1
|
Haque MA, Islam S, Bashar SJ, Rahman ASMMH, Faruque ASG, Ahmed T, Mahfuz M. Influence of Enterocytozoon bieneusi Infection on Child Growth: A Secondary Analysis of the MAL-ED Birth Cohort Study. Am J Trop Med Hyg 2025; 112:797-803. [PMID: 39874594 PMCID: PMC11965769 DOI: 10.4269/ajtmh.23-0895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 10/13/2024] [Indexed: 01/30/2025] Open
Abstract
Malnutrition in the early days of life is a global public health concern that affects children's growth. It results from a variety of factors, including pathogenic infections. Enterocytozoon bieneusi is a microsporidian parasite that can cause diarrhea and malnutrition in children. The study aimed to assess the impact of E. bieneusi on child growth. The MAL-ED study, a multicountry birth cohort research project, investigated the relationship between enteric infections and malnutrition in participating children from eight countries. A customized real-time polymerase chain reaction-based TaqMan array card was used in this study to identify enteropathogens from stool samples, where E. bieneusi was one of the target pathogens. The impacts of E. bieneusi infection on growth measures were assessed. Mixed-effect linear models were used to investigate the relationship between E. bieneusi and growth outcomes, including length-for-age (LAZ), weight-for-age (WAZ), and weight-for-length (WLZ) Z scores. At the endpoint (last month of measurement), the infected group had significantly lower scores than the noninfected group for all outcomes. The adjusted difference-in-difference (D-in-D) values were -0.53 (95% CI: -0.67 to -0.38) for LAZ, -0.38 (95% CI: -0.52 to -0.23) for WAZ, and -0.22 (95% CI: -0.38 to -0.06) for WLZ. Enterocytozoon bieneusi infection has been identified as a factor associated with reduced linear growth, weight gain, and weight gain relative to linear growth in children, underscoring the importance of treating this infection.
Collapse
Affiliation(s)
| | - Shaumik Islam
- Nutrition Research Division, icddr,b, Dhaka, Bangladesh
| | | | | | | | - Tahmeed Ahmed
- Nutrition Research Division, icddr,b, Dhaka, Bangladesh
| | | |
Collapse
|
2
|
Fitria AL, Dinana Z, Maharani AT, Fumizuki AS, Mumtazya FN, Atmaka DR, Yamani LN, Juniastuti, Lusida MI, Soetjipto, Puspikawati SI. Level of fecal neopterin and calprotectin in asymptomatic and symptomatic norovirus-infected children with malnutrition in Indonesia. J Infect Public Health 2025; 18:102687. [PMID: 39933424 DOI: 10.1016/j.jiph.2025.102687] [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: 10/25/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Norovirus infection is the second most frequent viral agent causing gastroenteritis worldwide. Asymptomatic norovirus infection is prevalent in children and is associated with growth failure. Evaluating the intestinal inflammation of asymptomatic norovirus infection is pivotal to overcoming malnutrition, especially in resource-limited settings. The current study aimed to compare the level of fecal neopterin and calprotectin, as the well-established biomarker for intestinal inflammation, in malnourished children with symptomatic and asymptomatic norovirus infections. METHODS In total 78 stool samples from children under five years old were analyzed in this comparative study. The 67 of the participants were malnourished children, with 24 had asymptomatic (ANV) and 18 had symptomatic norovirus infection (SNV), while 25 had no infections (Mal). We assigned 11 healthy children without any infection as control (Healthy). We assessed the fecal neopterin and calprotectin from the stools samples collected during viral diarrhea surveillance in Surabaya, East Java, Indonesia, using commercially ELISA kit from BT Lab (Zhejiang, China) for human neopterin and calprotectin. The statistical differences were tested using Kruskal-Wallis nonparametric test. RESULTS The level of fecal neopterin (mean ± SD, 15.46 ± 1.75 nmol/L and 15.12 ± 2.41 nmol/L; p-value: 0.744) and calprotectin (mean ± SD, 184.35 ± 35.09 ng/ml and 177.95 ± 49.55 ng/ml; p-value: 0.457) between ANV and SNV was comparable, also between Mal and Healthy groups. Meanwhile, the level of both biomarkers was significantly different in norovirus positive group and norovirus negative group (p-value < 0.000). CONCLUSIONS Norovirus infection, regardless of the presence of symptoms, triggered intestinal inflammation, represented by the increased of fecal neopterin and calprotectin level.
Collapse
Affiliation(s)
- Anisa Lailatul Fitria
- Department of Nutrition, Faculty of Public Health, Universitas Airlangga, Indonesia; Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia
| | - Zayyin Dinana
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia
| | - Aussie Tahta Maharani
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia
| | - Alinda Syifa Fumizuki
- Undergraduate Program of Nutrition, Faculty of Public Health, Universitas Airlangga, Indonesia
| | - Faiz Nabila Mumtazya
- Undergraduate Program of Nutrition, Faculty of Public Health, Universitas Airlangga, Indonesia
| | | | - Laura Navika Yamani
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia; Department of Epidemiology, Faculty of Public Health, Universitas Airlangga, Indonesia
| | - Juniastuti
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia; Department of Microbiology, Faculty of Medicine, Universitas Airlangga, Indonesia
| | - Maria Inge Lusida
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia; Department of Microbiology, Faculty of Medicine, Universitas Airlangga, Indonesia
| | - Soetjipto
- Viral Diarrhea Study Group, Institute of Tropical Disease, Universitas Airlangga, Indonesia; Department of Biochemistry, Faculty of Medicine, Universitas Airlangga, Indonesia.
| | | |
Collapse
|
3
|
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
|
4
|
Verster AJ, Salerno P, Valls R, Barrack K, Price CE, McClure EA, Madan JC, O’Toole GA, Sanville JL, Ross BD. Persistent delay in maturation of the developing gut microbiota in infants with cystic fibrosis. mBio 2025; 16:e0342024. [PMID: 39945545 PMCID: PMC11898760 DOI: 10.1128/mbio.03420-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/23/2025] [Indexed: 02/19/2025] Open
Abstract
The healthy human infant gut microbiome undergoes stereotypical changes in taxonomic composition between birth and maturation to an adult-like stable state. During this time, extensive communication between microbiota and the host immune system contributes to health status later in life. Although there are many reported associations between microbiota compositional alterations and disease in adults, less is known about how microbiome development is altered in pediatric diseases. One pediatric disease linked to altered gut microbiota composition is cystic fibrosis (CF), a multi-organ genetic disease involving impaired chloride secretion across epithelia and heightened inflammation both in the gut and at other body sites. Here, we use shotgun metagenomics to profile the strain-level composition and developmental dynamics of the infant fecal microbiota from several CF and non-CF longitudinal cohorts spanning from birth to greater than 36 months of life. We identify a set of keystone species that define microbiota development in early life in non-CF infants but are missing or decreased in relative abundance in infants with CF, resulting in a delayed pattern of microbiota maturation, persistent entrenchment in a transitional developmental phase, and subsequent failure to attain an adult-like stable microbiota. Delayed maturation is strongly associated with cumulative antibiotic treatments, and we also detect the increased relative abundance of oral-derived bacteria and higher levels of fungi in infants with CF, features that are associated with decreased gut bacterial density. These findings suggest the potential for future directed therapies targeted at overcoming developmental delays in microbiota maturation for infants with CF.IMPORTANCEThe human gastrointestinal tract harbors a diversity of microbes that colonize upon birth and collectively contribute to host health throughout life. Infants with the disease cystic fibrosis (CF) harbor altered gut microbiota compared to non-CF counterparts, with lower levels of beneficial bacteria. How this altered population is established in infants with CF and how it develops over the first years of life is not well understood. By leveraging multiple large non-CF infant fecal metagenomic data sets and samples from a CF cohort collected prior to highly effective modulator therapy, we define microbiome maturation in infants up to 3 years of age. Our findings identify conserved age-diagnostic species in the non-CF infant microbiome that are diminished in abundance in CF counterparts that instead exhibit an enrichment of oral-derived bacteria and fungi associated with antibiotic exposure. Together, our study builds toward microbiota-targeted therapy to restore healthy microbiota dynamics in infants with CF.
Collapse
Affiliation(s)
- Adrian J. Verster
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Paige Salerno
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Rebecca Valls
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Kaitlyn Barrack
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Courtney E. Price
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Emily A. McClure
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Juliette C. Madan
- Department of Pediatrics, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - George A. O’Toole
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Julie L. Sanville
- Department of Pediatrics, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Benjamin D. Ross
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
5
|
Phiri TN, Weatherill JW, Monford-Sanchez E, Serrano-Contreras JI, Melvin C, Kunaka M, Chisenga I, Ngalande P, Mweetwa MN, Besa E, Haider T, Mandal N, Thompson AJ, Edwards CA, Bourke CD, Robertson RC, Posma JM, Banda R, Mwiinga M, Kazhila L, Katsidzira L, Bwakura-Dangarembizi M, Amadi B, Garcia-Perez I, Maitland K, Marchesi JR, Morrison DJ, Frost G, Kelly P. Novel gastrointestinal tools (GI Tools) for evaluating gut functional capacity in adults with environmental enteropathy in Zambia and Zimbabwe: A cross-sectional study protocol. F1000Res 2025; 13:956. [PMID: 40110548 PMCID: PMC11920692 DOI: 10.12688/f1000research.154471.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Background Environmental enteropathy (EE) is a highly prevalent subclinical inflammatory intestinal disorder associated with growth failure, impaired neurocognitive development, poor response to oral vaccines, and micronutrient deficiencies. However, EE research and clinical trials are hampered by the lack of non-invasive tools for measuring intestinal function in detail. This study aims to develop new tools for the measurement of multiple domains of gut functional capacity. Methods The GI TOOLS project is a cross-sectional study that will recruit adults aged 18-65 years with EE in Lusaka, Zambia. Each participant will undergo assessment of gut functional capacity using novel near-point-of-care tools and provide multiple samples for detailed laboratory analyses. Participants will also undergo endoscopy for collection of duodenal biopsies. Novel techniques include stable isotopes approaches to measuring digestion, absorption, and bidirectional transmucosal amino acid flux, a non-invasive fluorescence tool for real-time evaluation of gut permeability, and assessment of reverse permeation of intravenous antibiotics to be carried out separately in Zimbabwe. Stool and duodenal microbiome sequencing using MinION sequencing, metabolome analysis applied to plasma and intestinal fluids, blood immune cell phenotyping, in vitro epithelial barrier models, and duodenal immunohistochemistry will also be used to explore EE in depth. These will all be integrated with gold standard histology and mucosal morphometry, alongside lactulose permeation data, and stool and plasma biomarker analysis. The protocol has been approved by ethics committees and regulators in Zambia, Zimbabwe, and the UK. Participants will give informed consent before they can participate. Anticipated outcomes Based on this extensive phenotyping, tests will be developed which can be simplified and refined for use in adults and children with EE, and for clinical trials. Findings from this project will be disseminated through in-person meetings with caregivers and regulatory bodies, presentations at conferences and in peer-reviewed journals.
Collapse
Affiliation(s)
- Tracy N Phiri
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - James W Weatherill
- Stable Isotope Biochemistry, Scottish Universities Environmental Research Centre, East Kilbride, Scotland, G75 0QF, UK
| | | | - Jose-Ivan Serrano-Contreras
- Department of Metabolism, Digestion and Reproduction, Imperial College London Section of Nutrition Research, London, England, W12 0NN, UK
| | - Callum Melvin
- School of Medicine Dentistry and Nursing, University of Glasgow College of Medical Veterinary and Life Sciences, Glasgow, Scotland, UK
| | - Mirriam Kunaka
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Ian Chisenga
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Perpetual Ngalande
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Monica N Mweetwa
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Ellen Besa
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Tafhima Haider
- Blizard Institute, Queen Mary University of London Barts and The London School of Medicine and Dentistry, London, England, E1 2AT, UK
| | - Nilanjan Mandal
- Imperial College London Faculty of Medicine, London, England, W12 0NN, UK
| | - Alex J Thompson
- Imperial College London Faculty of Medicine, London, England, W12 0NN, UK
| | - Christine A Edwards
- School of Medicine Dentistry and Nursing, University of Glasgow College of Medical Veterinary and Life Sciences, Glasgow, Scotland, UK
| | - Claire D Bourke
- Zvitambo Institute for Maternal and Child Health Research, Harare, Harare Province, Zimbabwe
- Centre for Immunobiology, University of Glasgow College of Medical Veterinary and Life Sciences, Glasgow, Scotland, UK
| | - Ruairi C Robertson
- Blizard Institute, Queen Mary University of London Barts and The London School of Medicine and Dentistry, London, England, E1 2AT, UK
| | - Joram M Posma
- Imperial College London Faculty of Medicine, London, England, W12 0NN, UK
| | - Rosemary Banda
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Mulima Mwiinga
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Lydia Kazhila
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Leolin Katsidzira
- Department of Internal Medicine, University of Zimbabwe College of Health Sciences, Harare, Harare Province, Zimbabwe
| | - Mutsa Bwakura-Dangarembizi
- Zvitambo Institute for Maternal and Child Health Research, Harare, Harare Province, Zimbabwe
- Department of Child, Adolescent and Women's Health, University of Zimbabwe College of Health Sciences, Harare, Harare Province, Zimbabwe
| | - Beatrice Amadi
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
| | - Isabel Garcia-Perez
- Department of Metabolism, Digestion and Reproduction, Imperial College London Section of Nutrition Research, London, England, W12 0NN, UK
| | - Kathryn Maitland
- Department of Infectious Disease and Institute of Global Health and Innovation, Imperial College London Faculty of Medicine, London, England, UK
| | - Julian R Marchesi
- Imperial College London Faculty of Medicine, London, England, W12 0NN, UK
| | - Douglas J Morrison
- Stable Isotope Biochemistry, Scottish Universities Environmental Research Centre, East Kilbride, Scotland, G75 0QF, UK
| | - Gary Frost
- Department of Metabolism, Digestion and Reproduction, Imperial College London Section of Nutrition Research, London, England, W12 0NN, UK
| | - Paul Kelly
- Tropical Gastroenterology & Nutrition group, University of Zambia School of Medicine, Lusaka, Lusaka Province, Zambia
- Blizard Institute, Queen Mary University of London Barts and The London School of Medicine and Dentistry, London, England, E1 2AT, UK
| |
Collapse
|
6
|
Magalhães L, Rodrigues F, Filho J, Gondim R, Ribeiro S, Rôla T, Clementino M, Maciel B, Magalhães P, Havt A, Santos A, Lima A. Quantifying lactulose and mannitol using LC-MS/MS in a clinical study of children with environmental enteric disease. Braz J Med Biol Res 2025; 58:e14156. [PMID: 40053037 PMCID: PMC11884778 DOI: 10.1590/1414-431x2024e14156] [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: 09/09/2024] [Accepted: 12/18/2024] [Indexed: 03/10/2025] Open
Abstract
Dysfunction of the intestinal epithelium barrier (DIEB) is frequent and can lead to serious complications in early childhood when diagnosis and clinical intervention are limited, especially in children with environmental enteric disease and malnutrition. The use of refined analytical techniques is increasingly necessary in this context. This study aimed to validate the high-performance liquid chromatography method coupled with tandem mass spectrometry (LC-MS/MS) to measure DIEB by lactulose:mannitol ratio detection (LM test) in samples of children with different social profiles from Fortaleza, Ceará. The first experimental set was conducted to validate the method through laboratory parameters, such as limit of detection (LD), limit of quantification (LQ), specificity/selectivity, linearity, accuracy, and precision. All validation parameters achieved detection and recovery standards within an acceptable coefficient of variation. Community samples (human development index (HDI) from 0.000 to ≤0.499) were obtained from children from the cohort study Malnutrition-Enteric Diseases, Fortaleza-CE (environmental enteric disease; EED group). The control group samples came from a school located in a region with a high HDI (>0.8). Mannitol excretion was lower in the EED group than in the control group (P<0.0001). On the other hand, LM was higher in this group compared to the control group (P<0.0001). For the first time, a robust analytical approach was used to detect biomarkers of environmental enteropathy (LM) in community samples, confirming with high-sensitivity the damage to the intestinal epithelial barrier function in populations living in low socio-economic conditions.
