Rakotondrainipiana M, Andriantsalama P, Randriamparany R, Rabenandrasana MAN, Weill FX, Sauvonnet N, Randremanana RV, Guillemot V, Vonaesch P, Sansonetti PJ. High prevalence of small intestine bacteria overgrowth and asymptomatic carriage of enteric pathogens in stunted children in Antananarivo, Madagascar. PLoS Negl Trop Dis 2022; 16:e0009849. [PMID: 35533199 PMCID: PMC9119516 DOI: 10.1371/journal.pntd.0009849] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 05/19/2022] [Accepted: 04/01/2022] [Indexed: 11/26/2022] Open
Abstract
Environmental Enteric Dysfunction (EED) refers to an incompletely defined syndrome of inflammation, reduced absorptive capacity, and reduced barrier function in the small intestine. It is widespread among children and adults in low- and middle-income countries and is also associated with poor sanitation and certain gut infections possibly resulting in an abnormal gut microbiota, small intestinal bacterial overgrowth (SIBO) and stunting. We investigated bacterial pathogen exposure in stunted and non-stunted children in Antananarivo, Madagascar by collecting fecal samples from 464 children (96 severely stunted, 104 moderately stunted and 264 non-stunted) and the prevalence of SIBO in 109 duodenal aspirates from stunted children (61 from severely stunted and 48 from moderately stunted children). SIBO assessed by both aerobic and anaerobic plating techniques was very high: 85.3% when selecting a threshold of ≥105 CFU/ml of bacteria in the upper intestinal aspirates. Moreover, 58.7% of the children showed more than 106 bacteria/ml in these aspirates. The most prevalent cultivated genera recovered were Streptococcus, Neisseria, Staphylococcus, Rothia, Haemophilus, Pantoea and Branhamella. Feces screening by qPCR showed a high prevalence of bacterial enteropathogens, especially those categorized as being enteroinvasive or causing mucosal disruption, such as Shigella spp., enterotoxigenic Escherichia coli, enteropathogenic E. coli and enteroaggregative E. coli. These pathogens were detected at a similar rate in stunted children and controls, all showing no sign of severe diarrhea the day of inclusion but both living in a highly contaminated environment (slum-dwelling). Interestingly Shigella spp. was the most prevalent enteropathogen found in this study (83.3%) without overrepresentation in stunted children. About 2 million children under the age of 5 suffer from stunted growth in Madagascar. Although deficient diet is the major cause of undernutrition, impaired absorption or assimilation caused by Environmental Enteric dysfunction (EED) has been proposed to play an important role in stunting. EED is widespread among children and adults in low- and middle-income countries (LMIC) and is also associated with undernutrition, poor sanitation, certain gut infections resulting in an abnormal gut microbiota and small intestinal bacterial overgrowth (SIBO) although the role of SIBO in EED remains unclear. The current study highlights the presence at high concentrations of bacterial taxa usually found in the oro-pharyngeal sphere in a high number of duodenal fluids of stunted children. This uncommon presence suggests a decompartmentalization of the gastrointestinal tract and a possible pro-inflammatory effect due to the ectopic presence of some of these bacteria in the duodenum. The study also points to a high prevalence of enteropathogens (especially Shigella spp.) in the feces of both stunted and control children, hence preventing from proposing a direct association with stunting. This suggests that, beside combatting poverty and improving diet, environmental sanitation, quality of water sources, hygiene promotion and health education are key points to mitigate stunting and restore nutritional benefits.
Collapse
Affiliation(s)
- Jean-Marc Collard
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, Antananarivo, Madagascar
- * E-mail:
| | - Lova Andrianonimiadana
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - Azimdine Habib
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | | | - Prisca Andriantsalama
- Unité d’Epidémiologie et de Recherche Clinique, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - Ravaka Randriamparany
- Unité d’Epidémiologie et de Recherche Clinique, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - M. A. N. Rabenandrasana
- Unité de Bactériologie Expérimentale, Institut Pasteur de Madagascar, Antananarivo, Madagascar
| | - François-Xavier Weill
- Centre National de Référence des Escherichia coli, Shigella et Salmonella, Unité des Bactéries Pathogènes Entériques, Institut Pasteur, Paris, France
| | - Nathalie Sauvonnet
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
| | | | - Vincent Guillemot
- Hub of Bioinformatics and Biostatistics, Institut Pasteur, Paris, France
| | - Pascale Vonaesch
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France
| | | | | |
Collapse
|