Collapse
Affiliation(s)
- L.M.V.C. Magalhães
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - F.A.P. Rodrigues
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
- Departamento de Educação Física e Esporte, Instituto Federal de Educação, Ciência e Tecnologia do Ceará, Fortaleza, CE, Brasil
| | - J.Q. Filho
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - R.N.D.G. Gondim
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - S.A. Ribeiro
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - T.B.M. Rôla
- Faculdade de Medicina, Centro Universitário Christus, Fortaleza, CE, Brasil
| | - M.A.F. Clementino
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - B.L.L. Maciel
- Programa de Pós-Graduação em Nutrição, Departamento de Nutrição, Universidade Federal de Rio Grande do Norte, Natal, RN, Brasil
| | - P.J.C. Magalhães
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A. Havt
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A.A. Santos
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - A.A.M. Lima
- Centro de Biomedicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
- Departamento de Fisiologia e Farmacologia, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| |
Collapse
|
7
|
Dobani S, Kirsty Pourshahidi L, Ternan NG, McDougall GJ, Pereira-Caro G, Bresciani L, Mena P, Almutairi TM, Crozier A, Tuohy KM, Del Rio D, Gill CIR. A review on the effects of flavan-3-ols, their metabolites, and their dietary sources on gut barrier integrity. Food Funct 2025; 16:815-830. [PMID: 39807528 DOI: 10.1039/d4fo04721d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Impairment of gut barrier integrity is associated with the pathogenesis of gastrointestinal diseases, including inflammatory bowel disease, colorectal cancer, and coeliac disease. While many aspects of diet have been linked to improved barrier function, (poly)phenols, a broad group of bioactive phytochemicals, are of potential interest. The (poly)phenolic sub-class, flavan-3-ols, have been investigated in some detail owing to their abundance in commonly consumed foods, including grapes, tea, apples, cocoa, berries, and nuts. This review summarises studies on the effects of flavan-3-ols, their microbiome-mediated metabolites, and food sources of these compounds, on gut barrier structure. Extensive evidence demonstrates that flavan-3-ol rich foods, individual flavan-3-ols (e.g., (epi)catechin, epi(gallo)catechin-3-O-gallate, and pro(antho)cyanidins), and their related microbiota-mediated metabolites, could be effective in protecting and restoring the integrity of the gut barrier. In this context, various endpoints are assessed, including transepithelial electrical resistance of the epithelial layer and expression of tight junction proteins and mucins, in ex vivo, in vitro, and animal models. The differences in bioactivity reported for barrier integrity are structure-function dependent, related to the (poly)phenolic source or the tested compound, as well as their dose, exposure time, and presence or absence of a stressor in the experimental system. Overall, these results suggest that flavan-3-ols and related compounds could help to maintain, protect, and restore gut barrier integrity, indicating that they might contribute to the beneficial properties associated with the intake of their dietary sources. However, rigorous and robustly designed human intervention studies are needed to confirm these experimental observations.
Collapse
Affiliation(s)
- Sara Dobani
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - L Kirsty Pourshahidi
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - Nigel G Ternan
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| | - Gordon J McDougall
- Environmental and Biochemical Sciences Department, The James Hutton Institute, Invergowrie, Dundee, UK
| | - Gema Pereira-Caro
- Department of Agroindustry and Food Quality, IFAPA-Alameda Del Obispo, Córdoba, Spain
| | - Letizia Bresciani
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
| | - Pedro Mena
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
- Microbiome Research Hub, Department of Food and Drug, University of Parma, Parma, Italy
| | | | - Alan Crozier
- Department of Chemistry, King Saud University Riyadh, Saudi Arabia
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Kieran M Tuohy
- School of Food Science & Nutrition, University of Leeds, Leeds, UK
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food and Drug, University of Parma, Parma, Italy
- Microbiome Research Hub, Department of Food and Drug, University of Parma, Parma, Italy
| | - Chris I R Gill
- Nutrition Innovation Centre for Food and Health (NICHE), Ulster University, Coleraine, UK.
| |
Collapse
|
8
|
Mehta R, Wenndt AJ. Mycotoxins and bone growth: a review of the literature on associations between xenobiotic exposure and bone growth and development. Nutr Rev 2025; 83:e493-e505. [PMID: 38578611 DOI: 10.1093/nutrit/nuae032] [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: 04/06/2024] Open
Abstract
Mycotoxins are secondary metabolites of fungi that are known to be associated with linear growth faltering because of their impact on inflammation, intestinal damage, inhibition of protein synthesis, and micronutrient absorption. In this narrative review, we aim to extend this analysis to further explore associations between mycotoxins (aflatoxins, ochratoxins, trichothecenes including deoxynivalenol, T-2 toxin, and fumonisins) and long-bone growth, particularly during the saltatory periods of development. Linear growth is a direct function of skeletal development and long-bone growth. We therefore explored biological pathways and mechanisms of impact of these toxins in both animal and human studies, in addition to the epidemiology literature (post-2020). Given what is known of the effects of individual and combinations of mycotoxins based on the animal literature, we have identified a need for further research and examination of how these toxins and exposures may be studied in humans to elucidate the downstream impact on bone-related biomarkers and anthropometric indices used to identify and predict stunting in population-based studies.
Collapse
Affiliation(s)
- Rukshan Mehta
- Centre for Global Child Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, USA
| | | |
Collapse
|
9
|
Lauer JM, Pyykkö J, Chembe M, Billima-Mulenga T, Sikazwe D, Chibwe B, Henderson S, Parkerson D, Leppänen JM, Fink G, Locks LM, Rockers PC. Markers of Environmental Enteric Dysfunction are Associated with Poor Growth and Developmental Outcomes among Young Children in Lusaka, Zambia. J Pediatr 2025; 277:114408. [PMID: 39551093 DOI: 10.1016/j.jpeds.2024.114408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/25/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVE To examine cross-sectional relationships between biomarkers of environmental enteric dysfunction (EED), an acquired subclinical condition of the small intestine, and anthropometric and developmental outcomes among children in Lusaka, Zambia. STUDY DESIGN Serum samples were collected from 240 children aged 27 to 35 months enrolled in a cluster-randomized trial assessing the effects of growth charts and small-quantity lipid-based nutrient supplements on linear growth. Samples were analyzed using the 11-plex Micronutrient and EED Assessment Tool, which incorporates 2 biomarkers of EED, namely intestinal fatty acid-binding protein (I-FABP), a marker of epithelial damage, and soluble CD14 (sCD14), a marker of microbial translocation. Associations between log2-transformed biomarker concentrations and anthropometric (height-for-age z-score [HAZ], weight-for-height z-score, and weight-for-age z-score) and developmental (Global Scales of Early Development development for age z-score and saccadic reaction time [SRT]) outcomes were assessed using linear regression analyses adjusted for background characteristics. RESULTS Mean ± SD HAZ was -1.94 ± 1.10. Higher sCD14 and I-FABP concentrations were significantly associated with lower HAZ (β: -0.21, 95% CI: -0.41, -0.01 and β: -0.20, 95% CI: -0.32, -0.08, respectively). Higher I-FABP concentrations were significantly associated with lower development-for-age z-score (β: -0.22, 95% CI: -0.40, -0.03) and slower SRT (β: 7.37 ms, 95% CI: 2.02, 12.72) as were higher alpha-1-acid glycoprotein concentrations (HAZ β: -0.38, 95% CI: -0.72, -0.03; SRT β: 11.14 ms, 95% CI: 0.94, 21.72). CONCLUSIONS In children in Lusaka, biomarkers of EED were associated with poor anthropometric and developmental outcomes, underscoring the need for interventions to address EED to improve child health globally. CLINICAL TRIAL REGISTRY ClinicalTrials.gov identifier for parent trial: NCT05120427. https://clinicaltrials.gov/ct2/show/NCT05120427.
Collapse
Affiliation(s)
- Jacqueline M Lauer
- Department of Health Sciences, Sargent College of Health & Rehabilitation Sciences, Boston University, Boston, MA.
| | - Juha Pyykkö
- Department of Global Health, Boston University School of Public Health, Boston, MA
| | - Mpela Chembe
- Innovations for Poverty Action Zambia, Lusaka, Zambia
| | | | | | - Bertha Chibwe
- Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | | | | | - Jukka M Leppänen
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
| | - Günther Fink
- University of Basel and Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Lindsey M Locks
- Department of Health Sciences, Sargent College of Health & Rehabilitation Sciences, Boston University, Boston, MA; Department of Global Health, Boston University School of Public Health, Boston, MA.
| | - Peter C Rockers
- Department of Global Health, Boston University School of Public Health, Boston, MA
| |
Collapse
|
10
|
Jesser K, Zhou NA, Hemlock C, Miller-Petrie MK, Contreras JD, Ballard A, Sosa-Moreno A, Calvopiña M, Arnold BF, Cevallos W, Trueba G, Lee GO, Eisenberg JN, Levy K. Environmental Exposures Associated with Enteropathogen Infection in Six-Month-Old Children Enrolled in the ECoMiD Cohort along a Rural-Urban Gradient in Northern Ecuador†. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:103-118. [PMID: 39807583 PMCID: PMC11740902 DOI: 10.1021/acs.est.4c07753] [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: 07/26/2024] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025]
Abstract
Enteropathogens are major contributors to mortality and morbidity, particularly in settings with limited access to water, sanitation, and hygiene infrastructure. To assess transmission pathways associated with enteropathogen infection, we measured household environmental conditions and assayed 22 enteropathogens using TaqMan Array Cards in stool samples from 276 six-month-old children living in communities along a rural-urban gradient in Northern Ecuador. We utilized multivariable models, risk factor importance, and distance-based statistical methods to test factors associated with infection. Most children (89%) carried at least one pathogen, and 72% carried two or more. Bacterial infections (82% of participants) were more common than viruses (58%) or parasites (9.1%). Infants living in the urban site had decreased infection risks compared to those in rural locations. Improved water and sanitation were most predictive of reduced infection risk. Improved water was associated with decreased enterotoxigenic E. coli prevalence, and improved sanitation was associated with lower prevalence of any infection and specifically norovirus. Animal exposure was associated with increased Salmonella prevalence. Children measured during the rainy season had fewer viral and more bacterial infections. Identifying environmental exposures associated with specific pathogen outcomes provides insights into transmission pathways, which contribute critical information for developing effective strategies to improve child health.
Collapse
Affiliation(s)
- Kelsey
J. Jesser
- Department
of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Nicolette A. Zhou
- Department
of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Caitlin Hemlock
- Department
of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Molly K. Miller-Petrie
- Department
of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Jesse D. Contreras
- Department
of Epidemiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - April Ballard
- Gangarosa
Department of Environmental Health, Emory
University, Atlanta, Georgia 30329, United States
| | - Andrea Sosa-Moreno
- Department
of Epidemiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Manuel Calvopiña
- One
Health Research Group, Facultad de Medicina, Universidad De Las Américas (UDLA), Quito 17901, Ecuador
| | - Benjamin F. Arnold
- F.I.
Proctor Foundation and Department of Ophthalmology, University of California at San Francisco, San Francisco, California 94158, United States
| | - William Cevallos
- Centro
de Biomedicina, Universidad Central del
Ecuador, Quito 170136, Ecuador
| | - Gabriel Trueba
- Instituto
de Microbiología Colegio de Ciencias Biológicas
y Ambientales, Universidad San Francisco
de Quito, Quito 170901, Ecuador
| | - Gwenyth O. Lee
- Rutgers
Global Health Institute, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Joseph N.S. Eisenberg
- Department
of Epidemiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Karen Levy
- Department
of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
11
|
Sturgeon JP, Mutasa K, Bwakura-Dangarembizi M, Amadi B, Ngosa D, Dzikiti A, Chandwe K, Besa E, Mutasa B, Murch SH, Hill S, Playford RJ, VanBuskirk K, Kelly P, Prendergast AJ. Therapeutic interventions targeting enteropathy in severe acute malnutrition modulate systemic and vascular inflammation and epithelial regeneration. EBioMedicine 2025; 111:105478. [PMID: 39662176 PMCID: PMC11697704 DOI: 10.1016/j.ebiom.2024.105478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Severe acute malnutrition (SAM) is the most life-threatening form of undernutrition, and children hospitalised with complications have unacceptably high mortality. Complicated SAM is a multisystem disease characterised pathophysiologically by muscle wasting, systemic inflammation, metabolic dysfunction, and malnutrition enteropathy including epithelial barrier dysfunction. There is a clear need for novel interventions to address the underlying pathogenic perturbations of complicated SAM. METHODS In this analysis of tertiary outcomes from a phase II multi-centre trial in Zambia and Zimbabwe, multiplex biomarkers were measured in 122 children (57% male) with SAM randomised following stabilisation ('baseline') to one of four interventions for 14 days to treat malnutrition enteropathy: budesonide, N-acetylglucosamine, colostrum, or teduglutide, compared with standard-of-care. Following measurement of 35 biomarkers from day 15 plasma samples using Luminex and ELISA, the dimensionality of biomarker data was reduced using principal component analysis. FINDINGS Both budesonide and colostrum reduced systemic inflammation (as measured by CD14, IL1-ra, CRP, and LBP), while children receiving colostrum had higher GLP2 and angiopoietin, and lower circulating lipopolysaccharide, suggesting better restoration of epithelial barrier function. N-acetylglucosamine, a precursor for epithelial glycosaminoglycan synthesis, increased biomarkers of epithelial regeneration (EGF, VEGF), and circulating growth factors (angiopoietin, IGFBP-3, and GCSF). INTERPRETATION Interventions aimed at ameliorating malnutrition enteropathy showed plausible effects on biomarkers of inflammation and epithelial regeneration, demonstrating an interdependence of systemic inflammation and enteropathy markers seen in structural analysis. Given the interplay between inflammation and tissue restoration in malnutrition, this mechanism of action supports larger trials to determine the clinical benefits of interventions, either alone or in combination, in children with complicated SAM. FUNDING This analysis of tertiary outcomes for the TAME trial was funded by a Wellcome grant to JPS (220566/Z/20/Z). The TAME trial was funded by a grant from the Medical Research Council (UK), number MR/P024033/1. AJP is funded by Wellcome (108065/Z/15/Z). Takeda UK provided teduglutide at a discounted price.
Collapse
Affiliation(s)
- Jonathan P Sturgeon
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Blizard Institute, Queen Mary University of London, London, UK.
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Mutsa Bwakura-Dangarembizi
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Beatrice Amadi
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Deophine Ngosa
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Anesu Dzikiti
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Kanta Chandwe
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Ellen Besa
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Batsirai Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | | | - Susan Hill
- Great Ormond Street Hospital, London, UK
| | | | - Kelley VanBuskirk
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia
| | - Paul Kelly
- Tropical Gastroenterology and Nutrition Group, University of Zambia, Lusaka, Zambia; Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe; Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
12
|
Colt S, Edielu A, Lewander D, Wu HW, Webb EL, Mawa PA, Nakyesige R, Ayebazibwe AGK, Friedman JF, Bustinduy AL. Associations of poor water, sanitation, and hygiene and parasite burden with markers of environmental enteric dysfunction in preschool-age children infected with Schistosoma mansoni in Uganda. Trop Med Int Health 2025; 30:14-21. [PMID: 39618064 PMCID: PMC11698645 DOI: 10.1111/tmi.14061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is an acquired subclinical condition of the small intestine with lasting health implications for nutritional status, linear growth and development among children. EED is characterised by structural and functional changes to the gut barrier. There are no standardised diagnostic criteria, however, a number of biomarkers have been evaluated to capture EED domains. While the causes of EED are not fully understood, risk factors include poor water, sanitation and hygiene conditions and exposure to enteric pathogens. Very few studies have evaluated the impact of schistosomiasis on EED despite repeated intestinal damage from parasite eggs passing across the gut barrier. METHODS In a cohort of 354 preschool-aged children aged 12-47 months with Schistosoma mansoni infection recruited from the Lake Albert region of Uganda, we assessed exposure to water, sanitation and hygiene conditions and measured markers from each EED domain: intestinal inflammation (faecal calprotectin), epithelial damage (serum intestinal fatty-acid binding-protein), increased permeability (urine lactulose to mannitol ratio and faecal alpha-1 antitrypsin) and microbial translocation (serum endotoxin core antibody). RESULTS In multivariable linear regression models, we found that children whose drinking water was sourced from Lake Albert had higher concentrations of intestinal fatty-acid binding-protein (β = 0.48, 95% CI 0.20-0.76, p < 0.001), and lack of toilet/latrine access was associated with higher concentrations of calprotectin (β = 0.48, 95% CI 0.18-0.78, p < 0.01). Higher schistosomiasis intensity (eggs per gram of stool) was associated with higher calprotectin (β = 0.10, 95% CI 0.02-0.17, p = 0.01), but not with other EED markers. CONCLUSIONS Few studies have investigated schistosomiasis-related morbidities in very young children infected with schistosomiasis. Our findings from Uganda show that poor water, sanitation and hygiene conditions and heavier schistosomiasis burden are associated with intestinal inflammation and damage, contributing to EED. Improved treatment coverage for preschool-aged children infected with schistosomiasis may reduce the burden from EED and associated long-term morbidities.
Collapse
Affiliation(s)
- Susannah Colt
- Center for International Health Research, Rhode Island Hospital, Providence, RI, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Andrew Edielu
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - David Lewander
- Global Health Program, Boston Children’s Hospital, Boston, MA, USA
| | - Hannah W. Wu
- Center for International Health Research, Rhode Island Hospital, Providence, RI, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Emily L. Webb
- Medical Research Council International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Patrice A. Mawa
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Racheal Nakyesige
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - A. Gloria K. Ayebazibwe
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Jennifer F. Friedman
- Center for International Health Research, Rhode Island Hospital, Providence, RI, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Amaya L. Bustinduy
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
13
|
Carboo JA, Malan L, Lombard M, Nienaber A, Dolman-Macleod RC. The relationship between 25-hydroxyvitamin D and markers of intestinal and systemic inflammation in undernourished and non-undernourished children, 6-59 months. Cytokine 2025; 185:156807. [PMID: 39550924 DOI: 10.1016/j.cyto.2024.156807] [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: 03/15/2023] [Revised: 07/05/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Elevated inflammation contributes to growth faltering in children. Vitamin D (vitD) suppresses pro-inflammatory and enhances anti-inflammatory molecule production, thus vitamin D deficiency (VDD) has been associated with heightened inflammation. In undernourished children, VDD and inflammation co-exist, however, little is known about their interaction. OBJECTIVE This cross-sectional study aimed to investigate the association of serum 25-hydroxyvitamin D (25(OH)D) concentration with markers of inflammation in undernourished and non-undernourished children, as well as the effect of vitD supplementation on inflammatory markers in the children with low 25(OH)D in a nested before-and-after trial. METHODS Serum 25(OH)D, IL-1β, IL-8, IL-10, TNF-α, CRP, AGP, IFABP, sCD14, IGF-1 and FGF-21 of 121 undernourished and 51 non-undernourished children aged 6-59 months were measured cross-sectionally. Children with serum 25(OH)D < 30 ng/mL received 50,000 IU/week of vitD for three weeks. RESULTS TNF-α and FGF-21 in the overall and undernourished group were higher in those with serum 25(OH)D < 30 ng/mL compared to those with serum ≥ 30 ng/mL (p < 0.05), while IFABP concentration was higher in the non-undernourished children with serum 25(OH)D < 30 ng/mL (p = 0.047). Serum 25(OH)D was negatively associated with TNF-α in the overall group (β = -0.012, p = 0.034); and FGF-21 (β = -0.013, p = 0.023) in the undernourished group. After the supplementation trial, TNF-α was reduced by 55.9 % (p = 0.008) and 64.7 % (p = 0.017) in the overall and undernourished groups respectively, and AGP showed a trend of 41.6 % reduction (p = 0.099) in the overall group. IL-1β concentration increased post-supplementation in the overall (p = 0.011) and undernourished groups (p = 0.039). CONCLUSION Optimising vitD status may potentially be a strategy for reducing systemic and gut inflammation, and subsequently improving growth, particularly in undernourished children.
Collapse
Affiliation(s)
- Janet Adede Carboo
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa.
| | - Linda Malan
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Martani Lombard
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Arista Nienaber
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | | |
Collapse
|
14
|
González-Fernández D, Yousafzai A, Cousens S, Rizvi A, Ahmed I, Soofi SB, Bhutta ZA. Early life adverse environmental, nutrition and infection factors are associated with lower developmental scores in Pakistani children at 5 years: a cohort study. BMJ Nutr Prev Health 2024; 7:e000900. [PMID: 39882303 PMCID: PMC11773647 DOI: 10.1136/bmjnph-2024-000900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/14/2024] [Indexed: 01/31/2025] Open
Abstract
Background The effects of multiple early adverse psychosocial and biological factors on child development at preschool age in deprived settings are not fully understood. Methods The 'Etiology, Risk Factors and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health and Development' (MAL-ED) project followed children from eight countries, recording sociodemographic, nutritional, illness, enteroinfection biomarkers and scores for quality of home environment (Home Observation for Measurement of the Environment (HOME)), development (Bayley) and maternal depression during the first year of life. In the Pakistan cohort, we investigated associations of these early factors with Z-scores (derived from the eight participating countries) of three developmental outcomes at 5 years: Executive Functions (Z-EF), the Wechsler Preschool and Primary Scale for Intelligence (Z-WPPSI) and the externalising behaviours component of the Strength and Difficulties test (Z-externalising behaviours). Results Most children had 5-year development measurements below other MAL-ED countries (Z-EF<0, 80.3%, Z-WPPSI<0, 69.3%) and 45.6% had Z-externalising behaviours>0. Higher Z-EF was associated with higher HOME (coeff: 0.03 (95% CI 0.005, 0.05), p=0.017) and Bayley scores (0.01 (0.002, 0.01), p=0.010). Higher Z-WPPSI was associated with more household assets (0.02 (0.01, 0.03), p=0.003), but with lower alpha-1 antitrypsin (µmol/L, protein-losing enteropathy) (-0.01 (-0.02, -0.005), p=0.003). Lower externalising behaviour was associated with female sex (-0.30 (-0.53, -0.08), p=0.009), higher soluble-transferrin-receptors (mg/L) (-0.07 (-0.14, -0.01), p=0.024) and initiation of solids/semisolids≥6 months (-0.16 (-0.31, -0.01), p=0.033), but higher externalising behaviour was associated with underweight (0.35 (0.07, 0.62), p=0.014), more diarrhoeal episodes (0.03 (0.004, 0.06), p=0.022) and higher Maternal Depression Score (0.04 (0.01, 0.07), p=0.003) in the first year. Conclusion Adverse environmental, nutrition and infectious factors, and indicators of deprived early development in the first year of life have a negative association with developmental scores at 5 years. Addressing early stressors, improving diet, infections and environment stimulation early in life could positively impact child development in resource-constrained settings.
Collapse
Affiliation(s)
| | - Aisha Yousafzai
- Department of Global Health and Population, Harvard University T H Chan School of Public Health, Boston, Massachusetts, USA
| | - Simon Cousens
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | | | | | - Sajid Bashir Soofi
- Pediatrics & Child Health, Aga Khan University, Karachi, Pakistan
- Center of Excellence in Women & Child Health, Aga Khan University, Karachi, Pakistan
| | - Zulfiqar Ahmed Bhutta
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute for Global Health and Development, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
15
|
Lauer JM, Kirby MA, Muhihi A, Ulenga N, Aboud S, Liu E, Choy RK, Arndt MB, Kou J, Fawzi WW, Gewirtz AT, Sudfeld CR, Manji KP, Duggan CP. Effects of Vitamin D-3 Supplementation During Pregnancy and Lactation on Maternal and Infant Biomarkers of Environmental Enteric Dysfunction, Systemic Inflammation, and Growth: A Secondary Analysis of a Randomized Controlled Trial. J Nutr 2024; 154:3400-3406. [PMID: 39278411 PMCID: PMC11600094 DOI: 10.1016/j.tjnut.2024.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/10/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is an acquired, subclinical state of intestinal inflammation common in children and adults in low-income and middle-income countries. Although vitamin D-3 supplementation has purported anti-inflammatory properties, its ability to ameliorate biomarkers of EED remains unclear. OBJECTIVES This study aimed to examine the effects of maternal vitamin D-3 supplementation during pregnancy and lactation on biomarkers of EED, systemic inflammation, and growth in women living with HIV and their infants in Dar es Salaam, Tanzania. METHODS We conducted subgroup analyses among randomly selected mothers (n = 720) and infants (n = 365 at 6 wk of age, and n = 266 at 6 mo of age) who participated in a randomized, triple-blind, placebo-controlled trial of daily maternal 3000 IU vitamin D-3 supplementation from the second trimester of pregnancy until 1 y postpartum. Biomarkers of EED (soluble CD14 and intestinal fatty acid-binding protein), systemic inflammation (C-reactive protein and α1-acid glycoprotein), and growth factors (insulin-like growth factor 1 and fibroblast growth factor 21) were measured via the Micronutrient and Environmental Enteric Dysfunction Assessment Tool. Anti-flagellin and anti-lipopolysaccharide immunoglobulins were measured via enzyme-linked immunosorbent assay. Comparisons by randomized treatment arm were performed using ordinary least squares regression models with log2-transformed biomarkers. RESULTS At 32 wk of gestation, intestinal fatty acid-binding protein (β: -0.19; P = 0.03) and α1-acid glycoprotein (β:-0.11; P = 0.04) were significantly lower in mothers in the vitamin D-3 group than those in mothers in the placebo group. At 6 wk of age, insulin-like growth factor 1 (β:-0.31; P = 0.03) was significantly lower in infants whose mothers were in the vitamin D-3 group than that in infants whose mothers were in the placebo group. CONCLUSIONS Vitamin D-3 supplementation during pregnancy and lactation reduced selected EED and systemic inflammation biomarkers among women living with HIV. While the effects of maternal vitamin D-3 supplementation do not appear to extend to infants, there may be an effect on growth factors. This trial was registered at clinicaltrials.gov as NCT02305927 (https://clinicaltrials.gov/study/NCT02305927).
Collapse
Affiliation(s)
- Jacqueline M Lauer
- Department of Health Sciences, Sargent College of Health & Rehabilitation Sciences, Boston University, Boston, Massachusetts, United States; Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts, United States.
| | - Miles A Kirby
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Alfa Muhihi
- Africa Academy for Public Health, Dar es Salaam, Tanzania
| | - Nzovu Ulenga
- Management and Development for Health, Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; National Institute for Medical Research, Dar es Salaam, Tanzania
| | - Enju Liu
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts, United States; Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Robert Km Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, United States
| | - Michael B Arndt
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, Washington, United States
| | - Jianqun Kou
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States
| | - Wafaie W Fawzi
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Andrew T Gewirtz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States
| | - Christopher R Sudfeld
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Karim P Manji
- Department of Pediatrics and Child Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Christopher P Duggan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, Massachusetts, United States; Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| |
Collapse
|
16
|
Kivelä L, Lindfors K, Lundin KEA, Størdal K. Review article: Faecal biomarkers for assessing small intestinal damage in coeliac disease and environmental enteropathy. Aliment Pharmacol Ther 2024; 60:988-1004. [PMID: 39233618 DOI: 10.1111/apt.18234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/19/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND In coeliac disease and environmental enteropathy, dietary gluten and enteric infections cause reversible inflammation and morphological changes to the small intestinal mucosa that can be detected in biopsy samples obtained by endoscopy. However, there is a clear need for non-invasive biomarkers. Constant shedding of mucosal material into the bowel lumen and faeces, together with easy availability of stool, makes it an interesting sample matrix. AIMS To conduct a systematic literature search and summarize the existing evidence for host mucosa-derived faecal biomarkers in evaluating small intestinal damage. METHODS We searched for studies on PubMed (MEDLINE) until 1 March 2024. RESULTS We identified 494 studies and included 35 original case-control and cohort studies. These assessed host mucosal transcripts and 14 other markers aiming specifically to reflect inflammation and cell-mediated, innate and gluten-induced immune responses. In coeliac disease, faecal calprotectin and anti-gliadin, tissue transglutaminase, endomysium and deamidated gliadin peptide antibodies were the most studied but with inconsistent results. Single studies reported positive findings about microRNA transcripts, β-defensin-2, lipocalin-2, zonulin-related proteins and angiotensin-converting enzyme. In environmental enteropathy, a non-significant association was reported between calprotectin and urine lactulose/mannitol ratio; there were conflicting results for neopterin, myeloperoxidase and host transcripts. Single studies reported a positive association for lactoferrin, and a negative association for regenerating islet-derived protein 1. Studies comparing faecal markers against small intestinal biopsy findings were not identified in environmental enteropathy. CONCLUSIONS Further studies are needed to determine reliable faecal markers as a proxy for small intestinal mucosal damage.
Collapse
Affiliation(s)
- Laura Kivelä
- Department of Pediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Children's Hospital, Helsinki University Hospital, Helsinki, Finland
- Department of Pediatrics, Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Knut E A Lundin
- Department of Gastroenterology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ketil Størdal
- Department of Pediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Mahfuz M, Coomes D, Abdalla M, Mweetwa M, VanBuskirk K, Iqbal NT, Ali SA, Chandwe K, Das S, Kelly P, Shaikh N, Tarr PI, Denno DM. Biomarker relationships with small bowel histopathology among malnourished children with environmental enteric dysfunction in a multicountry cohort study. Am J Clin Nutr 2024; 120 Suppl 1:S73-S83. [PMID: 39300665 DOI: 10.1016/j.ajcnut.2024.02.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Validated biomarkers could catalyze environmental enteric dysfunction (EED) research. OBJECTIVES Leveraging an EED histology scoring system, this multicountry analysis examined biomarker associations with duodenal histology features among children with EED. We also examined differences in 2-h compared with 1-h urine collections in the lactulose rhamnose (LR) dual sugar test. METHODS Three cohorts of undernourished children unresponsive to nutrition intervention underwent esophagogastroduodenoscopy and duodenal biopsies. Histopathology scores were compared to fecal calprotectin (CAL), myeloperoxidase (MPO), neopterin (NEO), and urinary LR ratio and lactulose percentage recovery. Log-transformed biomarkers were used in linear regressions adjusted for age, center, and sample collection-biopsy time interval in multivariable models. RESULTS Data on >1 biomarker were available for 120 Bangladeshi (CAL, MPO, NEO, and LR), 63 Pakistani (MPO, NEO, and LR), and 63 Zambian children (CAL). Median age at endoscopy was similar (19 mo) across centers. Median sample collection prior to endoscopy was consistent with each center's study design: 2 wk in Bangladesh (urine and stool) and Zambia (stool), and 6 (urine) and 11 (stool) mo in Pakistan. In multivariable models, intraepithelial lymphocytes were associated with CAL (exponentiated [exp.] coefficient: 1.19; 95% confidence interval [CI]: 1, 1.41), intramucosal Brunner's glands with MPO (exp. coefficient: 1.33; 95% CI: 1.05, 1.69) and NEO (exp. coefficient: 1.37; 95% CI: 1.1, 1.7), and chronic inflammation with NEO (exp. coefficient: 1.61; 95% CI: 1.17, 2.17). Intraepithelial lymphocytes were associated with lactulose % recovery (exp. coefficient: 1.22; 95% CI: 1.05, 1.41). LR recovery was substantially lower in 1-h collections than in 2-h collections. CONCLUSIONS Four commonly used markers of enteric dysfunction were associated with specific histologic features. One-hour urine collection may be insufficient to reflect small bowel permeability in LR testing. While acknowledging the challenges with obtaining relevant tissue, these findings form the basis for further EED biomarker validation research.
Collapse
Affiliation(s)
- Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh.
| | - David Coomes
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, United States
| | - Marwa Abdalla
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, United States
| | - Monica Mweetwa
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Kelley VanBuskirk
- Department of Global Health, University of Washington School of Public Health, Seattle, WA, United States
| | - Najeeha T Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - S Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kanta Chandwe
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Subhasish Das
- 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
| | - Nurmohammad Shaikh
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Phillip I Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Donna M Denno
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States.
| |
Collapse
|
18
|
Blaauw J, Chikwana J, Chaima D, Khoswe S, Samikwa L, de Vries I, Voskuijl W. The presence of enteropathy in HIV infected children on antiretroviral therapy in Malawi. PLoS One 2024; 19:e0298310. [PMID: 38330085 PMCID: PMC10852317 DOI: 10.1371/journal.pone.0298310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Undernutrition and malnutrition in children in low- and middle-income countries contribute to high mortality rates. Stunting, a prevalent form of malnutrition, is associated with educational and productivity losses. Environmental enteric dysfunction (EED) and human immunodeficiency virus (HIV) infection worsen these conditions. This study seeks to investigate the presence of enteropathy using EED fecal biomarkers in HIV-infected children who are stable on antiretroviral therapy (ART) across various nutritional statuses. By understanding the interplay between EED, HIV, and nutritional status, this study aims to provide insights that can inform targeted interventions to optimize nutritional outcomes in HIV infected children. METHODS/PRINCIPAL FINDINGS This study evaluated the levels of alpha-1-antitrypsin, calprotectin and myeloperoxidase in frozen fecal samples from 61 HIV infected (mean age 9.16 ±3.08 years) and 31 HIV uninfected (6.65 ±3.41 years) children in Malawi. Anthropometric measurements and clinical data were collected. The height-for-age z-score (-1.66 vs -1.27, p = 0.040) and BMI-for-age z-score (-0.36 vs 0.01, p = 0.037) were lower in HIV infected children. Enzyme-linked immunosorbent assays were used to measure biomarker concentrations. Statistical tests were applied to compare biomarker levels based on HIV status and anthropometric parameters. Myeloperoxidase, alpha-1-antitrypsin, and calprotectin concentrations did not differ between HIV infected and HIV uninfected children of different age groups. In HIV infected children from 5-15 years, there is no difference in biomarker concentration between the stunted and non-stunted groups. CONCLUSION/SIGNIFICANCE Our study found a higher prevalence of stunting in HIV infected children compared to uninfected children, but no significant differences in biomarker concentrations. This suggests no causal relationship between enteropathy and stunting in HIV infected children. These results contribute to the understanding of growth impairment in HIV infected children and emphasize the need for further research, particularly a longitudinal, biopsy-controlled study.
Collapse
Affiliation(s)
- Julia Blaauw
- Department of Global Child Health, Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jessica Chikwana
- Department of Pediatrics & Child Health, The Kamuzu University of Health Sciences, Blantyre, Malawi
| | - David Chaima
- Department of Pediatrics & Child Health, The Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Stanley Khoswe
- Department of Pediatrics & Child Health, The Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Lyson Samikwa
- Department of Pediatrics & Child Health, The Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Isabelle de Vries
- Department of Global Child Health, Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Wieger Voskuijl
- Department of Global Child Health, Amsterdam Institute for Global Health and Development, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
19
|
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
|
20
|
He P, Shen X, Guo S. Intestinal flora and linear growth in children. Front Pediatr 2023; 11:1252035. [PMID: 38034825 PMCID: PMC10687454 DOI: 10.3389/fped.2023.1252035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
The gut microbiota plays a critical role in human growth and development as well as the regulation of human pathophysiological processes. According to research, the gut microbiota controls the host's growth and development in areas such as nutrition, metabolism, endocrine hormones, and immune modulation. The human gut microbiota has an important role in child and adolescent growth, especially when nutritional conditions are poor. In this review, we focus on recent findings about the gut microbiota's influence on child growth, including the relationship between the gut microbiota and linear growth during pregnancy, infancy, childhood, and adolescence. Furthermore, we also review some mechanisms by which intestinal flora influence the host's linear growth. Although the data supports a link between intestinal flora and linear development in children, our review has limitations that prohibit us from fully verifying the causal relationship between gut flora and linear development in children. Improving the gut microbiota, in conjunction with renutrition techniques, has the potential to ameliorate the growth and development impairments currently associated with chronic illness and malnutrition in children.
Collapse
Affiliation(s)
| | | | - Sheng Guo
- Department of Endocrine, Genetics and Metabolism, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Luoma J, Adubra L, Alber D, Ashorn P, Ashorn U, Cloutman-Green E, Diallo F, Ducker C, Elovainio R, Fan YM, Gates L, Gruffudd G, Haapaniemi T, Haidara F, Hallamaa L, Ihamuotila R, Klein N, Martell O, Sow S, Vehmasto T, Cheung YB. Statistical analysis plan for the LAKANA trial: a cluster-randomized, placebo-controlled, double-blinded, parallel group, three-arm clinical trial testing the effects of mass drug administration of azithromycin on mortality and other outcomes among 1-11-month-old infants in Mali. Trials 2023; 24:733. [PMID: 37968741 PMCID: PMC10648361 DOI: 10.1186/s13063-023-07771-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/01/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND The Large-scale Assessment of the Key health-promoting Activities of two New mass drug administration regimens with Azithromycin (LAKANA) trial in Mali aims to evaluate the efficacy and safety of azithromycin (AZI) mass drug administration (MDA) to 1-11-month-old infants as well as the impact of the intervention on antimicrobial resistance (AMR) and mechanisms of action of azithromycin. To improve the transparency and quality of this clinical trial, we prepared this statistical analysis plan (SAP). METHODS/DESIGN LAKANA is a cluster randomized trial that aims to address the mortality and health impacts of biannual and quarterly AZI MDA. AZI is given to 1-11-month-old infants in a high-mortality setting where a seasonal malaria chemoprevention (SMC) program is in place. The participating villages are randomly assigned to placebo (control), two-dose AZI (biannual azithromycin-MDA), and four-dose AZI (quarterly azithromycin-MDA) in a 3:4:2 ratio. The primary outcome of the study is mortality among the intention-to-treat population of 1-11-month-old infants. We will evaluate relative risk reduction between the study arms using a mixed-effects Poisson model with random intercepts for villages, using log link function with person-years as an offset variable. We will model outcomes related to secondary objectives of the study using generalized linear models with considerations on clustering. CONCLUSION The SAP written prior to data collection completion will help avoid reporting bias and data-driven analysis for the primary and secondary aims of the trial. If there are deviations from the analysis methods described here, they will be described and justified in the publications of the trial results. TRIAL REGISTRATION ClinicalTrials.gov ID NCT04424511 . Registered on 11 June 2020.
Collapse
Affiliation(s)
- Juho Luoma
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Laura Adubra
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Dagmar Alber
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Per Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Paediatrics, Tampere University Hospital, Tampere, Finland
| | - Ulla Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Elaine Cloutman-Green
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | | | - Riku Elovainio
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Yue-Mei Fan
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Lily Gates
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | - Tiia Haapaniemi
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Lotta Hallamaa
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Rikhard Ihamuotila
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Nigel Klein
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | - Samba Sow
- Center for Vaccine Development, Bamako, Mali
| | - Taru Vehmasto
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Yin Bun Cheung
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Program in Health Services and Systems Research and Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
22
|
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
|
23
|
Müller-Hauser AA, Huda TMN, Sobhan S, Lambrecht NJ, Waid JL, Wendt AS, Ali S, Rahman M, Gabrysch S. Effect of a Homestead Food Production and Food Hygiene Intervention on Biomarkers of Environmental Enteric Dysfunction in Children Younger Than 24 Months in Rural Bangladesh: A Cluster-Randomized Controlled Trial. Am J Trop Med Hyg 2023; 109:1166-1176. [PMID: 37783459 PMCID: PMC10622486 DOI: 10.4269/ajtmh.23-0153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/08/2023] [Indexed: 10/04/2023] Open
Abstract
Poor sanitation and hygiene practices and inadequate diets can contribute to environmental enteric dysfunction (EED). We evaluated the impact of a combined homestead food production and food hygiene intervention on EED biomarkers in young children in rural Bangladesh. The analysis was conducted within the Food and Agricultural Approaches to Reducing Malnutrition (FAARM) cluster-randomized trial in Sylhet, Bangladesh. The FAARM trial enrolled 2,705 married women and their children younger than 3 years of age in 96 settlements (geographic clusters): 48 intervention and 48 control. The 3-year intervention (2015-2018) included training on gardening, poultry rearing, and improved nutrition practices and was supplemented by an 8-month food hygiene behavior change component, implemented from mid-2017. We analyzed data on 574 children age 0 to 24 months with multilevel linear regression. We assessed fecal myeloperoxidase (MPO), neopterin (NEO), and alpha-1-antitrypsin (AAT) as biomarkers of EED, and serum C-reactive protein (CRP) and alpha-1-acid glycoprotein (AGP) as biomarkers of systemic inflammation, using ELISA. There was no intervention effect on NEO, AAT, CRP, and AGP concentrations, but, surprisingly, MPO levels were increased in children of the intervention group (0.11 log ng/mL; 95% CI, 0.001-0.22). This increase was greater with increasing child age and among intervention households with poultry that were not kept in a shed. A combined homestead food production and food hygiene intervention did not decrease EED in children in our study setting. Small-scale poultry rearing promoted by the intervention might be a risk factor for EED.
Collapse
Affiliation(s)
- Anna A. Müller-Hauser
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Public Health, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
| | - Tarique Md. Nurul Huda
- Department of Public Health, College of Public Health and Health Informatics, Qassim University, Al Bukairiyah, Saudi Arabia
- Environmental Interventions Unit, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Shafinaz Sobhan
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Public Health, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
| | - Nathalie J. Lambrecht
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Public Health, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
| | - Jillian L. Waid
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
- Heidelberg Institute of Global Health, Heidelberg University, Heidelberg, Germany
| | - Amanda S. Wendt
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
| | - Shahjahan Ali
- Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
- Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mahbubur Rahman
- Environmental Interventions Unit, Infectious Diseases Division, icddr,b, Dhaka, Bangladesh
| | - Sabine Gabrysch
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Public Health, Berlin, Germany
- Research Department 2, Potsdam Institute for Climate Impact Research, (PIK), Member of the Leibniz Association, Potsdam, Germany
- Heidelberg Institute of Global Health, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
24
|
Som SV, Wieringa FT, Campos Ponce M, Polman K, Dakurah P, Duncan D, Blomberg J, Rasphone S, Hoeven MVD. Association of both Water, Sanitation and Hygiene (WASH) and Infant and Young Child Feeding (IYCF) practices with childhood malnutrition in Lao PDR: a cross-sectional study of the 2017 Lao Social Indicator Survey II. BMJ Open 2023; 13:e073974. [PMID: 37832981 PMCID: PMC10582958 DOI: 10.1136/bmjopen-2023-073974] [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: 03/23/2023] [Accepted: 09/11/2023] [Indexed: 10/15/2023] Open
Abstract
OBJECTIVE We assessed whether Water, Sanitation and Hygiene (WASH) and Infant and Young Child Feeding (IYCF), either alone or combined, were associated with malnutrition among Lao People's Democratic Republic (Lao PDR) children aged 6 to <24 months. DESIGN This is a secondary analysis of the 2017 Lao Social Indicator Survey II (LSIS II), which used multistage probability proportional to size sampling. Logistic regression analyses were conducted with OR and its corresponding 95% CI. SETTING The LSIS II was conducted a nationwide household-based survey covering all 18 provinces in Lao PDR. PARTICIPANTS We had a total of 3375 children (weighted sample 3345) and 357 households with data on drinking water. OUTCOME MEASURES The outcomes of this study were stunting and wasting. RESULTS The prevalence of stunting and wasting was 28.9% and 10.1%, respectively. Even though households with access to a basic or improved water source were high (82.5%), over 83% of drinking water was contaminated with Escherichia coli. Access to improved sanitation, basic hygiene and adequate IYCF gave a significant lower risk of becoming stunted. The combined effect of these practices on stunting was (adjusted OR (AOR)=0.54; 95% CI=0.41 to 0.73) greater than each practice alone (improved sanitation: AOR=0.75; 95% CI=0.61 to 0.93; basic hygiene: AOR=0.69; 95% CI=0.57 to 0.83; adequate IYCF: AOR=0.79; 95% CI=0.64 to 0.98). Access to improved sanitation and adequate IYCF was associated with a significant lower risk for being wasted, and again the combined effect of these practices was (AOR=0.64; 95% CI=0.44 to 0.92) greater than each practice alone (improved sanitation: AOR=0.68; 95% CI=0.49 to 0.93 and adequate IYCF: AOR=0.66; 95% CI=0.47 to 0.92). CONCLUSION Given the strong associations with both stunting and wasting, and the added benefits when combining WASH and IYCF, there is a need of multisectoral interventions to reduce early childhood malnutrition in Lao PDR.
Collapse
Affiliation(s)
- Somphos Vicheth Som
- Section of Infectious Diseases, Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Frank T Wieringa
- UMR QualiSud, French National Research Institute for Sustainable Development, Montpellier, France
| | - Maiza Campos Ponce
- Section of Infectious Diseases, Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Katja Polman
- Section of Infectious Diseases, Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Prosper Dakurah
- United Nations Children's Emergency Fund Lao PDR, Vientiane, Lao People's Democratic Republic
| | - David Duncan
- United Nations Children's Emergency Fund Lao PDR, Vientiane, Lao People's Democratic Republic
| | - Janneke Blomberg
- United Nations Children's Emergency Fund Lao PDR, Vientiane, Lao People's Democratic Republic
| | - Sitthorot Rasphone
- The Ministry of Planning and Investment's National Economic Research Institute, Vientiane, Lao People's Democratic Republic
| | - Marinka van der Hoeven
- Section of Infectious Diseases, Department of Health Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Mertens A, Benjamin-Chung J, Colford JM, Coyle J, van der Laan MJ, Hubbard AE, Rosete S, Malenica I, Hejazi N, Sofrygin O, Cai W, Li H, Nguyen A, Pokpongkiat NN, Djajadi S, Seth A, Jung E, Chung EO, Jilek W, Subramoney V, Hafen R, Häggström J, Norman T, Brown KH, Christian P, Arnold BF. Causes and consequences of child growth faltering in low-resource settings. Nature 2023; 621:568-576. [PMID: 37704722 PMCID: PMC10511328 DOI: 10.1038/s41586-023-06501-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/31/2023] [Indexed: 09/15/2023]
Abstract
Growth faltering in children (low length for age or low weight for length) during the first 1,000 days of life (from conception to 2 years of age) influences short-term and long-term health and survival1,2. Interventions such as nutritional supplementation during pregnancy and the postnatal period could help prevent growth faltering, but programmatic action has been insufficient to eliminate the high burden of stunting and wasting in low- and middle-income countries. Identification of age windows and population subgroups on which to focus will benefit future preventive efforts. Here we use a population intervention effects analysis of 33 longitudinal cohorts (83,671 children, 662,763 measurements) and 30 separate exposures to show that improving maternal anthropometry and child condition at birth accounted for population increases in length-for-age z-scores of up to 0.40 and weight-for-length z-scores of up to 0.15 by 24 months of age. Boys had consistently higher risk of all forms of growth faltering than girls. Early postnatal growth faltering predisposed children to subsequent and persistent growth faltering. Children with multiple growth deficits exhibited higher mortality rates from birth to 2 years of age than children without growth deficits (hazard ratios 1.9 to 8.7). The importance of prenatal causes and severe consequences for children who experienced early growth faltering support a focus on pre-conception and pregnancy as a key opportunity for new preventive interventions.
Collapse
Affiliation(s)
- Andrew Mertens
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA.
| | - Jade Benjamin-Chung
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - John M Colford
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Jeremy Coyle
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Mark J van der Laan
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Alan E Hubbard
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Sonali Rosete
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Ivana Malenica
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Nima Hejazi
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Oleg Sofrygin
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Wilson Cai
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Haodong Li
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Anna Nguyen
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Nolan N Pokpongkiat
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Stephanie Djajadi
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Anmol Seth
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Esther Jung
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Esther O Chung
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | - Wendy Jilek
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA, USA
| | | | - Ryan Hafen
- Hafen Consulting, West Richland, WA, USA
| | | | - Thea Norman
- Quantitative Sciences, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Kenneth H Brown
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Parul Christian
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Benjamin F Arnold
- Francis I. Proctor Foundation, University of California, San Francisco, San Francisco, CA, USA.
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
26
|
Tickell KD, Denno DM, Saleem A, Kazi Z, Singa BO, Achieng C, Mutinda C, Richardson BA, Ásbjörnsdóttir KH, Hawes SE, Berkley JA, Walson JL. Plasma proteomic signatures of enteric permeability among hospitalized and community children in Kenya and Pakistan. iScience 2023; 26:107294. [PMID: 37554451 PMCID: PMC10405056 DOI: 10.1016/j.isci.2023.107294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/04/2023] [Accepted: 07/03/2023] [Indexed: 08/10/2023] Open
Abstract
We aimed to establish if enteric permeability was associated with similar biological processes in children recovering from hospitalization and relatively healthy children in the community. Extreme gradient boosted models predicting the lactulose-rhamnose ratio (LRR), a biomarker of enteric permeability, using 7,500 plasma proteins and 34 fecal biomarkers of enteric infection among 89 hospitalized and 60 community children aged 2-23 months were built. The R2 values were calculated in test sets. The models performed better among community children (R2: 0.27 [min-max: 0.19, 0.53]) than hospitalized children (R2: 0.07 [min-max: 0.03, 0.11]). In the community, LRR was associated with biomarkers of humoral antimicrobial and cellular lipopolysaccharide responses and inversely associated with anti-inflammatory and innate immunological responses. Among hospitalized children, the selected biomarkers had few shared functions. This suggests enteric permeability among community children was associated with a host response to pathogens, but this association was not observed among hospitalized children.
Collapse
Affiliation(s)
- Kirkby D. Tickell
- Department of Global Health, University of Washington, Seattle, WA, USA
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| | - Donna M. Denno
- Department of Global Health, University of Washington, Seattle, WA, USA
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| | - Ali Saleem
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Aga Khan University, Karachi, Pakistan
| | - Zaubina Kazi
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Aga Khan University, Karachi, Pakistan
| | - Benson O. Singa
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Kenya Medical Research Institute, Nairobi, Kenya
| | - Catherine Achieng
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Kenya Medical Research Institute, Nairobi, Kenya
| | - Charles Mutinda
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Kenya Medical Research Institute, Nairobi, Kenya
| | | | | | - Stephen E. Hawes
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - James A. Berkley
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Judd L. Walson
- Department of Global Health, University of Washington, Seattle, WA, USA
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| |
Collapse
|
27
|
Scharf RJ, McQuade ETR, Svensen E, Huggins A, Maphula A, Bayo E, Blacy L, Pamplona E. de Souza P, Costa H, Houpt ER, Bessong PO, Mduma E, Lima AAM, Guerrant RL. Early-Life Enteric Pathogen Exposure, Socioeconomic Status, and School-Age Cognitive Outcomes. Am J Trop Med Hyg 2023; 109:436-442. [PMID: 37536666 PMCID: PMC10397442 DOI: 10.4269/ajtmh.22-0584] [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: 09/08/2022] [Accepted: 04/05/2023] [Indexed: 08/05/2023] Open
Abstract
Early-life experiences of enteric infections and diarrheal illness are common in low-resource settings and are hypothesized to affect child development. However, longer-term associations of enteric infections with school-age cognitive outcomes are difficult to estimate due to lack of long-term studies. The objective of this study was to examine the relationship between enteropathogen exposure in the first 2 years of life with school-age cognitive skills in a cohort of children followed from birth until 6 to 8 years in low-resource settings in Brazil, Tanzania, and South Africa. The study included participants from three sites from the Etiology, Risk Factors, and Interactions of Enteric Infections and Malnutrition and the Consequences for Child Health Study who were enrolled just after birth and followed for enteric infections, diarrheal illness, and cognitive development until 2 years of age. When the children were school-age, further data were collected on reasoning skills and semantic/phonemic fluency. We estimated associations between the burden of specific enteric pathogens and etiology-specific diarrhea from 0 to 2 years with cognitive test scores at 6 to 8 years using linear regression and adjusting for confounding variables. In this study, children who carried more enteric pathogens in the first 2 years of life showed overall decreases in school-age cognitive abilities, particularly children who carried protozoa, although this was not statistically significant in this sample. Socioeconomic factors such as maternal education and income were more closely associated with school-age cognitive abilities. Early-life enteric pathogens may have a small, lasting influence on school-age cognitive outcomes, although other socioeconomic factors likely contribute more significantly.
Collapse
Affiliation(s)
- Rebecca J. Scharf
- Department of Pediatrics, Neurology and Public Health, University of Virginia, Charlottesville, Virginia
| | | | - Erling Svensen
- Department of Organizational Psychology, Haukeland University Hospital, Bergen, Norway
| | - Amber Huggins
- Department of Public Health, University of Virginia, Charlottesville, Virginia
| | - Angelina Maphula
- Department of Psychology, University of Venda, Thohoyandou, South Africa
| | | | | | | | - Hilda Costa
- Department of Psychology, Federal University of Ceará, Fortaleza, Brazil
| | - Eric R. Houpt
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Pascal O. Bessong
- Department of Microbiology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Aldo A. M. Lima
- Department of Microbiology, Federal University of Ceará, Fortaleza, Brazil
| | | |
Collapse
|
28
|
Salerno P, Verster A, Valls R, Barrack K, Price C, Madan J, O'Toole GA, Ross BD. Persistent delay in maturation of the developing gut microbiota in infants with cystic fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539134. [PMID: 37205374 PMCID: PMC10187160 DOI: 10.1101/2023.05.02.539134] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The healthy human infant gut microbiome undergoes stereotypical changes in taxonomic composition between birth and maturation to an adult-like stable state. During this time, extensive communication between microbiota and the host immune system contributes to health status later in life. Although there are many reported associations between microbiota compositional alterations and disease in adults, less is known about how microbiome development is altered in pediatric diseases. One pediatric disease linked to altered gut microbiota composition is cystic fibrosis (CF), a multi-organ genetic disease involving impaired chloride secretion across epithelia and heightened inflammation both in the gut and at other body sites. Here, we use shotgun metagenomics to profile the strain-level composition and developmental dynamics of the infant fecal microbiota from several CF and non-CF longitudinal cohorts spanning from birth to greater than 36 months of life. We identify a set of keystone species whose prevalence and abundance reproducibly define microbiota development in early life in non-CF infants, but are missing or decreased in relative abundance in infants with CF. The consequences of these CF-specific differences in gut microbiota composition and dynamics are a delayed pattern of microbiota maturation, persistent entrenchment in a transitional developmental phase, and subsequent failure to attain an adult-like stable microbiota. We also detect the increased relative abundance of oral-derived bacteria and higher levels of fungi in CF, features that are associated with decreased gut bacterial density in inflammatory bowel diseases. Our results define key differences in the gut microbiota during ontogeny in CF and suggest the potential for directed therapies to overcome developmental delays in microbiota maturation.
Collapse
|
29
|
Chen C, Patil CL, Mduma ER, Groopman JD, Riley RT, Wu F. Mycotoxins were not associated with environmental enteropathy in a cohort of Tanzanian children. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2023; 43:860-866. [PMID: 35618664 DOI: 10.1111/risa.13956] [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] [Indexed: 06/15/2023]
Abstract
Enteropathy is a pathophysiological condition characterized by decreased intestinal barrier function and absorption. Past studies have hypothesized that mycotoxins might impair children's growth by causing intestinal enteropathy, including interactions between mycotoxins and pathogens. We investigated the association of two mycotoxins, aflatoxin B1 (AFB1 ) and fumonisin B1 (FB1 ), independently and in conjunction with microbial pathogens, with fecal biomarkers of environmental enteropathy in children. As part of a larger MAL-ED study, 196 children were recruited in Haydom, Tanzania, and followed for the first 36 months of life. The gut inflammation biomarkers myeloperoxidase (MPO), neopterin (NEO), and alpha-1-antitrypsin (A1AT) were analyzed in stool samples at 24 months; with mean concentrations 5332.5 ng/L MPO, 807.2 nmol/L NEO, and 0.18 mg/g A1AT. Forty-eight children were measured for AFB1 -lys, with a mean of 5.30 (95% CI: 3.93-6.66) pg/mg albumin; and 87 were measured for FB1 , with a mean of 1.25 (95% CI: 0.72-1.76) ng/ml urine. Although the pathogens adenovirus and Campylobacter were associated with A1AT (p = 0.049) and NEO (p = 0.004), respectively, no association was observed between aflatoxin (MPO, p = 0.30; NEO, p = 0.08; A1AT, p = 0.24) or fumonisin (MPO, p = 0.38; NEO, p = 0.65; A1AT, p = 0.20) exposure and any gut inflammation biomarkers; nor were interactive effects found between mycotoxins and pathogens in contributing to intestinal enteropathy in this cohort. Although further studies are needed to confirm these results, it is possible that mycotoxins contribute to child growth impairment via mechanisms other than disrupting children's intestinal function.
Collapse
Affiliation(s)
- Chen Chen
- School of Public Health, Shandong University, Jinan, China
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan, USA
| | - Crystal L Patil
- Department of Women, Children and Family Health Science, University of Illinois at Chicago, College of Nursing, Chicago, Illinois, USA
| | | | - John D Groopman
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ronald T Riley
- Department of Environmental Health Science, University of Georgia, Athens, Georgia, USA
| | - Felicia Wu
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, Michigan, USA
- Department of Agricultural, Food, and Resource Economics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
30
|
Cowardin CA, Syed S, Iqbal N, Jamil Z, Sadiq K, Iqbal J, Ali SA, Moore SR. Environmental enteric dysfunction: gut and microbiota adaptation in pregnancy and infancy. Nat Rev Gastroenterol Hepatol 2023; 20:223-237. [PMID: 36526906 PMCID: PMC10065936 DOI: 10.1038/s41575-022-00714-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 03/31/2023]
Abstract
Environmental enteric dysfunction (EED) is a subclinical syndrome of intestinal inflammation, malabsorption and barrier disruption that is highly prevalent in low- and middle-income countries in which poverty, food insecurity and frequent exposure to enteric pathogens impair growth, immunity and neurodevelopment in children. In this Review, we discuss advances in our understanding of EED, intestinal adaptation and the gut microbiome over the 'first 1,000 days' of life, spanning pregnancy and early childhood. Data on maternal EED are emerging, and they mirror earlier findings of increased risks for preterm birth and fetal growth restriction in mothers with either active inflammatory bowel disease or coeliac disease. The intense metabolic demands of pregnancy and lactation drive gut adaptation, including dramatic changes in the composition, function and mother-to-child transmission of the gut microbiota. We urgently need to elucidate the mechanisms by which EED undermines these critical processes so that we can improve global strategies to prevent and reverse intergenerational cycles of undernutrition.
Collapse
Affiliation(s)
- Carrie A Cowardin
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA
| | - Sana Syed
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeha Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Zehra Jamil
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Junaid Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Syed Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sean R Moore
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
31
|
Oriá RB, Freitas RS, Roque CR, Nascimento JCR, Silva AP, Malva JO, Guerrant RL, Vitek MP. ApoE Mimetic Peptides to Improve the Vicious Cycle of Malnutrition and Enteric Infections by Targeting the Intestinal and Blood-Brain Barriers. Pharmaceutics 2023; 15:pharmaceutics15041086. [PMID: 37111572 PMCID: PMC10141726 DOI: 10.3390/pharmaceutics15041086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Apolipoprotein E (apoE) mimetic peptides are engineered fragments of the native apoE protein’s LDL-receptor binding site that improve the outcomes following a brain injury and intestinal inflammation in a variety of models. The vicious cycle of enteric infections and malnutrition is closely related to environmental-driven enteric dysfunction early in life, and such chronic inflammatory conditions may blunt the developmental trajectories of children with worrisome and often irreversible physical and cognitive faltering. This window of time for microbiota maturation and brain plasticity is key to protecting cognitive domains, brain health, and achieving optimal/full developmental potential. This review summarizes the potential role of promising apoE mimetic peptides to improve the function of the gut-brain axis, including targeting the blood-brain barrier in children afflicted with malnutrition and enteric infections.
Collapse
Affiliation(s)
- Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
- Correspondence: ; Tel.: +55-85-3366-8239
| | - Raul S. Freitas
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
| | - Cássia R. Roque
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
| | - José Carlos R. Nascimento
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
- Institute of Health Sciences, Medicine, University of International Integration of Afro-Brazilian Lusofonia, Redenção 62790-970, Brazil
| | - Ana Paula Silva
- Institute of Pharmacology and Experimental Therapeutics and Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - João O. Malva
- Institute of Pharmacology and Experimental Therapeutics and Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Michael P. Vitek
- Division of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
32
|
Lauer JM, Kirby MA, Muhihi A, Ulenga N, Aboud S, Liu E, Choy RKM, Arndt MB, Kou J, Fawzi W, Gewirtz A, Sudfeld CR, Manji KP, Duggan CP. Assessing environmental enteric dysfunction via multiplex assay and its relation to growth and development among HIV-exposed uninfected Tanzanian infants. PLoS Negl Trop Dis 2023; 17:e0011181. [PMID: 36943785 PMCID: PMC10030025 DOI: 10.1371/journal.pntd.0011181] [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: 09/20/2022] [Accepted: 02/21/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) may contribute to poor growth and development in young children. While validated EED biomarkers are currently lacking, multiplex assays are able to capture multiple domains of the condition. The purpose of this exploratory study was to examine the relationship between biomarkers of EED and subsequent growth and development among Tanzanian HIV-exposed uninfected (HEU) infants. METHODOLOGY We enrolled 467 infants of mothers living with HIV who had participated in a trial of vitamin D3 supplementation during pregnancy. Infant serum samples collected at 6 weeks (n = 365) and 6 months (n = 266) were analyzed for anti-flagellin and anti-lipopolysaccharide (LPS) IgA and IgG via ELISA as well as the 11-plex Micronutrient and EED Assessment Tool (MEEDAT), which incorporates two biomarkers of EED [intestinal fatty acid-binding protein (I-FABP) and soluble CD14 (sCD14)]. Outcomes were 12-month growth [length-for-age z-score (LAZ), weight-for-length z-score (WLZ), and weight-for-age z-score (WAZ)] and development [Caregiver Reported Early Development Instruments (CREDI) z-scores] and were assessed using linear regression. FINDINGS In primary analyses, higher quartiles of 6-month anti-LPS IgG concentrations were significantly associated with lower LAZ at 12 months (ptrend = 0.040). In secondary analyses, higher log2-transformed 6-week anti-flagellin IgA and 6-month anti-LPS IgA concentrations were significantly associated with lower LAZ at 12 months. No associations were observed between I-FABP or sCD14 and infant growth. However, higher log2-transformed 6-week sCD14 concentrations were significantly associated with lower overall CREDI z-scores, while higher log2-transformed 6-month I-FABP concentrations were significantly associated with higher overall CREDI z-scores. CONCLUSIONS Unlike anti-flagellin and anti-LPS Igs, MEEDAT's biomarkers of EED (I-FABP and sCD14) were not associated with subsequent linear growth among HEU infants in Tanzania. The relationship between EED and infant development warrants further study.
Collapse
Affiliation(s)
- Jacqueline M. Lauer
- Department of Health Sciences, College of Health & Rehabilitation Sciences: Sargent College, Boston University, Boston, Massachusetts, United States of America
| | - Miles A. Kirby
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Alfa Muhihi
- Management and Development for Health, Dar es Salaam, Tanzania
| | - Nzovu Ulenga
- Management and Development for Health, Dar es Salaam, Tanzania
| | - Said Aboud
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Enju Liu
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, United States of America
| | - Michael B. Arndt
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, Washington, United States of America
| | - Jianqun Kou
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Wafaie Fawzi
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Andrew Gewirtz
- Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Christopher R. Sudfeld
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Karim P. Manji
- Department of Pediatrics and Child Health, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Christopher P. Duggan
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
33
|
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
|
34
|
Uddin IM, Endres K, Parvin T, Bhuyian MSI, Zohura F, Masud J, Monira S, Hasan MT, Biswas SK, Sultana M, Thomas ED, Perin J, Sack DA, Faruque A, Alam M, George CM. Food Hygiene and Fecal Contamination on the Household Compound are Associated with Increased Pediatric Diarrhea in Urban Bangladesh (CHoBI7 Program). Am J Trop Med Hyg 2023; 108:524-529. [PMID: 36746654 PMCID: PMC9978565 DOI: 10.4269/ajtmh.22-0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/27/2022] [Indexed: 02/08/2023] Open
Abstract
In this prospective cohort study, we explored individual- and household-level risk factors associated with diarrheal diseases among 251 children younger than 5 years in slum areas of urban Dhaka, Bangladesh. During the 3-month study period, diarrhea surveillance was conducted monthly, and spot checks of the household compound were performed at baseline to assess the presence of feces (animal or human) in the household compound and in cooking and food storage areas, and to assess whether cooked food was covered and refrigerated. We also collected caregiver reports on child mouthing behaviors. Children living in households with feces within 10 steps of cooking and food storage areas (odds ratio [OR], 8.43; 95% CI, 1.01-70.18), those with visible feces found on the ground of the household compound (OR, 4.05; 95% CI, 1.24-13.22), and those in households found to keep cooked food uncovered and without refrigeration (OR, 6.16; 95% CI, 1.11-34.25) during spot checks had a significantly greater odds of diarrhea. There was no significant association between pediatric diarrhea and caregiver-reported child mouthing behaviors or presence of animals in the cooking area. These study findings demonstrate that presence of visible feces in the household compound and near cooking and food storage areas, and poor household food hygiene practices, were significant risk factors for diarrheal disease among young children in Dhaka, Bangladesh. Health communication programs are needed to target these exposure pathways to fecal pathogens.
Collapse
Affiliation(s)
- Ismat Minhaj Uddin
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Kelly Endres
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Tahmina Parvin
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | | | - Fatema Zohura
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Jahed Masud
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Shirajum Monira
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - M. Tasdik Hasan
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Shwapon Kumar Biswas
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Marzia Sultana
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Elizabeth D. Thomas
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jamie Perin
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - David A. Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - A.S.G. Faruque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Munirul Alam
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Christine Marie George
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
35
|
González-Fernández D, Cousens S, Rizvi A, Chauhadry I, Soofi SB, Bhutta ZA. Infections and nutrient deficiencies during infancy predict impaired growth at 5 years: Findings from the MAL-ED study in Pakistan. Front Nutr 2023; 10:1104654. [PMID: 36875830 PMCID: PMC9982131 DOI: 10.3389/fnut.2023.1104654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/10/2023] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND Socio-economic, nutritional, and infectious factors have been associated with impaired infant growth, but how the presence of these factors during infancy affects growth around 5 years is not well understood. METHODS This secondary analysis of the MAL-ED cohort included 277 children from Pakistan for whom socio-demographic, breastfeeding, complementary foods, illness, nutritional biomarkers, stool pathogens and environmental enteropathy indicators between 0 and 11 months were recorded. We used linear regression models to analyze associations of these indicators with height-for-age (HAZ), weight-for-age (WAZ) and weight-for-height (WLZ) at 54-66 months (~5 years), and Poisson regression with robust standard errors to estimate risk ratios for stunting and underweight ~5 years, controlling for gender, first available weight, and income. RESULTS Among the 237 infants followed longitudinally and evaluated at about 5 years of age, exclusive breastfeeding was short (median = 14 days). Complementary feeding started before 6 months with rice, bread, noodles, or sugary foods. Roots, dairy products, fruits/vegetables, and animal-source foods were provided later than recommended (9-12 months). Anemia (70.9%), deficiencies in iron (22.0%), zinc (80.0%), vitamin A (53.4%) and iodine (13.3%) were common. Most infants (>90%) presented with diarrhea and respiratory infections in their first year. At ~5 years, low WAZ (mean-1.91 ± 0.06) and LAZ (-2.11 ± 0.06) resulted in high prevalence of stunting (55.5%) and underweight (44.4%) but a relatively low rate of wasting (5.5%). While 3.4% had concurrent stunting and wasting ~5 years, 37.8% of children had coexisting stunting and underweight. A higher income and receiving formula or dairy products during infancy were associated with a higher LAZ ~5 years, but infant's history of hospitalizations and more respiratory infections were associated with lower LAZ and higher risk of stunting ~5 years. Infants' intake of commercial baby foods and higher serum-transferrin receptors were associated with higher WAZ and lower risk of underweight ~5 years. Presence of Campylobacter and fecal neopterin >6.8 nmol/L in the first year were associated with increased risk of underweight ~5 years. CONCLUSION Growth indicators ~5 years were associated with poverty, inappropriate complementary feeding, and infections during the first year of life, which supports the early start of public health interventions for preventing growth delay ~5 years.
Collapse
Affiliation(s)
| | - Simon Cousens
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Arjumand Rizvi
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Imran Chauhadry
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Sajid Bashir Soofi
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Zulfiqar Ahmed Bhutta
- SickKids Centre for Global Child Health, Toronto, ON, Canada
- Center of Excellence in Women and Child Health, The Aga Khan University, Karachi, Pakistan
- Institute for Global Health and Development, The Aga Khan University, London, United Kingdom
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
George CM, Birindwa A, Li S, Williams C, Kuhl J, Thomas E, François R, Presence AS, Claude BRJ, Mirindi P, Bisimwa L, Perin J, Stine OC. Akkermansia muciniphila Associated with Improved Linear Growth among Young Children, Democratic Republic of the Congo. Emerg Infect Dis 2023; 29:81-88. [PMID: 36573546 PMCID: PMC9796213 DOI: 10.3201/eid2901.212118] [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: 12/29/2022] Open
Abstract
To investigate the association between enteric pathogens, fecal microbes, and child growth, we conducted a prospective cohort study of 236 children <5 years of age in rural eastern Democratic Republic of the Congo. We analyzed baseline fecal specimens by quantitative PCR and measured child height and weight at baseline and growth at a 6-month follow-up. At baseline, 66% (156/236) of children had >3 pathogens in their feces. We observed larger increases in height-for-age-z-scores from baseline to the 6-month follow-up among children with Akkermansia muciniphila in their feces (coefficient 0.02 [95% CI 0.0001-0.04]; p = 0.04). Children with Cryptosporidium in their feces had larger declines in weight-for-height/length z-scores from baseline to the 6-month follow-up (coefficient -0.03 [95% CI -0.05 to -0.005]; p = 0.02). Our study showed high prevalence of enteric pathogens among this pediatric cohort and suggests A. muciniphila can potentially serve as a probiotic to improve child growth.
Collapse
|
39
|
Luoma J, Adubra L, Ashorn P, Ashorn U, Bendabenda J, Dewey KG, Hallamaa L, Coghlan R, Horton WA, Hyöty H, Kortekangas E, Lehto KM, Maleta K, Matchado A, Nkhoma M, Oikarinen S, Parkkila S, Purmonen S, Fan YM. Association between asymptomatic infections and linear growth in 18-24-month-old Malawian children. MATERNAL & CHILD NUTRITION 2023; 19:e13417. [PMID: 36111423 DOI: 10.1111/mcn.13417] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 12/15/2022]
Abstract
Inadequate diet and frequent symptomatic infections are considered major causes of growth stunting in low-income countries, but interventions targeting these risk factors have achieved limited success. Asymptomatic infections can restrict growth, but little is known about their role in global stunting prevalence. We investigated factors related to length-for-age Z-score (LAZ) at 24 months by constructing an interconnected network of various infections, biomarkers of inflammation (as assessed by alpha-1-acid glycoprotein [AGP]), and growth (insulin-like growth factor 1 [IGF-1] and collagen X biomarker [CXM]) at 18 months, as well as other children, maternal, and household level factors. Among 604 children, there was a continuous decline in mean LAZ and increased mean length deficit from birth to 24 months. At 18 months of age, the percentage of asymptomatic children who carried each pathogen was: 84.5% enterovirus, 15.5% parechovirus, 7.7% norovirus, 4.6% rhinovirus, 0.6% rotavirus, 69.6% Campylobacter, 53.8% Giardia lamblia, 11.9% malaria parasites, 10.2% Shigella, and 2.7% Cryptosporidium. The mean plasma IGF-1 concentration was 12.5 ng/ml and 68% of the children had systemic inflammation (plasma AGP concentration >1 g/L). Shigella infection was associated with lower LAZ at 24 months through both direct and indirect pathways, whereas enterovirus, norovirus, Campylobacter, Cryptosporidium, and malaria infections were associated with lower LAZ at 24 months indirectly, predominantly through increased systemic inflammation and reduced plasma IGF-1 and CXM concentration at 18 months.
Collapse
Affiliation(s)
- Juho Luoma
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Laura Adubra
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Per Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Paediatrics, Tampere University Hospital, Tampere, Finland
| | - Ulla Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jaden Bendabenda
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Kathryn G Dewey
- Department of Nutrition, Institute for Global Nutrition, University of California, Davis, California, USA
| | - Lotta Hallamaa
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ryan Coghlan
- Research Center, Shriners Hospitals for Children, Portland, Oregon, USA.,Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - William A Horton
- Research Center, Shriners Hospitals for Children, Portland, Oregon, USA.,Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon, USA
| | - Heikki Hyöty
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Ltd., Tampere University Hospital, Tampere, Finland
| | - Emma Kortekangas
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi-Maarit Lehto
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kenneth Maleta
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Andrew Matchado
- School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Minyanga Nkhoma
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sami Oikarinen
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Seppo Parkkila
- Fimlab Ltd., Tampere University Hospital, Tampere, Finland.,Clinical Medicine, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Sami Purmonen
- Clinical Medicine, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Yue-Mei Fan
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
40
|
Crosstalk between Resveratrol and Gut Barrier: A Review. Int J Mol Sci 2022; 23:ijms232315279. [PMID: 36499603 PMCID: PMC9739931 DOI: 10.3390/ijms232315279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/08/2022] Open
Abstract
The plant-based nutraceuticals are receiving increasing interest in recent time. The high attraction to the phytochemicals is associated with their anti-inflammatory and antioxidant activities, which can lead to reduced risk of the development of cardiovascular and other non-communicable diseases. One of the most disseminated groups of plant bioactives are phenolic compounds. It was recently hypothesized that phenolic compounds can have the ability to improve the functioning of the gut barrier. The available studies showed that one of the polyphenols, resveratrol, has great potential to improve the integrity of the gut barrier. Very promising results have been obtained with in vitro and animal models. Still, more clinical trials must be performed to evaluate the effect of resveratrol on the gut barrier, especially in individuals with increased intestinal permeability. Moreover, the interplay between phenolic compounds, intestinal microbiota and gut barrier should be carefully evaluated in the future. Therefore, this review offers an overview of the current knowledge about the interaction between polyphenols with a special emphasis on resveratrol and the gut barrier, summarizes the available methods to evaluate the intestinal permeability, discusses the current research gaps and proposes the directions for future studies in this research area.
Collapse
|
41
|
Gizaw Z, Yalew AW, Bitew BD, Lee J, Bisesi M. Stunting among children aged 24-59 months and associations with sanitation, enteric infections, and environmental enteric dysfunction in rural northwest Ethiopia. Sci Rep 2022; 12:19293. [PMID: 36369357 PMCID: PMC9652362 DOI: 10.1038/s41598-022-23981-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
Abstract
Stunting is a public health issue of global concern. Despite, poor sanitation, diarrhea, parasitic infections, and environmental enteric dysfunction (EED) are associated with stunting, their link is poorly understood and has not been investigated in Ethiopia. This study was conducted to assess the associations of stunting with sanitation, enteric infections, and EED among children aged 24-59 months in rural northwest Ethiopia. A community-based cross-sectional study was conducted among 224 randomly selected children aged 24-59 months in rural areas of the east Dembiya district. We collected information on household food insecurity and dietary diversity using pre-tested questionnaires adopted from the food and nutrition technical assistance (FANTA) project. We used height-for-age-z score (HAZ) to define stunting. We also used the data collected to measure the environmental exposures of children to intestinal parasitic infections and fecal biomarkers of EED. A multivariable binary logistic regression model was used to assess the association of stunting with sanitation, enteric infections, and EED. Of the 224 children, 33% (95% CI 27, 39%) were stunted. Stunting in children was significantly associated with poor dietary intake (AOR 3.0, 95% CI 1.2, 7.3), open defecation practice (AOR 3.0, 95% CI 1.2, 7.9), presence of animal excreta in the living environment (AOR 3.4, 95% CI 1.2, 9.9), E. coli contamination of drinking water (AOR 4.2, 95% CI 1.1, 15.3), diarrheal disease incidence (AOR 3.4, 95% CI 1.5, 7.7), intestinal parasites in children (AOR 3.3, 95% CI 1.3, 8.8), and higher EED disease activity scores (AOR 2.9, 95% CI 1.2, 6.7). One-third of the children in the study area were stunted and this high prevalence of stunting was associated with poor dietary intake, poor hygiene and sanitation conditions, enteric infections, and EED. Thus, stunting can be prevented by improving sanitation and hygienic conditions to prevent repeated enteric infections in children and by promoting dietary diversity of children.
Collapse
Affiliation(s)
- Zemichael Gizaw
- grid.59547.3a0000 0000 8539 4635Department of Environmental and Occupational Health and Safety, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia ,grid.458355.a0000 0004 9341 7904Addis Continental Institute of Public Health, Addis Ababa, Ethiopia ,grid.261331.40000 0001 2285 7943Global One Health Initiative (GOHi), The Ohio State University, Columbus, OH USA
| | - Alemayehu Worku Yalew
- grid.7123.70000 0001 1250 5688School of Public Health, Addis Ababa University, Addis Ababa, Ethiopia
| | - Bikes Destaw Bitew
- grid.59547.3a0000 0000 8539 4635Department of Environmental and Occupational Health and Safety, Institute of Public Health, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Jiyoung Lee
- grid.261331.40000 0001 2285 7943Division of Environmental Health Sciences, College of Public Health, The Ohio State University, 1841 Neil Avenue, Columbus, OH 43210 USA ,grid.261331.40000 0001 2285 7943Department of Food Science and Technology, The Ohio State University, Columbus, OH USA
| | - Michael Bisesi
- grid.261331.40000 0001 2285 7943Division of Environmental Health Sciences, College of Public Health, The Ohio State University, 1841 Neil Avenue, Columbus, OH 43210 USA
| |
Collapse
|
42
|
George CM, Birindwa A, Li S, Williams C, Kuhl J, Thomas E, François R, Presence AS, Claude BRJ, Mirindi P, Bisimwa L, Perin J, Stine OC. Akkermansia muciniphila Associated with Improved Linear Growth among Young Children, Democratic Republic of the Congo. Emerg Infect Dis 2022. [DOI: 10.3201/eid2811.212118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
43
|
Rogawski McQuade ET, Scharf RJ, Svensen E, Huggins A, Maphula A, Bayo E, Blacy L, Pamplona E. de Souza P, Costa H, Houpt ER, Bessong PO, Mduma ER, Lima AAM, Guerrant RL. Impact of Shigella infections and inflammation early in life on child growth and school-aged cognitive outcomes: Findings from three birth cohorts over eight years. PLoS Negl Trop Dis 2022; 16:e0010722. [PMID: 36149931 PMCID: PMC9534434 DOI: 10.1371/journal.pntd.0010722] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/05/2022] [Accepted: 08/06/2022] [Indexed: 11/30/2022] Open
Abstract
Background Shigella infections cause inflammation, which has been hypothesized to mediate the associations between Shigella and child development outcomes among children in low-resource settings. We aimed to assess whether early life inflammation and Shigella infections affect school-aged growth and cognitive outcomes from 6–8 years of age. Methodology/principal findings We conducted follow-up assessments of anthropometry, reasoning skills, and verbal fluency in 451 children at 6–8 years of age in the Brazil, Tanzania, and South Africa sites of MAL-ED, a longitudinal birth cohort study. We estimated the associations between Shigella burden and inflammation with linear growth at 2, 5, and 6–8 years of age, and with the cognitive test scores using linear regression and adjusting for potential confounding variables. We also assessed whether inflammation mediated the associations between Shigella and school-aged outcomes using a regression-based approach to mediation analysis. A high prevalence of Shigella was associated with a 0.32 (95% CI: 0.08, 0.56) z-score lower height-for-age z-score (HAZ) at 6–8 years compared to a low prevalence of Shigella. Intestinal inflammation had a smaller association with HAZ at 6–8 years. Shigella burden had small and consistently negative associations with cognitive outcomes in Brazil and Tanzania, but not South Africa, and the estimates were not statistically significant. Systemic inflammation was strongly associated with lower verbal fluency scores in Brazil (semantic fluency z-score difference: -0.57, 95% CI: -1.05, -0.10; phonemic fluency z-score difference: -0.48, 95% CI: -0.93, -0.03). There was no evidence that intestinal inflammation mediated the association between Shigella and HAZ or cognitive outcomes. Conclusions/significance While Shigella infections were consistently associated with long-term deficits in linear growth, the estimates of the negative associations between Shigella and cognitive outcomes were imprecise and only observed in the Brazil and Tanzania sites. Systemic inflammation was strongly associated with lower semantic and phonemic fluency scores in Brazil only, highlighting the site-specificity of effects. Shigella infections are common among children in low-resource settings and cause inflammation, which may contribute to poor child development outcomes. We studied children from birth to 6–8 years of age in Brazil, Tanzania, and South Africa to assess whether Shigella infections and markers of inflammation in the first 2 years of life were associated with child development outcomes at school age, including height and cognitive assessment scores. Shigella infections were consistently associated with long-term deficits in linear growth. However, the associations between Shigella and cognitive outcomes were smaller, not statistically significant, and inconsistent across the three sites. There was no evidence that inflammation caused by Shigella explained the associations between Shigella and school-aged outcomes. Systemic inflammation was strongly associated with lower cognitive assessment scores in Brazil but not in the other sites, highlighting the differences in determinants of child development across settings. Quantifying the impact and understanding mechanisms for the effect of early life exposures to Shigella on long-term cognitive outcomes remains challenging.
Collapse
Affiliation(s)
| | - Rebecca J. Scharf
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | | | - Amber Huggins
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | | | - Eliwaza Bayo
- Haydom Global Health Research Centre, Haydom Lutheran Hospital, Haydom, Tanzania
| | - Ladislaus Blacy
- Haydom Global Health Research Centre, Haydom Lutheran Hospital, Haydom, Tanzania
| | | | - Hilda Costa
- Universidade Federal do Ceara, Fortaleza, Brazil
| | - Eric R. Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| | | | - Estomih R. Mduma
- Haydom Global Health Research Centre, Haydom Lutheran Hospital, Haydom, Tanzania
| | | | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
44
|
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
|
45
|
Palit P, Ahmed MMM, Gazi MA, Haque MA, Alam MA, Haque R, Mahfuz M, Ahmed T. Association of Secretor Status with Enteropathy and Growth among Children in Bangladesh Aged 1-24 Months. Am J Trop Med Hyg 2022; 107:449-456. [PMID: 35895378 PMCID: PMC9393443 DOI: 10.4269/ajtmh.22-0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/30/2022] [Indexed: 11/24/2022] Open
Abstract
Secretor status refers to the ability of an individual to secrete blood group antigens into body fluids and onto the different epithelial surfaces. Concurrent findings have demonstrated an association of the secretor status of children with susceptibility to a plethora of enteropathogens. We aimed to determine a possible association of secretor status of children with childhood enteropathy, an important causal factor for childhood growth failure. Participants of the Malnutrition and Enteric Disease (MAL-ED) birth cohort study from the Bangladesh site were enrolled along with their mothers. Saliva was analyzed for determining blood groups and secretor status of the children and their mothers by using an in-house ELISA. Approximately 59% of children and 65% of mothers were found to be secretor positive. Secretor-positive children were found to have a significantly positive association with alpha-1-antitrypsin (β-coefficient: 0.11, 95% CI: 0.07, 0.21, P < 0.01) and with environmental enteric dysfunction score (β-coefficient: 0.32, 95% CI: 0.29, 0.65, P = 0.05). However, despite a negative effect size, secretor-positive children did not show any statistical significance with length-for-age and weight-for-age z scores (LAZ and WAZ), respectively. Our findings indicate toward the genetic factor of secretor status of children being associated with childhood growth faltering, through increased susceptibility to distinct enteropathogens and the consequent development of enteric inflammation and enteropathy among children. However, these findings are only applicable in Bangladeshi settings and thus need to be validated in several other similar settings, to establish a possible relationship between the secretor status of children with enteropathy and resulting childhood growth failure.
Collapse
Affiliation(s)
- Parag Palit
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Diseases Research, Dhaka, Bangladesh (icddr,b)
| | - Mondar Maruf Moin Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Diseases Research, Dhaka, Bangladesh (icddr,b)
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Diseases Research, Dhaka, Bangladesh (icddr,b)
| | - Md Ahshanul Haque
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Diseases Research, Dhaka, Bangladesh (icddr,b)
| | - Md Ashraful Alam
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Diseases Research, Dhaka, Bangladesh (icddr,b)
| | - Rashidul Haque
- Emerging Infections and Parasitology Laboratory, International Centre for Diarrhoeal Diseases Research, Dhaka, Bangladesh (icddr,b)
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Diseases Research, Dhaka, Bangladesh (icddr,b)
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Diseases Research, Dhaka, Bangladesh (icddr,b)
| |
Collapse
|
46
|
Hasan MM, Gazi MA, Das S, Fahim SM, Hossaini F, Khan AR, Ferdous J, Alam MA, Mahfuz M, Ahmed T. Gut biomolecules (I-FABP, TFF3 and lipocalin-2) are associated with linear growth and biomarkers of environmental enteric dysfunction (EED) in Bangladeshi children. Sci Rep 2022; 12:13905. [PMID: 35974137 PMCID: PMC9381788 DOI: 10.1038/s41598-022-18141-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
In the current world, a major challenge to diagnose environmental enteric dysfunction (EED) is the lack of validated non-invasive biomarkers. Intestine derived molecules, including intestinal fatty acid binding protein (I-FABP), trefoil factor-3 (TFF3), lactoferrin, lipocalin-2 (LCN2), and mucin-2, have been reported as indicators of intestinal inflammation and gut health. Therefore, we aimed to investigate the levels of these bio-molecules as biomarkers of EED among under-2 children in Bangladesh. A total of 140 children were recruited in a case-control design. All the biomarkers were measured by ELISA. Spearman's rank correlation was performed to see the correlation between the biomarkers and the EED score. Moreover, multivariable linear regression was performed to investigate the association of biomarkers with length-for-age z-score (LAZ). TFF3 correlates positively with myeloperoxidase (r = 0.26, p < 0.05) and EED score (r = 0.17, p < 0.05). Likewise, LCN2 correlates positively with myeloperoxidase (r = 0.37, p < 0.05), neopterin (r = 0.33, p < 0.05) and EED score (r = 0.31, p < 0.05). Moreover, multivariable linear regression revealed a negative association of I-FABP with LAZ of the study participants. Our results imply that TFF3 and LCN2 might be promising biomarkers to diagnose intestinal inflammation and EED, while I-FABP is negatively associated with linear growth of Bangladeshi children.
Collapse
Affiliation(s)
- Md Mehedi Hasan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Md Amran Gazi
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh.
| | - Subhasish Das
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Shah Mohammad Fahim
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Farzana Hossaini
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Ar-Rafi Khan
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Jafrin Ferdous
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Md Ashraful Alam
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
- James P. Grant School of Public Health, BRAC University, Dhaka, Bangladesh
- Department of Global Health, University of Washington, Seattle, WA, USA
| |
Collapse
|
47
|
Mogaka JN, Owuor PM, Odhiambo S, Waterman C, McGuire MK, Fuchs GJ, Attia SL. Investigating the Impact of Moringa oleifera Supplemented to Kenyan Breastfeeding Mothers on Maternal and Infant Health: A Cluster Randomized Single-Blinded Controlled Pilot Trial Protocol. JPGN REPORTS 2022; 3:e237. [PMID: 37168619 PMCID: PMC10158460 DOI: 10.1097/pg9.0000000000000237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/22/2022] [Indexed: 05/13/2023]
Abstract
Undernutrition contributes to up to 45% of deaths globally in children <5 years, with an optimal time for intervention before 24 months of age. Breastmilk microbiome helps establish the infant intestinal microbiome and impacts infant intestinal and nutritional health. Inadequacies in breastmilk composition such as low vitamin A contribute to infant nutrient deficiencies. Changes in milk fatty acid composition (reduced saturated and increased unsaturated fatty acids) may reduce susceptibility to enteric infection and increase protective intestinal bacteria. Moringa oleifera leaves (moringa) provide high nutrient concentrations (including protein, iron, vitamin A) and increase milk production; this may enhance breastmilk quantity and quality and improve infant health. Objective To investigate the role of moringa supplementation to improve maternal and infant nutritional and intestinal health via changes in maternal milk quantity and quality. Methods Fifty mother-infant pairs exclusively breastfeeding will be enrolled in a single-blinded randomized controlled trial in Kombewa County Hospital and Chulaimbo SubCounty Hospital, Kisumu, Kenya. Intervention Dietary Supplementation of 20 g of Moringa oleifera leaf powder divided twice daily in corn porridge consumed daily for 3 months while control comparator will receive porridge daily for 3 months. Outcomes Change in infant growth and maternal milk output (primary); maternal and infant vitamin A and iron status, changes in infant and maternal intestinal health (secondary). Participating Centers Pamoja Community Based Organization, Kombewa Sub-County Hospital, and Chulaimbo Sub-County Hospital.
Collapse
Affiliation(s)
| | - Patrick Mbullo Owuor
- Pamoja Community Based Organization, Kisumu, Kenya
- Department of Anthropology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, Illinois
| | | | - Carrie Waterman
- Institute of Global Nutrition, University of California, Davis, Davis, California
| | - Michelle K. McGuire
- College of Agricultural and Life Sciences, Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, Idaho
| | - George J. Fuchs
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Epidemiology and Department of Preventive Medicine and Environmental Health, University of Kentucky College of Public Health, Lexington, Kentucky
| | - Suzanna L. Attia
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
48
|
Chaima D, Hart J, Pickering H, Burr S, Maleta K, Kalua K, Bailey R, Holland M. Association between intestinal bacterial carriage, biomarkers of environmental enteric dysfunction, and stunting in rural Malawian children. Gates Open Res 2022. [DOI: 10.12688/gatesopenres.13671.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background. Available data from murine studies suggest that intestinal bacteria may have a role in modulating growth phenotypes in the host. We investigated the prevalence of four gut bacteria known in murine models to impair growth (Bifidobacterium longum, Faecalibacterium prausnitzii, Dorea formicigenerans, and Akkermansia muciniphila), the level of fecal biomarkers of environmental enteric dysfunction (EED) and stunting in rural Malawian children. Methods. DNA and protein were extracted from fecal samples of rural Malawian children (aged 1-59 months) at a baseline cross-sectional survey in the Mangochi district of Malawi conducted within the framework of the Macrolides Oraux pour Réduire les Décès avec un Oeil sur la Résistance (MORDOR) trial. Intestinal carriage of bacteria was measured by PCR. Neopterin (NEO), myeloperoxidase (MPO), and alpha-1 antitrypsin (AAT), biomarkers of EED, were measured by an enzyme-linked immunosorbent assay (ELISA) test. Height-for-age Z (HAZ) score <-2 defined stunting. Tests of proportions and regression models were used to explore the relationship between bacterial carriage, EED, and stunting. Results. Fecal samples from 613 children were available for laboratory analyses. F. prausnitzii and D. formicigenerans were prevalent in over 70% of children while B. longum was the least prevalent. B. longum carriage in younger children was associated with elevated EED biomarkers. Two thirds of children had elevated NEO, 33% had elevated MPO, and 16% had elevated AAT. Stunting was found in 38%. No significant associations were found between EED biomarkers or intestinal bacteria carriage and stunting. Conclusion. Intestinal carriage of these four bacteria was not associated with stunting in Malawian children. Carriage was also not associated with EED, nor EED biomarker levels associated with stunting. Further factors acting in concert are necessary to impact EED, perturb growth, and alter gut bacterial carriage.
Collapse
|
49
|
Liu Z, Ashorn U, Chingwanda C, Maleta K, Hallamaa L, Matchado A, Kortekangas E, Dewey KG, Ashorn P, Fan Y. Provision of small‐quantity lipid‐based nutrient supplements does not improve intestinal health among rural Malawian children. MATERNAL & CHILD NUTRITION 2022; 18:e13331. [PMID: 35128820 PMCID: PMC9218311 DOI: 10.1111/mcn.13331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Zhifei Liu
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Ulla Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | | | - Kenneth Maleta
- Department of Public Health University of Malawi Zomba Malawi
| | - Lotta Hallamaa
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Andrew Matchado
- Department of Public Health University of Malawi Zomba Malawi
| | - Emma Kortekangas
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Kathryn G Dewey
- Department of Nutrition University of California Davis CA USA
| | - Per Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
- Department of Paediatrics Tampere University Hospital Tampere Finland
| | - Yue‐Mei Fan
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology Tampere University Tampere Finland
| |
Collapse
|
50
|
Modern G, Mpolya E, Sauli E. Causal relationship between Environmental Enteric Dysfunction (EED), poor WaSH practices and growth failure in children from Rukwa-Tanzania. SCIENTIFIC AFRICAN 2022. [DOI: 10.1016/j.sciaf.2022.e01281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|